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


  • Sebastian Kadener Laboratory of Molecular Neurobiology, Hebrew University, Jerusalem
    Herman, N., Kadener, S. and Shifman, S. (2022). The chromatin factor ROW cooperates with BEAF-32 in regulating long-range inducible genes. EMBO Rep 23(12): e54720. PubMed ID: 36245419

    Pamudurti, N. R., Patop, I. L., Krishnamoorthy, A., Bartok, O., Maya, R., Lerner, N., Ashwall-Fluss, R., Konakondla, J. V. V., Beatus, T. and Kadener, S. (2022). circMbl functions in cis and in trans to regulate gene expression and physiology in a tissue-specific fashion. Cell Rep 39(4): 110740. PubMed ID: 35476987

    Martin Anduaga, A., Evantal, N., Patop, I. L., Bartok, O., Weiss, R. and Kadener, S. (2019). Thermosensitive alternative splicing senses and mediates temperature adaptation in Drosophila. Elife 8. PubMed ID: 31702556

    Buchumenski, I., Bartok, O., Ashwal-Fluss, R., Pandey, V., Porath, H. T., Levanon, E. Y. and Kadener, S. (2017). Dynamic hyper-editing underlies temperature adaptation in Drosophila. PLoS Genet 13(7): e1006931. PubMed ID: 28746393

    Afik, S., Bartok, O., Artyomov, M. N., Shishkin, A. A., Kadri, S., Hanan, M., Zhu, X., Garber, M. and Kadener, S. (2017). Defining the 5 and 3 landscape of the Drosophila transcriptome with Exo-seq and RNaseH-seq. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 28335028

    Lerner, I., Bartok, O., Wolfson, V., Menet, J. S., Weissbein, U., Afik, S., Haimovich, D., Gafni, C., Friedman, N., Rosbash, M. and Kadener, S. (2015). Clk post-transcriptional control denoises circadian transcription both temporally and spatially. Nat Commun 6: 7056. PubMed ID: 25952406

    Ashwal-Fluss, R., Meyer, M., Pamudurti, N. R., Ivanov, A., Bartok, O., Hanan, M., Evantal, N., Memczak, S., Rajewsky, N. and Kadener, S. (2014). circRNA biogenesis competes with pre-mRNA splicing. Mol Cell 56: 55-66. PubMed ID: 25242144

    Weiss, R., Bartok, O., Mezan, S., Malka, Y. and Kadener, S. (2014). Synergistic Interactions between the Molecular and Neuronal Circadian Networks Drive Robust Behavioral Circadian Rhythms in Drosophila melanogaster. PLoS Genet 10: e1004252. PubMed ID: 24698952

    Mezan, S., Ashwal-Fluss, R., Shenhav, R., Garber, M. and Kadener, S. (2013). Genome-wide assessment of post-transcriptional control in the fly brain. Front Mol Neurosci 6: 49. PubMed ID: 24367289

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

  • James Kadonaga Biological Sciences, U. C. San Diego
    Vo Ngoc, L., Kassavetis, G. A. and Kadonaga, J. T. (2019). The RNA Polymerase II core promoter in Drosophila. Genetics 212(1): 13-24. PubMed ID: 31053615

    Shir-Shapira, H., Sloutskin, A., Adato, O., Ovadia-Shochat, A., Ideses, D., Zehavi, Y., Kassavetis, G., Kadonaga, J. T., Unger, R. and Juven-Gershon, T. (2019). Identification of evolutionarily conserved downstream core promoter elements required for the transcriptional regulation of Fushi tarazu target genes. PLoS One 14(4): e0215695. PubMed ID: 30998799

    Khuong, M. T., Fei, J., Cruz-Becerra, G. and Kadonaga, J. T. (2017). A simple and versatile system for the ATP-dependent assembly of chromatin. J Biol Chem 292(47): 19478-19490. PubMed ID: 28982979

    Wang, Y. L., Duttke, S. H., Chen, K., Johnston, J., Kassavetis, G. A., Zeitlinger, J. and Kadonaga, J. T. (2014). TRF2, but not TBP, mediates the transcription of ribosomal protein genes. Genes Dev. PubMed ID: 24958592

    Kassavetis, G. A. and Kadonaga, J. T. (2014). The Annealing Helicase and Branch Migration Activities of Drosophila HARP. PLoS One 9: e98173. PubMed ID: 24866343

    Torigoe, S. E., Patel, A., Khuong, M. T., Bowman, G. D. and Kadonaga, J. T. (2013). ATP-dependent chromatin assembly is functionally distinct from chromatin remodeling. Elife 2: e00863. PubMed ID: 23986862

    Kadonaga, J. T. (2012). Perspectives on the RNA polymerase II core promoter. Wiley Interdiscip Rev Dev Biol 1: 40-51. PubMed ID: 23801666

  • Ilona Kadow International Max Planck, Research School for Molecular and Cellular Life Sciences, Martinsried/Munich
    Hussain, A., Pooryasin, A., Zhang, M., Loschek, L. F., La Fortezza, M., Friedrich, A. B., Blais, C. M., Ucpunar, H. K., Yepez, V. A., Lehmann, M., Gompel, N., Gagneur, J., Sigrist, S. J. and Grunwald Kadow, I. C. (2018). Inhibition of oxidative stress in cholinergic projection neurons fully rescues aging-associated olfactory circuit degeneration in Drosophila. Elife 7. PubMed ID: 29345616

    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

    Hartl, M., et al. (2011). A new Prospero and microRNA-279 pathway restricts CO2 receptor neuron formation. J. Neurosci. 31(44): 15660-73. PubMed ID: 22049409

  • Laurie Kaguni Biochemistry and Molecular Biology, Michigan State University, East Lansing
    Salminen, T. S., Cannino, G., Oliveira, M. T., Lillsunde, P., Jacobs, H. T. and Kaguni, L. S. (2019). Lethal interaction of nuclear and mitochondrial genotypes in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 31076384

    Salminen, T.S., Oliveira, M.T., Cannino, G., Lillsunde, P., Jacobs, H.T. and Kaguni, L.S. (2017). Mitochondrial genotype modulates mtDNA copy number and organismal phenotype in Drosophila. Mitochondrion [Epub ahead of print]. PubMed ID: 28214560

    Stiban, J., Farnum, G. A., Hovde, S. L. and Kaguni, L. S. (2014). The N-terminal domain of the Drosophila mitochondrial replicative DNA helicase contains an iron-sulfur cluster and binds DNA. J Biol Chem. PubMed ID: 25023283

    Fernandez-Moreno, M. A., Hernandez, R., Adan, C., Roberti, M., Bruni, F., Polosa, P. L., Cantatore, P., Matsushima, Y., Kaguni, L. S. and Garesse, R. (2013). Drosophila nuclear factor DREF regulates the expression of the mitochondrial DNA helicase and mitochondrial transcription factor B2 but not the mitochondrial translation factor B1. Biochim Biophys Acta 1829: 1136-1146. PubMed ID: 23916463

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

  • Toshie Kai Temasek Life Sciences Laboratory, National University of Singapore
    Iki, T., Takami, M. and Kai, T. (2020). Modulation of Ago2 Loading by Cyclophilin 40 Endows a Unique Repertoire of Functional miRNAs during Sperm Maturation in Drosophila. Cell Rep 33(6): 108380. PubMed ID: 33176138

    Kawaguchi, S., Ueki, M. and Kai, T. (2020). Drosophila MARF1 ensures proper oocyte maturation by regulating nanos expression. PLoS One 15(4): e0231114. PubMed ID: 32243476

    Teo, R. Y. W., Anand, A., Sridhar, V., Okamura, K. and Kai, T. (2018). Heterochromatin protein 1a functions for piRNA biogenesis predominantly from pericentric and telomeric regions in Drosophila. Nat Commun 9(1): 1735. Pubmed ID: 29728561

    Quénerch'du, E., Anand, A. and Kai, T. (2016). The piRNA pathway is developmentally regulated during spermatogenesis in Drosophila. RNA [Epub ahead of print]. PubMed ID: 27208314

    Ghodsi, Z., Silva, E. S. and Hassani, H. (2015). Bicoid Signal Extraction with a Selection of Parametric and Nonparametric Signal Processing Techniques. Genomics Proteomics Bioinformatics. PubMed ID: 26197438

    Patil, V. S., Anand, A., Chakrabarti, A. and Kai, T. (2014). The Tudor domain protein Tapas, a homolog of the vertebrate Tdrd7, functions in piRNA pathway to regulate retrotransposons in germline of Drosophila melanogaster. BMC Biol 12: 61. PubMed ID: 25287931

    Pek, J. W., Ng, B. F. and Kai, T. (2012). Polo-mediated phosphorylation of Maelstrom regulates oocyte determination during oogenesis in Drosophila. Development 139: 4505-4513. PubMed ID: 23136393

    Lim, R. S., Osato, M. and Kai, T. (2012). Isolation of undifferentiated female germline cells from adult Drosophila ovaries. Curr Protoc Stem Cell Biol Chapter 2: Unit2E 3. PubMed ID: 22872426

  • Daniel Kalderon Biological Sciences,Columbia University
    Reilein, A., Kogan, H. V., Misner, R., Park, K. S. and Kalderon, D. (2021). Adult stem cells and niche cells segregate gradually from common precursors that build the adult Drosophila ovary during pupal development. Elife 10. PubMed ID: 34590579
    Little, J. C., Garcia-Garcia, E., Sul, A. and Kalderon, D. (2020). Drosophila hedgehog can act as a morphogen in the absence of regulated Ci processing. Elife 9. PubMed ID: 33084577

    Melamed, D. and Kalderon, D. (2020). Opposing JAK-STAT and Wnt signaling gradients define a stem cell domain by regulating differentiation at two borders. Elife 9. PubMed ID: 33135631

    Reilein, A., Melamed, D., Tavare, S. and Kalderon, D. (2018). Division-independent differentiation mandates proliferative competition among stem cells. Proc Natl Acad Sci U S A. PubMed ID: 29555768

    Huang, J., Reilein, A. and Kalderon, D. (2017). Yorkie and Hedgehog independently restrict BMP production in Escort cells to permit germline differentiation in the Drosophila ovary. Development. PubMed ID: 28619819

    Garcia Garcia, E., Little, J. C. and Kalderon, D. (2017). The exon junction complex and Srp54 contribute to Drosophila Hedgehog signaling via ci RNA Splicing. Genetics [Epub ahead of print]. PubMed ID: 28637711

    Reilein, A., Melamed, D., Park, K. S., Berg, A., Cimetta, E., Tandon, N., Vunjak-Novakovic, G., Finkelstein, S. and Kalderon, D. (2017). Alternative direct stem cell derivatives defined by stem cell location and graded Wnt signalling. Nat Cell Biol 19(5):433-444. PubMed ID: 28414313

    Huang, J. and Kalderon, D. (2014). Coupling of Hedgehog and Hippo pathways promotes stem cell maintenance by stimulating proliferation. J Cell Biol. PubMed ID: 24798736

    Vied, C., Reilein, A., Field, N. S. and Kalderon, D. (2012). Regulation of stem cells by intersecting gradients of long-range niche signals. Dev Cell 23: 836-848. PubMed ID: 23079600

    Wang, Z. A., Huang, J. and Kalderon, D. (2012). Drosophila follicle stem cells are regulated by proliferation and niche adhesion as well as mitochondria and ROS. Nat Commun 3: 769. PubMed ID: 22473013

  • Alla Ivanovna Kalmykova Laboratory of Eukaryotic Genomic Repeats, Kurchatov Institute, Moscow
    Morgunova, V. V., Sokolova, O. A., Sizova, T. V., Malaev, L. G., Babaev, D. S., Kwon, D. A. and Kalmykova, A. I. (2022). Dysfunction of Lamin B and Physiological Aging Cause Telomere Instability in Drosophila Germline. Biochemistry (Mosc) 87(12): 1600-1610. PubMed ID: 36717449

    Komarov, P. A., Sokolova, O., Akulenko, N., Brasset, E., Jensen, S. and Kalmykova, A. (2020). Epigenetic Requirements for Triggering Heterochromatinization and Piwi-Interacting RNA Production from Transgenes in the Drosophila Germline. Cells 9(4). PubMed ID: 32290057

    Kordyukova, M., Sokolova, O., Morgunova, V., Ryazansky, S., Akulenko, N., Glukhov, S. and Kalmykova, A. (2019). Nuclear Ccr4-Not mediates the degradation of telomeric and transposon transcripts at chromatin in the Drosophila germline. Nucleic Acids Res. PubMed ID: 31724732

    Radion, E., Sokolova, O., Ryazansky, S., Komarov, P. A., Abramov, Y. and Kalmykova, A. (2019). The integrity of piRNA clusters is abolished by insulators in the Drosophila germline. Genes (Basel) 10(3). PubMed ID: 30862119

    Kordyukova, M., Morgunova, V., Olovnikov, I., Komarov, P. A., Mironova, A., Olenkina, O. M. and Kalmykova, A. (2018). Subcellular localization and Egl-mediated transport of telomeric retrotransposon HeT-A ribonucleoprotein particles in the Drosophila germline and early embryogenesis. PLoS One 13(8): e0201787. PubMed ID: 30157274

    Akulenko, N., Ryazansky, S., Morgunova, V., Komarov, P. A., Olovnikov, I., Vaury, C., Jensen, S. and Kalmykova, A. (2018). Transcriptional and chromatin changes accompanying de novo formation of transgenic piRNA clusters. Rna. PubMed ID: 29358235

    Ryazansky, S., Radion, E., Mironova, A., Akulenko, N., Abramov, Y., Morgunova, V., Kordyukova, M.Y., Olovnikov, I. and Kalmykova, A. (2017). Natural variation of piRNA expression affects immunity to transposable elements. PLoS Genet 13: e1006731. PubMed ID: 28448516

    Radion, E., Ryazansky, S., Akulenko, N., Rozovsky, Y., Kwon, D., Morgunova, V., Olovnikov, I. and Kalmykova, A. (2016). Telomeric retrotransposon HeT-A contains a bidirectional promoter that initiates divergent transcription of piRNA precursors in Drosophila germline. J Mol Biol [Epub ahead of print]. PubMed ID: 27939293

    Olovnikov, I. A., Morgunova, V. V., Mironova, A. A., Kordyukova, M. Y., Radion, E. I., Olenkina, O. M., Akulenko, N. V. and Kalmykova, A. I. (2016). Interaction of Telomeric Retroelement HeT-A Transcripts and Their Protein Product Gag in Early Embryogenesis of Drosophila. Biochemistry (Mosc) 81(9): 1023-1030. PubMed ID: 27682174

  • Azusa Kamikouchi Laboratory of Cellular Neurobiology, Tokyo University of Pharmacy & Life Science
    Ishimoto, H. and Kamikouchi, A. (2020). A feedforward circuit regulates action selection of pre-mating courtship behavior in female Drosophila. Curr Biol 30(3): 396-407.e394. PubMed ID: 31902724

    Ishikawa, Y., Fujiwara, M., Wong, J., Ura, A. and Kamikouchi, A. (2019). Stereotyped combination of hearing and wind/gravity-sensing neurons in the Johnston's organ of Drosophila. Front Physiol 10: 1552. PubMed ID: 31969834

    Yamada, D., Ishimoto, H., Li, X., Kohashi, T., Ishikawa, Y. and Kamikouchi, A. (2018). GABAergic local interneurons shape female fruit fly response to mating songs. J Neurosci 38(18): 4329-4347. PubMed ID: 29691331

    Li, X., Ishimoto, H. and Kamikouchi, A. (2018). Auditory experience controls the maturation of song discrimination and sexual response in Drosophila. Elife 7. PubMed ID: 29555017

    :Matsuo, E., Seki, H., Asai, T., Morimoto, T., Miyakawa, H., Ito, K. and Kamikouchi, A. (2016). The organization of projection neurons and local neurons of the primary auditory center in the fruit fly Drosophila melanogaster. J Comp Neurol 24(6):1099-164. PubMed ID: 26762251

    Yoon, J., Matsuo, E., Yamada, D., Mizuno, H., Morimoto, T., Miyakawa, H., Kinoshita, S., Ishimoto, H. and Kamikouchi, A. (2013). Selectivity and Plasticity in a Sound-Evoked Male-Male Interaction in. PLoS One 8: e74289. PubMed ID: 24086330

    Kamikouchi, A., Wiek, R., Effertz, T., Gopfert, M. C. and Fiala, A. (2010). Transcuticular optical imaging of stimulus-evoked neural activities in the Drosophila peripheral nervous system. Nat Protoc 5: 1229-1235. PubMed ID: 20595952

    Kamikouchi, A., Albert, J. T. and Gopfert, M. C. (2010). Mechanical feedback amplification in Drosophila hearing is independent of synaptic transmission. Eur J Neurosci 31: 697-703. PubMed ID: 20384813

    Morgunova, V., Akulenko, N., Radion, E., Olovnikov, I., Abramov, Y., Olenina, L. V., Shpiz, S., Kopytova, D. V., Georgieva, S. G. and Kalmykova, A. (2016). Telomeric repeat silencing in germ cells is essential for early development in Drosophila. Nucleic Acids Res 44(15): 7509. PubMed ID: 27220463

    Shpiz, S., Ryazansky, S., Olovnikov, I., Abramov, Y. and Kalmykova, A. (2014). Euchromatic transposon insertions trigger production of novel Pi- and endo-siRNAs at the target sites in the drosophila germline. PLoS Genet 10(2): e1004138. PubMed ID: 24516406

  • KyeongJin Kang Laboratory of Behavioral Neurogenetics, Sungkyunkwan University, Seoul, Korea
    Lee, M. J., Sung, H. Y., Jo, H., Kim, H. W., Choi, M. S., Kwon, J. Y. and Kang, K. (2017). Ionotropic Receptor 76b is required for gustatory aversion to excessive Na+ in Drosophila. Mol Cells 40(10): 787-795. PubMed ID: 29081083

    Du, E. J., Ahn, T. J., Wen, X., Seo, D. W., Na, D. L., Kwon, J. Y., Choi, M., Kim, H. W., Cho, H. and Kang, K. (2016). Nucleophile sensitivity of Drosophila TRPA1 underlies light-induced feeding deterrence. Elife 5. PubMed ID: 27656903

    Choi, J., van Giesen, L., Choi, M. S., Kang, K., Sprecher, S. G. and Kwon, J. Y. (2016). A Pair of Pharyngeal Gustatory Receptor Neurons Regulates Caffeine-Dependent Ingestion in Drosophila Larvae. Front Cell Neurosci 10: 181. PubMed ID: 27486388

    Park, J. H., Chen, J., Jang, S., Ahn, T. J., Kang, K., Choi, M. S. and Kwon, J. Y. (2016). A subset of enteroendocrine cells is activated by amino acids in the Drosophila midgut. FEBS Lett 590: 493-500. PubMed ID: 26801353

    Kim, H., Choi, M. S., Kang, K. and Kwon, J. Y. (2016). Behavioral Analysis of Bitter Taste Perception in Drosophila Larvae. Chem Senses 41: 85-94. PubMed ID: 26512069

  • Madhuri Kango-Singh Department of Biology, University of Dayton
    Gangwani, K., Snigdha, K. and Kango-Singh, M. (2020). Tep1 Regulates Yki Activity in Neural Stem Cells in Drosophila Glioma Model. Front Cell Dev Biol 8: 306. PubMed ID: 32457905

    Waghmare, I. and Kango-Singh, M. (2016). Loss of cell adhesion increases tumorigenic potential of polarity deficient Scribble mutant cells. PLoS One 11: e0158081. PubMed ID: 27327956

    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. (2015). Drosophila C-terminal Src kinase regulates growth via the Hippo signaling pathway. Dev Biol 397: 67-76. PubMed ID: 25446534

  • Pankaj Kapahi Buck Institute for Research on Aging, Novato, CA
    Hodge, B. A., Meyerhof, G. T., Katewa, S. D., Lian, T., Lau, C., Bar, S., Leung, N. Y., Li, M., Li-Kroeger, D., Melov, S., Schilling, B., Montell, C. and Kapahi, P. (2022). Dietary restriction and the transcription factor clock delay eye aging to extend lifespan in Drosophila Melanogaster. Nat Commun 13(1): 3156. PubMed ID: 35672419

    Hilsabeck, T. A. U., Liu-Bryan, R., Guo, T., Wilson, K. A., Bose, N., Raftery, D., Beck, J. N., Lang, S., Jin, K., Nelson, C. S., Oron, T., Stoller, M., Promislow, D., Brem, R. B., Terkeltaub, R. and Kapahi, P. (2022). A fly GWAS for purine metabolites identifies human FAM214 homolog medusa, which acts in a conserved manner to enhance hyperuricemia-driven pathologies by modulating purine metabolism and the inflammatory response. Geroscience. PubMed ID: 35381951

    Sharma, A., Akagi, K., Pattavina, B., Wilson, K. A., Nelson, C., Watson, M., Maksoud, E., Harata, A., Ortega, M., Brem, R. B. and Kapahi, P. (2020). Musashi expression in intestinal stem cells attenuates radiation-induced decline in intestinal permeability and survival in Drosophila. Sci Rep 10(1): 19080. PubMed ID: 33154387

    Wilson, K. A., Beck, J. N., Nelson, C. S., Hilsabeck, T. A., Promislow, D., Brem, R. B. and Kapahi, P. (2020). GWAS for Lifespan and Decline in Climbing Ability in Flies upon Dietary Restriction Reveal decima as a Mediator of Insulin-like Peptide Production. Curr Biol. PubMed ID: 32502405

    Lang, S., Hilsabeck, T. A., Wilson, K. A., Sharma, A., Bose, N., Brackman, D. J., Beck, J. N., Chen, L., Watson, M. A., Killilea, D. W., Ho, S., Kahn, A., Giacomini, K., Stoller, M. L., Chi, T. and Kapahi, P. (2019). A conserved role of the insulin-like signaling pathway in diet-dependent uric acid pathologies in Drosophila melanogaster. PLoS Genet 15(8): e1008318. PubMed ID: 31415568

    Huang, K., Chen, W., Zhu, F., Li, P. W., Kapahi, P. and Bai, H. (2019). RiboTag translatomic profiling of Drosophila oenocytes under aging and induced oxidative stress. BMC Genomics 20(1): 50. PubMed ID: 30651069

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

    Chi, T., Kim, M. S., Lang, S., Bose, N., Kahn, A., Flechner, L., Blaschko, S. D., Zee, T., Muteliefu, G., Bond, N., Kolipinski, M., Fakra, S. C., Mandel, N., Miller, J., Ramanathan, A., Killilea, D. W., Bruckner, K., Kapahi, P. and Stoller, M. L. (2015). A Drosophila model identifies a critical role for zinc in mineralization for kidney stone disease. PLoS One 10: e0124150. PubMed ID: 25970330

    Miller, J., Chi, T., Kapahi, P., Kahn, A. J., Kim, M. S., Hirata, T., Romero, M. F., Dow, J. A. and Stoller, M. L. (2013). Drosophila Melanogaster as an Emerging Translational Model of Human Nephrolithiasis. J Urol. PubMed ID: 23500641

    Bruce, K. D., Hoxha, S., Carvalho, G. B., Yamada, R., Wang, H. D., Karayan, P., He, S., Brummel, T., Kapahi, P. and Ja, W. W. (2013). High carbohydrate-low protein consumption maximizes Drosophila lifespan. Exp Gerontol. PubMed ID: 23403040

    Katewa, S. D., Demontis, F., Kolipinski, M., Hubbard, A., Gill, M. S., Perrimon, N., Melov, S. and Kapahi, P. (2012). Intramyocellular fatty-acid metabolism plays a critical role in mediating responses to dietary restriction in Drosophila melanogaster. Cell Metab 16: 97-103. PubMed ID: 22768842

  • Martin Kapun University of Zurich
    Kawecki, T. J., Erkosar, B., Dupuis, C., Hollis, B., Stillwell, R. C. and Kapun, M. (2021). The genomic architecture of adaptation to larval malnutrition points to a trade-off with adult starvation resistance in Drosophila. Mol Biol Evol. PubMed ID: 33677563

    Kapopoulou, A., Kapun, M., Pieper, B., Pavlidis, P., Wilches, R., Duchen, P., Stephan, W. and Laurent, S. (2020). Demographic analyses of a new sample of haploid genomes from a Swedish population of Drosophila melanogaster. Sci Rep 10(1): 22415. PubMed ID: 33376238

    Kapun, M. and Flatt, T. (2019). The adaptive significance of chromosomal inversion polymorphisms in Drosophila melanogaster. Mol Ecol 28(6): 1263-1282. PubMed ID: 30230076

  • Francois Karch Department of Genetics and Evolution, University of Geneva
    Castro Alvarez, J. J., Revel, M., Carrasco, J., Cleard, F., Pauli, D., Hilgers, V., Karch, F. and Maeda, R. K. (2021). Repression of the Hox gene abd-A by ELAV-mediated Transcriptional Interference. PLoS Genet 17(11): e1009843. PubMed ID: 34780465

    Immarigeon, C., Frei, Y., Delbare, S. Y. N., Gligorov, D., Machado Almeida, P., Grey, J., Fabbro, L., Nagoshi, E., Billeter, J. C., Wolfner, M. F., Karch, F. and Maeda, R. K. (2021). Identification of a micropeptide and multiple secondary cell genes that modulate Drosophila male reproductive success. Proc Natl Acad Sci U S A 118(15). PubMed ID: 33876742

    Prince, E., Kroeger, B., Gligorov, D., Wilson, C., Eaton, S., Karch, F., Brankatschk, M. and Maeda, R. K. (2018). Rab-mediated trafficking in the secondary cells of Drosophila male accessory glands and its role in fecundity. Traffic. PubMed ID: 30426623

    Maeda, R. K., Sitnik, J. L., Frei, Y., Prince, E., Gligorov, D., Wolfner, M. F. and Karch, F. (2018). The lncRNA male-specific abdominal plays a critical role in Drosophila accessory gland development and male fertility. PLoS Genet 14(7): e1007519. PubMed ID: 30011265

    Cleard, F., Wolle, D., Taverner, A. M., Aoki, T., Deshpande, G., Andolfatto, P., Karch, F. and Schedl, P. (2016). Different Evolutionary Strategies to Conserve Chromatin Boundary Function in the Bithorax Complex. Genetics. PubMed ID: 28007886

    Sitnik, J.L., Gligorov, D., Maeda, R.K., Karch, F. and Wolfner, M.F. (2016). Secondary cell expressed genes in the male accessory gland are needed for the Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 26746709

    Wolle, D., Cleard, F., Aoki, T., Deshpande, G., Schedl, P. and Karch, F. (2015). Functional requirements for Fab-7 boundary activity in the Bithorax Complex. Mol Cell Biol. PubMed ID: 26303531

    Maeda, R. K. and Karch, F. (2015). The open for business model of the bithorax complex in Drosophila. Chromosoma [Epub ahead of print]. PubMed ID: 26067031

    Gummalla, M., Galetti, S., Maeda, R. K. and Karch, F. (2014). Hox gene regulation in the central nervous system of. Front Cell Neurosci 8: 96. PubMed ID: 24795565

    Bocksberger, M., Karch, F. and Gibert, J. M. (2014). In vivo analysis of a fluorescent SUMO fusion in transgenic. Fly (Austin) 8. PubMed ID: 24583630

    Gligorov, D., Sitnik, J. L., Maeda, R. K., Wolfner, M. F., Karch, F. (2013) A novel function for the hox gene abd-B in the male accessory gland regulates the long-term female post-mating response in Drosophila. PLoS Genet 9: e1003395. PubMed ID: 23555301

  • Roger Karess Institut Jacques Monod, Paris
    Menant, A. and Karess, R. E. (2020). Mutations in the Drosophila rough deal gene affecting RZZ kinetochore function. Biol Cell. PubMed ID: 32602944

    Raich, N., Mahmoudi, S., Emre, D. and Karess, R. E. (2018). Mad1 influences interphase nucleoplasm organization and chromatin regulation in Drosophila. Open Biol 8(10). PubMed ID: 30333236

    Caous, R., Pascal, A., Rome, P., Richard-Parpaillon, L., Karess, R. and Giet, R. (2015). Spindle assembly checkpoint inactivation fails to suppress neuroblast tumour formation in aurA mutant Drosophila. Nat Commun 6: 8879. PubMed ID: 26568519

    Defachelles, L., Raich, N., Terracol, R., Baudin, X., Williams, B., Goldberg, M. and Karess, R. E. (2015). RZZ and Mad1 dynamics in Drosophila mitosis. Chromosome Res [Epub ahead of print]. PubMed ID: 25772408

    Defachelles, L., Hainline, S. G., Menant, A., Lee, L. A. and Karess, R. E. (2015). A maternal effect rough deal mutation suggesting multiple pathways regulating Drosophila RZZ kinetochore recruitment. J Cell Sci [Epub ahead of print]. PubMed ID: 25616898

    Menant, A. and Karess, R. E. (2012). Inducing "cytokinesis" without mitosis in unfertilized Drosophila eggs. Cell Cycle 11: 2856-2863. PubMed ID: 22801541

    Althoff, F., Karess, R. E. and Lehner, C. F. (2012). Spindle checkpoint-independent inhibition of mitotic chromosome segregation by Drosophila Mps1. Mol Biol Cell 23: 2275-2291. PubMed ID: 22553353

  • Jason Karpac Department of Molecular and Cellular Medicine. Texas A&M University School of Medicine, College Station, Texas
    Mlih, M. and Karpac, J. (2022). Integrin-ECM interactions and membrane-associated Catalase cooperate to promote resilience of the Drosophila intestinal epithelium. PLoS Biol 20(5): e3001635. PubMed ID: 35522719

    Zhao, X. and Karpac, J. (2021). Glutamate metabolism directs energetic trade-offs to shape host-pathogen susceptibility in Drosophila. Cell Metab. PubMed ID: 34710355

    Zhao, X., Li, X., Shi, X. and Karpac, J. (2020). Diet-MEF2 interactions shape lipid droplet diversification in muscle to influence Drosophila lifespan. Aging Cell 19(7). PubMed ID: 32537848

    Vandehoef, C., Molaei, M. and Karpac, J. (2020). Dietary Adaptation of Microbiota in Drosophila Requires NF-κB-Dependent Control of the Translational Regulator 4E-BP. Cell Rep 31(10): 107736. PubMed ID: 32521261

    Molaei, M., Vandehoef, C. and Karpac, J. (2019). NF-kappaB shapes metabolic adaptation by attenuating Foxo-mediated lipolysis in Drosophila. Dev Cell. PubMed ID: 31080057

    Zhao, X. and Karpac, J. (2017). Muscle directs diurnal energy homeostasis through a myokine-dependent hormone module in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 28669758

    Karpac, J., Biteau, B. and Jasper, H. (2013). Misregulation of an adaptive metabolic response contributes to the age-related disruption of lipid homeostasis in Drosophila. Cell Rep 4(6): 1250-1261. PubMed ID: 24035390

    Kapuria, S., Karpac, J., Biteau, B., Hwangbo, D. and Jasper, H. (2012). Notch-mediated suppression of TSC2 expression regulates cell differentiation in the Drosophila intestinal stem cell lineage. PLoS Genet 8(11): e1003045. PubMed ID: 23144631

  • Gary Karpen Lawrence Berkeley Labs
    Kasinathan, B., Colmenares, S. U., 3rd, McConnell, H., Young, J. M., Karpen, G. H. and Malik, H. S. (2020). Innovation of heterochromatin functions drives rapid evolution of essential ZAD-ZNF genes in Drosophila. Elife 9. PubMed ID: 33169670

    Lee, Y. C. G., Ogiyama, Y., Martins, N. M. C., Beliveau, B. J., Acevedo, D., Wu, C. T., Cavalli, G. and Karpen, G. H. (2020). Pericentromeric heterochromatin is hierarchically organized and spatially contacts H3K9me2 islands in euchromatin. PLoS Genet 16(3): e1008673. PubMed ID: 32203508

    Janssen, A., Colmenares, S. U., Lee, T. and Karpen, G. H. (2019). Timely double-strand break repair and pathway choice in pericentromeric heterochromatin depend on the histone demethylase dKDM4A. Genes Dev 33(1-2): 103-115. PubMed ID: 30578303

    Colmenares, S. U., Swenson, J. M., Langley, S. A., Kennedy, C., Costes, S. V. and Karpen, G. H. (2017). Drosophila Histone Demethylase KDM4A Has Enzymatic and Non-enzymatic Roles in Controlling Heterochromatin Integrity. Dev Cell 42(2): 156-169.e155. PubMed ID: 28743002

    Lee, Y. C. G. and Karpen, G. H. (2017). Pervasive epigenetic effects of Drosophila euchromatic transposable elements impact their evolution. Elife 6. PubMed ID: 28695823

    Colmenares, S. U., Swenson, J. M., Langley, S. A., Kennedy, C., Costes, S. V. and Karpen, G. H. (2017). Drosophila histone demethylase KDM4A has enzymatic and non-enzymatic roles in controlling heterochromatin integrity. Dev Cell 42(2): 156-169.e155. PubMed ID: 28743002

    Strom, A. R., Emelyanov, A. V., Mir, M., Fyodorov, D. V., Darzacq, X. and Karpen, G. H. (2017). Phase separation drives heterochromatin domain formation. Nature [Epub ahead of print]. PubMed ID: 28636597

    Swenson, J.M., Colmenares, S.U., Strom, A.R., Costes, S.V. and Karpen, G.H. (2016). The composition and organization of Drosophila heterochromatin are heterogeneous and dynamic. Elife 5. PubMed ID: 27514026

    Janssen, A., Breuer, G. A., Brinkman, E. K., van der Meulen, A. I., Borden, S. V., van Steensel, B., Bindra, R. S., LaRocque, J. R. and Karpen, G. H. (2016). A single double-strand break system reveals repair dynamics and mechanisms in heterochromatin and euchromatin. Genes Dev 30: 1645-1657. PubMed ID: 27474442

    Ryu, T., Spatola, B., Delabaere, L., Bowlin, K., Hopp, H., Kunitake, R., Karpen, G. H. and Chiolo, I. (2015). Heterochromatic breaks move to the nuclear periphery to continue recombinational repair. Nat Cell Biol 17: 1401-1411. PubMed ID: 26502056

  • Timothy L Karr The Biodesign Institute, Arizona State University, Tempe, Arizona,
    Garlovsky, M. D., Sandler, J. A. and Karr, T. L. (2022). Functional Diversity and Evolution of the Drosophila Sperm Proteome. Mol Cell Proteomics 21(10): 100281. PubMed ID: 35985624

    Ibaraki, K., Nakatsuka, M., Ohsako, T., Watanabe, M., Miyazaki, Y., Shirakami, M., Karr, T. L., Sanuki, R., Tomaru, M. and Takano-Shimizu-Kouno, T. (2021). A cross-species approach for the identification of Drosophila male sterility genes. G3 (Bethesda) 11(8). PubMed ID: 34849808

    Karr, T. L., Southern, H., Rosenow, M. A., Gossmann, T. I. and Snook, R. R. (2019). The Old and the New: Discovery Proteomics Identifies Putative Novel Seminal Fluid Proteins in Drosophila. Mol Cell Proteomics 18(Suppl 1): S23-S33. PubMed ID: 30760537

  • Judy Kassis NICHD, NIH, Bethesda
    De, S., Gehred, N. D., Fujioka, M., Chan, F. W., Jaynes, J. B. and Kassis, J. A. (2020). Defining the Boundaries of Polycomb Domains in Drosophila. Genetics. PubMed ID: 32948625

    De, S., Cheng, Y., Sun, M. A., Gehred, N. D. and Kassis, J. A. (2019). Structure and function of an ectopic Polycomb chromatin domain. Sci Adv 5(1): eaau9739. PubMed ID: 30662949

    De, S., Mitra, A., Cheng, Y., Pfeifer, K. and Kassis, J. A. (2016). Formation of a Polycomb-Domain in the Absence of Strong Polycomb Response Elements. PLoS Genet 12: e1006200. PubMed ID: 27466807

    Ray, P., De, S., Mitra, A., Bezstarosti, K., Demmers, J. A., Pfeifer, K. and Kassis, J. A. (2016). Combgap contributes to recruitment of Polycomb group proteins in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 27001825

    Cheng, Y., Brunner, A. L., Kremer, S., DeVido, S. K., Stefaniuk, C. M. and Kassis, J. A. (2014). Co-regulation of invected and engrailed by a complex array of regulatory sequences in Drosophila. Dev Biol. PubMed ID: 25172431

    Dorsett, D. and Kassis, J. A. (2014). Checks and Balances between Cohesin and Polycomb in Gene Silencing and Transcription. Curr Biol 24: R535-R539. PubMed ID: 24892918

    Brown, J. L. and Kassis, J. A. (2013). Architectural and Functional Diversity of Polycomb Group Response Elements in Drosophila. Genetics. PubMed ID: 23934890

    Kassis, J. A., Brown, J. L. (2013) Polycomb Group Response Elements in Drosophila and Vertebrates. Adv Genet 81C: 83-118. PubMed ID: 23419717

    Langlais, K. K., Brown, J. L. and Kassis, J. A. (2012). Polycomb Group Proteins Bind an engrailed PRE in Both the "ON" and "OFF" Transcriptional States of engrailed. PLoS One 7: e48765. PubMed ID: 23139817

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

    Noyes, A., Stefaniuk, C., Cheng, Y., Kennison, J. A. and Kassis, J. A. (2011). Modulation of the activity of a polycomb-group response element in Drosophila by a mutation in the transcriptional activator woc. G3 (Bethesda) 1: 471-478. PubMed ID: 22384357

  • Vladimir Katanaev University of Lausanne
    Larasati, Y. A., Savitsky, M., Koval, A., Solis, G. P., Valnohova, J. and Katanaev, V. L. (2022). Restoration of the GTPase activity and cellular interactions of Gα(o) mutants by Zn(2+) in GNAO1 encephalopathy models. Sci Adv 8(40): eabn9350. PubMed ID: 36206333

    Savitsky, M., Solis, G. P., Kryuchkov, M. and Katanaev, V. L. (2020). Humanization of Drosophila Gαo to Model GNAO1 Paediatric Encephalopathies. Biomedicines 8(10). PubMed ID: 33036271
    Solis, G. P., Bilousov, O., Koval, A., Luchtenborg, A. M., Lin, C. and Katanaev, V. L. (2017). Golgi-resident Galphao promotes protrusive membrane dynamics. Cell 170(5):939-955. PubMed ID: 28803726

    Moon, S., Kim, W., Kim, S., Kim, Y., Song, Y., Bilousov, O., Kim, J., Lee, T., Cha, B., Kim, M., Kim, H., Katanaev, V. L. and Jho, E. H. (2016). Phosphorylation by NLK inhibits YAP-14-3-3-interactions and induces its nuclear localization. EMBO Rep [Epub ahead of print]. PubMed ID: 27979972

    Pouly, D., Chenaux, S., Martin, V., Babis, M., Koch, R., Nagoshi, E., Katanaev, V. L., Gachon, F. and Staub, O. (2016). USP2-45 is a circadian clock output effector regulating calcium absorption at the post-translational level. PLoS One 11: e0145155. PubMed ID: 26756164

    Luchtenborg, A. M., Purvanov, V., Melnik, B. S., Becker, S. and Katanaev, V. L. (2015). Mode of interaction of the Galphao subunit of heterotrimeric G proteins with the GoLoco1 motif of Drosophila Pins is determined by guanine nucleotides. Biosci Rep. PubMed ID: 26487707

    Blagodatski, A., Sergeev, A., Kryuchkov, M., Lopatina, Y. and Katanaev, V.L. (2015). Diverse set of Turing nanopatterns coat corneae across insect lineages. Proc Natl Acad Sci U S A 112: 10750-10755. PubMed ID: 26307762

    Luchtenborg, A. M. and Katanaev, V. L. (2014). Lack of evidence of the interaction of the Ass peptide with the Wnt signaling cascade in Drosophila models of Alzheimer's disease. Mol Brain 7: 81. PubMed ID: 25387847

    Luchtenborg, A. M., Solis, G. P., Egger-Adam, D., Koval, A., Lin, C., Blanchard, M. G., Kellenberger, S. and Katanaev, V. L. (2014). Heterotrimeric Go protein links Wnt-Frizzled signaling with ankyrins to regulate the neuronal microtubule cytoskeleton. Development 141: 3399-3409. PubMed ID: 25139856

    Bilousov, O., Koval, A., Keshelava, A. and Katanaev, V. L. (2014). Identification of novel elements of the Drosophila blisterome sheds light on potential pathological mechanisms of several human diseases. PLoS One 9: e101133. PubMed ID: 24968325

    Lin, C., Koval, A., Tishchenko, S., Gabdulkhakov, A., Tin, U., Solis, G. P. and Katanaev, V. L. (2014). Double Suppression of the Galpha Protein Activity by RGS Proteins. Mol Cell 53: 663-671. PubMed ID: 24560274

  • Matthew Kaiser Department of Psychiatry, University of Pennsylvania, Philadelphia
    Gong, N. N., Dilley, L. C., Williams, C. E., Moscato, E. H., Szuperak, M., Wang, Q., Jensen, M., Girirajan, S., Tan, T. Y., Deardorff, M. A., Li, D., Song, Y. and Kayser, M. S. (2021). The chromatin remodeler ISWI acts during Drosophila development to regulate adult sleep. Sci Adv 7(8). PubMed ID: 33597246

    Chakravarti Dilley, L., Szuperak, M., Gong, N. N., Williams, C. E., Saldana, R. L., Garbe, D. S., Syed, M. H., Jain, R. and Kayser, M. S. (2020). Identification of a molecular basis for the juvenile sleep state. Elife 9. PubMed ID: 32202500

    Churgin, M. A., Szuperak, M., Davis, K. C., Raizen, D. M., Fang-Yen, C. and Kayser, M. S. (2019). Quantitative imaging of sleep behavior in Caenorhabditis elegans and larval Drosophila melanogaster. Nat Protoc. PubMed ID: 30953041

    Belfer, S. J., Bashaw, A. G., Perlis, M. L. and Kayser, M. S. (2019). A Drosophila model of sleep restriction therapy for insomnia. Mol Psychiatry. PubMed ID: 30824866

    Dilley, L. C., Vigderman, A., Williams, C. E. and Kayser, M. S. (2018). Behavioral and Genetic Features of Sleep Ontogeny in Drosophila. Sleep. PubMed ID: 29746663

    Szuperak, M., Churgin, M. A., Borja, A. J., Raizen, D. M., Fang-Yen, C. and Kayser, M. S. (2018). A sleep state in Drosophila larvae required for neural stem cell proliferation. Elife 7. PubMed ID: 29424688

    Chakravarti, L., Moscato, E. H. and Kayser, M. S. (2017). Unraveling the Neurobiology of Sleep and Sleep Disorders Using Drosophila. Curr Top Dev Biol 121: 253-285. PubMed ID: 28057302

    Garbe, D. S., Vigderman, A. S., Moscato, E., Dove, A. E., Vecsey, C. G., Kayser, M. S. and Sehgal, A. (2016). Changes in Female Drosophila Sleep following Mating Are Mediated by SPSN-SAG Neurons. J Biol Rhythms 31(6): 551-567. PubMed ID: 27658900

  • Hokto Kazama RIKEN Brain Science Institute, Wako, Japan
    Endo, K., Tsuchimoto, Y. and Kazama, H. (2020). Synthesis of Conserved Odor Object Representations in a Random, Divergent-Convergent Network. Neuron. PubMed ID: 32814018

    Shiozaki, H. M., Ohta, K. and Kazama, H. (2020). A Multi-regional Network Encoding Heading and Steering Maneuvers in Drosophila. Neuron. PubMed ID: 32023429

    Mercier, D., Tsuchimoto, Y., Ohta, K. and Kazama, H. (2018). Olfactory landmark-based communication in interacting Drosophila. Curr Biol 28(16): 2624-2631.e2625. PubMed ID: 30078566

    Shiozaki, H. M. and Kazama, H. (2017). Parallel encoding of recent visual experience and self-motion during navigation in Drosophila. Nat Neurosci. PubMed ID: 28869583

    Inada, K., Tsuchimoto, Y. and Kazama, H. (2017). Origins of Cell-Type-Specific Olfactory Processing in the Drosophila Mushroom Body Circuit. Neuron 95(2): 357-367 e354. PubMed ID: 28728024

    Badel, L., Ohta, K., Tsuchimoto, Y. and Kazama, H. (2016). Decoding of Context-Dependent Olfactory Behavior in Drosophila. Neuron 91(1): 155-167. PubMed ID: 27321924

    Kazama, H. (2015). Systems neuroscience in Drosophila: Conceptual and technical advantages. Neuroscience 296: 3-14. PubMed ID: 24973655

  • Thomas Kaufman Department of Biology, Indiana University
    Eisman, R. C., Phelps, M. A. and Kaufman, T. C. (2016). The End of a Monolith: Deconstructing the Cnn-Polo interaction. Fly (Austin): [Epub ahead of print]. PubMed ID: 27096551

    Eisman, R. C., Phelps, M. A. and Kaufman, T. (2015). An amino-terminal Polo kinase interaction motif acts in the regulation of centrosome formation and reveals a novel function for centrosomin (cnn) in Drosophila. Genetics 201: 685-706. PubMed ID: 26447129

    Eisman, R. C. and Kaufman, T. C. (2013). Probing the Boundaries of Orthology: The Unanticipated Rapid Evolution of Drosophila Centrosomin. Genetics. PubMed ID: 23749319

    Cook, R. K., Christensen, S. J., Deal, J. A., Coburn, R. A., Deal, M. E., Gresens, J. M., Kaufman, T. C. and Cook, K. R. (2012). The generation of chromosomal deletions to provide extensive coverage and subdivision of the Drosophila melanogaster genome. Genome Biol 13: R21. PubMed ID: 22445104

    Holtzman, S., Miller, D., Eisman, R., Kuwayama, H., Niimi, T. and Kaufman, T. (2010). Transgenic tools for members of the genus Drosophila with sequenced genomes. Fly (Austin) 4: 349-362. PubMed ID: 20890112

  • Nancy Kaufmann Department of Biological Sciences, University of Pittsburgh
    Hildebrand, J. D., Leventry, A. D., Aideyman, O. P., Majewski, J. C., Haddad, J. A., Bisi, D. C. and Kaufmann, N. (2021). A modifier screen identifies regulators of cytoskeletal architecture as mediators of Shroom-dependent changes in tissue morphology. Biol Open 10(2). PubMed ID: 33504488

    Yi, S. X., Benoit, J. B., Elnitsky, M. A., Kaufmann, N., Brodsky, J. L., Zeidel, M. L., Denlinger, D. L. and Lee, R. E., Jr. (2011). Function and immuno-localization of aquaporins in the Antarctic midge Belgica antarctica. J Insect Physiol 57(8): 1096-1105. PubMed ID: 21315725

    Spring, J. H., Robichaux, S. R., Kaufmann, N. and Brodsky, J. L. (2007). Localization of a Drosophila DRIP-like aquaporin in the Malpighian tubules of the house cricket, Acheta domesticus. Comp Biochem Physiol A Mol Integr Physiol 148(1): 92-100. PubMed ID: 17267256

  • Karla Kaun Neuroscience, Brown University, Providence, RI
    Scaplen, K. M., Talay, M., Fisher, J. D., Cohn, R., Sorkac, A., Aso, Y., Barnea, G. and Kaun, K. R. (2021). Transsynaptic mapping of Drosophila mushroom body output neurons. Elife 10. PubMed ID: 33570489

    Scaplen, K. M., Talay, M., Nunez, K. M., Salamon, S., Waterman, A. G., Gang, S., Song, S. L., Barnea, G. and Kaun, K. R. (2020). Circuits that encode and guide alcohol-associated preference. Elife 9. PubMed ID: 32497004

    Petruccelli, E., Brown, T., Waterman, A., Ledru, N. and Kaun, K. R. (2020). Alcohol Causes Lasting Differential Transcription in Drosophila Mushroom Body Neurons. Genetics 215(1): 103-116. PubMed ID: 32132098

    Scaplen, K. M., Mei, N. J., Bounds, H. A., Song, S. L., Azanchi, R. and Kaun, K. R. (2019). Automated real-time quantification of group locomotor activity in Drosophila melanogaster. Sci Rep 9(1): 4427. PubMed ID: 30872709

    Petruccelli, E., Feyder, M., Ledru, N., Jaques, Y., Anderson, E. and Kaun, K. R. (2018). Alcohol Activates Scabrous-Notch to Influence Associated Memories. Neuron 100(5): 1209-1223 e1204. PubMed ID: 30482693

  • Tadeusz Kawecki Department of Ecology and Evolution, University of Lausanne
    Erkosar, B., Yashiro, E., Zajitschek, F., Friberg, U., Maklakov, A. A., van der Meer, J. R. and Kawecki, T. J. (2018). Host diet mediates a negative relationship between abundance and diversity of Drosophila gut microbiota. Ecol Evol 8(18): 9491-9502. PubMed ID: 30377517

    Erkosar, B., Kolly, S., van der Meer, J. R. and Kawecki, T. J. (2017). Adaptation to Chronic Nutritional Stress Leads to Reduced Dependence on Microbiota in Drosophila melanogaster. MBio 8(5). PubMed ID: 29066546

    Hollis, B., Keller, L. and Kawecki, T. J. (2016). Sexual selection shapes development and maturation rates in Drosophila. Evolution [Epub ahead of print]. PubMed ID: 27883363

    Hollis, B., Houle, D. and Kawecki, T. J. (2015). Evolution of reduced postcopulatory molecular interactions in Drosophila populations lacking sperm competition. J Evol Biol [Epub ahead of print]. PubMed ID: 26395588

    Babin, A., Kolly, S. and Kawecki, T. J. (2014). Virulent bacterial infection improves aversive learning performance in Drosophila melanogaster. Brain Behav Immun. PubMed ID: 24863366

    Babin, A., Kolly, S., Schneider, F., Dolivo, V., Zini, M. and Kawecki, T. J. (2014). Fruit flies learn to avoid odours associated with virulent infection. Biol Lett 10: 20140048. PubMed ID: 24598110

    Hollis, B. and Kawecki, T. J. (2014). Male cognitive performance declines in the absence of sexual selection. Proc Biol Sci 281: 20132873. PubMed ID: 24573848

    Zrelec, V., Zini, M., Guarino, S., Mermoud, J., Oppliger, J., Valtat, A., Zeender, V. and Kawecki, T. J. (2013). Drosophila rely on learning while foraging under semi-natural conditions. Ecol Evol 3: 4139-4148. PubMed ID: 24324865

    Vijendravarma, R. K. and Kawecki, T. J. (2013). Epistasis and maternal effects in experimental adaptation to chronic nutritional stress in Drosophila. J Evol Biol. PubMed ID: 24118120

    Vijendravarma, R. K., Narasimha, S. and Kawecki, T. J. (2013). Predatory cannibalism in Drosophila melanogaster larvae. Nat Commun 4: 1789. PubMed ID: 23653201

  • Alex Keene Department of Biology, University of Nevada, Reno
    Murakami, K., Palermo, J., Stanhope, B. A., Gibbs, A. G. and Keene, A. C. (2021). A screen for sleep and starvation resistance identifies a wake-promoting role for the auxiliary channel unc79. G3 (Bethesda) 11(8). PubMed ID: 34849820

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

    Brown, E. B., Shah, K. D., Faville, R., Kottler, B. and Keene, A. C. (2020). Drosophila insulin-like peptide 2 mediates dietary regulation of sleep intensity. PLoS Genet 16(3): e1008270. PubMed ID: 32160200

    Sah Pamboro, E. L., Brown, E. B. and Keene, A. C. (2019). Dietary fatty acids promote sleep through a taste-independent mechanism. Genes Brain Behav: e12629. PubMed ID: 31845509

    Brown, E. B., Slocumb, M. E., Szuperak, M., Kerbs, A., Gibbs, A. G., Kayser, M. S. and Keene, A. C. (2019). Starvation resistance is associated with developmentally specified changes in sleep, feeding and metabolic rate. J Exp Biol. PubMed ID: 30606795

    Stahl, B. A., Peco, E., Davla, S., Murakami, K., Caicedo Moreno, N. A., van Meyel, D. J. and Keene, A. C. (2018). The taurine transporter Eaat2 functions in ensheathing glia to modulate sleep and metabolic Rate. Curr Biol. PubMed ID: 30416062

    Yurgel, M. E., Shah, K. D., Brown, E. B., Burns, C., Bennick, R. A., DiAngelo, J. R. and Keene, A. C. (2018). Ade2 functions in the Drosophila fat body to promote sleep. G3 (Bethesda). PubMed ID: 30249751

    Tauber, J. M., Brown, E. B., Li, Y., Yurgel, M. E., Masek, P. and Keene, A. C. (2017). A subset of sweet-sensing neurons identified by IR56d are necessary and sufficient for fatty acid taste. PLoS Genet 13(11): e1007059. PubMed ID: 29121639

    Stahl, B. A., Slocumb, M. E., Chaitin, H., Diangelo, J. R. and Keene, A. C. (2017). Sleep-dependent modulation of metabolic rate in Drosophila. Sleep [Epub ahead of print]. PubMed ID: 28541527

    Garbe, D.S., Bollinger, W.L., Vigderman, A., Masek, P., Gertowski, J., Sehgal, A. and Keene, A.C. (2015). Context-specific comparison of sleep acquisition systems in Drosophila. Biol Open [Epub ahead of print]. PubMed ID: 26519516

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

  • Krystyna Keleman Janelia Research Campus
    ei, Z., Henderson, K. and Keleman, K. (2022). A neural circuit linking learning and sleep in Drosophila long-term memory. Nat Commun 13(1): 609. PubMed ID: 35105888

    Dag, U., Lei, Z., Le, J. Q., Wong, A., Bushey, D. and Keleman, K. (2019). Neuronal reactivation during post-learning sleep consolidates long-term memory in Drosophila. Elife 8. PubMed ID: 30801246

    Zhao, X., Lenek, D., Dag, U., Dickson, B. and Keleman, K. (2018). Persistent activity in a recurrent circuit underlies courtship memory in Drosophila. Elife 7. PubMed ID: 29322941

    Koemans, T. S., Oppitz, C., Donders, R. A. T., van Bokhoven, H., Schenck, A., Keleman, K. and Kramer, J. M. (2017). Drosophila courtship conditioning as a measure of learning and memory. J Vis Exp(124) [Epub ahead of print]. PubMed ID: 28605393

    Kruttner, S., Traunmuller, L., Dag, U., Jandrasits, K., Stepien, B., Iyer, N., Fradkin, L. G., Noordermeer, J. N., Mensh, B. D. and Keleman, K. (2015). Synaptic Orb2A bridges memory acquisition and late memory consolidation in Drosophila. Cell Rep 11: 1953-1965. PubMed ID: 26095367

    Walkinshaw, E., Gai, Y., Farkas, C., Richter, D., Nicholas, E., Keleman, K. and Davis, R.L. (2015). Identification of genes that promote or inhibit olfactory memory formation in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 25644700

    Keleman, K., Vrontou, E., Kruttner, S., Yu, J. Y., Kurtovic-Kozaric, A. and Dickson, B. J. (2012). Dopamine neurons modulate pheromone responses in Drosophila courtship learning. Nature 489: 145-149. PubMed ID: 22902500

    Kruttner, S., Stepien, B., Noordermeer, J. N., Mommaas, M. A., Mechtler, K., Dickson, B. J. and Keleman, K. (2012). Drosophila CPEB Orb2A mediates memory independent of Its RNA-binding domain. Neuron 76: 383-395. PubMed ID: 23083740

  • Erin Kelleher University of Houston
    Lama, J., Srivastav, S., Tasnim, S., Hubbard, D., Hadjipanteli, S., Smith, B. R., Macdonald, S. J., Green, L. and Kelleher, E. S. (2022). Genetic variation in P-element dysgenic sterility is associated with double-strand break repair and alternative splicing of TE transcripts. PLoS Genet 18(12): e1010080. PubMed ID: 36477699

    Wang, L., Barbash, D. A. and Kelleher, E. S. (2020). Adaptive evolution among cytoplasmic piRNA proteins leads to decreased genomic auto-immunity. PLoS Genet 16(6): e1008861. PubMed ID: 32525870

    Zhang, S. and Kelleher, E. S. (2017). Targeted identification of TE insertions in a Drosophila genome through hemi-specific PCR. Mob DNA 8: 10. PubMed ID: 28775768

    Srivastav, S. P. and Kelleher, E. S. (2017). Paternal Induction of Hybrid Dysgenesis in Drosophila melanogaster Is Weakly Correlated with Both P-Element and hobo Element Dosage. G3 (Bethesda) 7(5): 1487-1497. PubMed ID: 28315830

    Kelleher, E. S. (2016). Reexamining the P-Element Invasion of Drosophila melanogaster Through the Lens of piRNA Silencing. Genetics 203(4): 1513-1531. PubMed ID: 27516614

  • Manolis Kellis MIT Computer Science and Artificial Intelligence Laboratory
    Jungreis, I., Chan, C. S., Waterhouse, R. M., Fields, G., Lin, M. F. and Kellis, M. (2016). Evolutionary dynamics of abundant stop codon readthrough. Mol Biol Evol [Epub ahead of print]. PubMed ID: 27604222

    Hammonds, A. S., Bristow, C. A., Fisher, W. W., Weiszmann, R., Wu, S., Hartenstein, V., Kellis, M., Yu, B., Frise, E. and Celniker, S. E. (2013). Spatial expression of transcription factors in Drosophila embryonic organ development. Genome Biol 14: R140. PubMed ID: 24359758

    Spivakov, M., Akhtar, J., Kheradpour, P., Beal, K., Girardot, C., Koscielny, G., Herrero, J., Kellis, M., Furlong, E. E. and Birney, E. (2012). Analysis of variation at transcription factor binding sites in Drosophila and humans. Genome Biol 13: R49. PubMed ID: 22950968

    Marbach, D., Roy, S., Ay, F., Meyer, P. E., Candeias, R., Kahveci, T., Bristow, C. A. and Kellis, M. (2012). Predictive regulatory models in Drosophila melanogaster by integrative inference of transcriptional networks. Genome Res 22: 1334-1349. PubMed ID: 22456606

    Jungreis, I., Lin, M. F., Spokony, R., Chan, C. S., Negre, N., Victorsen, A., White, K. P. and Kellis, M. (2011). Evidence of abundant stop codon readthrough in Drosophila and other metazoa. Genome Res 21: 2096-2113. PubMed ID: 21994247

  • Jason Kennington University of Western Australia
    Robinson, S. P., Simmons, L. W. and Kennington, W. J. (2014). Sex-biased mortality associated with inbreeding in Drosophila melanogaster. BMC Evol Biol 14: 51. PubMed ID: 24636623

    Robinson, S. P., Simmons, L. W. and Kennington, W. J. (2013). Estimating relatedness and inbreeding using molecular markers and pedigrees: the effect of demographic history. Mol Ecol. PubMed ID: 24102888

    Kennington, W. J. and Hoffmann, A. A. (2013). Patterns of genetic variation across inversions: geographic variation in the In(2L)t inversion in populations of Drosophila melanogaster from eastern Australia. BMC Evol Biol 13: 100. PubMed ID: 23688159

    Robinson, S. P., Kennington, W. J. and Simmons, L. W. (2012). Assortative mating for relatedness in a large naturally occurring population of Drosophila melanogaster. J Evol Biol 25: 716-725. PubMed ID: 22321157

  • Jim Kennison NICHD, NIH, Bethesda
    Monribot-Villanueva, J., Juarez-Uribe, R. A., Palomera-Sanchez, Z., Gutierrez-Aguiar, L., Zurita, M., Kennison, J. A., Vazquez, M. (2013) TnaA, an SP-RING Protein, Interacts with Osa, a Subunit of the Chromatin Remodeling Complex BRAHMA and with the SUMOylation Pathway in Drosophila melanogaster. PLoS One 8: e62251. PubMed ID: 23620817

    Lindsley, D. L., Roote, J. and Kennison, J. A. (2013). Anent the Genomics of Spermatogenesis in Drosophila melanogaster. PLoS One 8: e55915. PubMed ID: 23409089

    Stultz, B. G., Park, S. Y., Mortin, M. A., Kennison, J. A. and Hursh, D. A. (2012). Hox proteins coordinate peripodial decapentaplegic expression to direct adult head morphogenesis in Drosophila. Dev Biol 369: 362-376. PubMed ID: 22824425

    Noyes, A., Stefaniuk, C., Cheng, Y., Kennison, J. A. and Kassis, J. A. (2011). Modulation of the activity of a polycomb-group response element in Drosophila by a mutation in the transcriptional activator woc. G3 (Bethesda) 1: 471-478. PubMed ID: 22384357

  • Andrew Kern Department of Genetics, Rutgers, Piscataway, N. J.
    Adrion, J. R., Galloway, J. G. and Kern, A. D. (2020). Predicting the landscape of recombination using deep learning. Mol Biol Evol. PubMed ID: 32077950

    Reinhardt, J. A., Kolaczkowski, B., Jones, C. D., Begun, D. J. and Kern, A. D. (2014). Parallel Geographic Variation in Drosophila melanogaster. Genetics. PubMed ID: 24610860

    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

    Kolaczkowski, B., Kern, A. D., Holloway, A. K. and Begun, D. J. (2011). Genomic differentiation between temperate and tropical Australian populations of Drosophila melanogaster. Genetics 187: 245-260. PubMed ID: 21059887

  • Haig Keshishian Molecular Cellular & Developmental Biology, Yale University
    McNeill, E. M., Thompson, C., Berke, B., Chou, V. T., Rusch, J., Duckworth, A., DeProto, J., Taylor, A., Gates, J., Gertler, F., Keshishian, H. and Van Vactor, D. (2020). Drosophila enabled promotes synapse morphogenesis and regulates active zone form and function. Neural Dev 15(1): 4. PubMed ID: 32183907

    Berke, B., Le, L. and Keshishian, H. (2020). Target-dependent retrograde signaling mediates synaptic plasticity at the Drosophila neuromuscular junction. Dev Neurobiol. PubMed ID: 31950660

    Vonhoff, F. and Keshishian, H. (2017). In vivo calcium signaling during synaptic refinement at the Drosophila neuromuscular junction. J Neurosci [Epub ahead of print]. PubMed ID: 28476946

    Vonhoff, F. and Keshishian, H. (2016). Cyclic nucleotide signaling is required during synaptic refinement at the Drosophila neuromuscular junction. Dev Neurobiol [Epub ahead of print]. PubMed ID: 27281494

    Berke, B., Wittnam, J., McNeill, E., Van Vactor, D. L. and Keshishian, H. (2013). Retrograde BMP Signaling at the Synapse: A Permissive Signal for Synapse Maturation and Activity-Dependent Plasticity. J Neurosci 33: 17937-17950. PubMed ID: 24198381

    Olsen, D. P. and Keshishian, H. (2012). Monitoring membrane excitability in Drosophila expressing modified shaker constructs. Cold Spring Harb Protoc 2012: 226-230. PubMed ID: 22301649

    Leiserson, W. M. and Keshishian, H. (2011). Maintenance and regulation of extracellular volume and the ion environment in Drosophila larval nerves. Glia 59: 1312-1321. PubMed ID: 21305613

  • Albert Ketterman Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand (see article in ASBMB Today)
    Wongtrakul, J., Janphen, K., Saisawang, C. and Ketterman, A. J. (2014). Interaction of Omega, Sigma, and Theta glutathione transferases with p38b mitogenactivated protein kinase from the fruit fly, Drosophila melanogaster. J Insect Sci 14: 60. PubMed ID: 25373207

    Saisawang, C. and Ketterman, A. J. (2014). Micro-plasticity of genomes as illustrated by the evolution of glutathione transferases in 12 Drosophila species. PLoS One 9: e109518. PubMed ID: 25310450

    Wongtrakul, J., Sukittikul, S., Saisawang, C., Ketterman, A. J. (2012) Mitogen-Activated Protein Kinase p38b Interaction with Delta Class Glutathione Transferases from the Fruit Fly, Drosophila melanogaster. J Insect Sci 12: 107. PubMed ID: 23438069

    Saisawang, C., Wongsantichon, J. and Ketterman, A. J. (2012). A preliminary characterization of the cytosolic glutathione transferase proteome from Drosophila melanogaster. Biochem J 442: 181-190. PubMed ID: 22082028

  • Thomas Kidd Biology Department, University of Nevada, Reno
    Myers, L., Perera, H., Alvarado, M. G. and Kidd, T. (2017). The Drosophila Ret gene functions in the stomatogastric nervous system with the Maverick TGFbeta ligand and the Gfrl co-receptor. Development. PubMed ID: 29361562

    Alavi, M., Song, M., King, G. L., Gillis, T., Propst, R., Lamanuzzi, M., Bousum, A., Miller, A., Allen, R. and Kidd, T. (2016). Dscam1 forms a complex with Robo1 and the N-terminal fragment of Slit to promote the growth of longitudinal axons. PLoS Biol 14: e1002560. PubMed ID: 27654876

    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

    Hernandez, K., Myers, L. G., Bowser, M. and Kidd, T. (2015). Genetic tools for the analysis of Drosophila stomatogastric nervous system development. PLoS One 10: e0128290. PubMed ID: 26053861

    Newquist, G., Hogan, J., Walker, K., Lamanuzzi, M., Bowser, M. and Kidd, T. (2013). Control of male and female fertility by the netrin axon guidance genes. PLoS One 8: e72524. PubMed ID: 23977313

    Newquist, G., Drennan, J. M., Lamanuzzi, M., Walker, K., Clemens, J. C., Kidd, T. (2013) Blocking Apoptotic Signaling Rescues Axon Guidance in Netrin Mutants. Cell Rep. PubMed ID: 23499445

    Kidd, T. (2009). Neuroscience. Crossing the line. Science 324: 893-894. PubMed ID: 19443775

    Andrews, G. L., Tanglao, S., Farmer, W. T., Morin, S., Brotman, S., Berberoglu, M. A., Price, H., Fernandez, G. C., Mastick, G. S., Charron, F. and Kidd, T. (2008). Dscam guides embryonic axons by Netrin-dependent and -independent functions. Development 135: 3839-3848. PubMed ID: 18948420

  • Dan Kiehart Department of Biology, Duke University
    Moore, R. P., Fogerson, S. M., Tulu, U. S., Yu, J. W., Cox, A. H., Sican, M. A., Li, D., Legant, W. R., Weigel, A. V., Crawford, J. M., Betzig, E. and Kiehart, D. P. (2022). Superresolution microscopy reveals actomyosin dynamics in medioapical arrays. Mol Biol Cell 33(11): ar94. PubMed ID: 35544300

    Sallee, J. L., Crawford, J. M., Singh, V. and Kiehart, D. P. (2021). Mutations in Drosophila crinkled/Myosin VIIA disrupt denticle morphogenesis. Dev Biol 470: 121-135. PubMed ID: 33248112

    Mortensen, R. D., Moore, R. P., Fogerson, S. M., Chiou, H. Y., Obinero, C. V., Prabhu, N. K., Wei, A. H., Crawford, J. M. and Kiehart, D. P. (2018). Identifying genetic players in cell sheet morphogenesis using a Drosophila deficiency screen for genes on Chromosome 2R involved in dorsal closure. G3 (Bethesda). PubMed ID: 29776969

    Aristotelous, A. C., Crawford, J. M., Edwards, G. S., Kiehart, D. P. and Venakides, S. (2018). Mathematical models of dorsal closure. Prog Biophys Mol Biol. PubMed ID: 29852207

    Lu, H., Sokolow, A., Kiehart, D. P. and Edwards, G. S. (2016). Quantifying dorsal closure in three dimensions. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27798232

    Wells, A. R., Zou, R. S., Tulu, U. S., Sokolow, A. C., Crawford, J. M., Edwards, G. S. and Kiehart, D. P. (2014). Complete canthi removal reveals that forces from the amnioserosa are alone sufficient to drive dorsal closure in Drosophila. Mol Biol Cell. PubMed ID: 25253724

    Hunter, G. L., Crawford, J. M., Genkins, J. Z. and Kiehart, D. P. (2013). Ion channels contribute to the regulation of cell sheet forces during Drosophila dorsal closure. Development. PubMed ID: 24306105

    Sokolow, A., Toyama, Y., Kiehart, D. P. and Edwards, G. S. (2012). Cell ingression and apical shape oscillations during dorsal closure in Drosophila. Biophys J 102: 969-979. PubMed ID: 22404919

    Franke, J. D., Montague, R. A. and Kiehart, D. P. (2010). Nonmuscle myosin II is required for cell proliferation, cell sheet adhesion and wing hair morphology during wing morphogenesis. Dev. Biol. 345(2): 117-32.

  • Amy Kiger, Cell and Developmental Biology, U. C. San Diego
    Fujita, N., Huang, W., Lin, T. H., Groulx, J. F., Jean, S., Kuchitsu, Y., Koyama-Honda, I., Mizushima, N., Fukuda, M. and Kiger, A. A. (2017). Genetic screen in Drosophila muscle identifies autophagy-mediated T-tubule remodeling and a Rab2 role in autophagy. Elife 6. PubMed ID: 28063257

    Velichkova, M., Juan, J., Kadandale, P., Jean, S., Ribeiro, I., Raman, V., Stefan, C. and Kiger, A. A. (2010). Drosophila Mtm and class II PI3K coregulate a PI(3)P pool with cortical and endolysosomal functions. J Cell Biol 190(3): 407-425. PubMed ID: 20696708

  • Changsoo Kim School of Biological Sciences and Technology, Chonnam National University,
    Jang, W., Lim, J. Y., Kang, S., Kim, M., Hwang, S. W. and Kim, C. (2022). Drosophila ppk19 encodes a proton-gated and mechanosensitive ion channel. Sci Rep 12(1): 18346. PubMed ID: 36319833

    To, V., Kim, H. J., Jang, W., Sreejith, P. and Kim, C. (2021). Lin28 and Imp are Required for Stability of Bowl Transcripts in Hub Cells of the Drosophila Testis. Dev Reprod 25(4): 313-319. PubMed ID: 35141457

    Malik, S., Jang, W., Kim, J. Y. and Kim, C. (2020). Mechanisms ensuring robust repression of the Drosophila female germline stem cell maintenance factor Nanos via posttranscriptional regulation. Faseb j. PubMed ID: 32654316

    Malik, S., Jang, W., Park, S. Y. Y., Kim, J. Y. Y., Kwon, K. S. and Kim, C. (2019). The target specificity of the RNA binding protein Pumilio is determined by distinct co-factors. Biosci Rep. PubMed ID: 31097674

    Sreejith, P., Jang, W., To, V., Hun Jo, Y., Biteau, B. and Kim, C. (2019). Lin28 is a critical factor in the function and aging of Drosophila testis stem cell niche. Aging (Albany NY). PubMed ID: 30713156

    Jang, W., Lee, S., Choi, S. I., Chae, H. S., Han, J., Jo, H., Hwang, S. W., Park, C. S. and Kim, C. (2019). Impairment of proprioceptive movement and mechanical mociception in Drosophila melanogaster larvae lacking Ppk30, a Drosophila member of the DEG/ENaC family. Genes Brain Behav: e12545. PubMed ID: 30675754

    Jang, W., Baek, M., Han, Y. S. and Kim, C. (2018). Duox mediates ultraviolet injury-induced nociceptive sensitization in Drosophila larvae. Mol Brain 11(1): 16. PubMed ID: 29540218
    Malik, S., Jang, W. and Kim, C. (2017). Protein interaction mapping of translational regulators affecting expression of the critical stem cell factor nos. Dev Reprod 21(4): 449-456. PubMed ID: 29354790

    Jang, W., Kim, J. Y., Cui, S., Jo, J., Lee, B. C., Lee, Y., Kwon, K. S., Park, C. S. and Kim, C. (2014). The Anoctamin family channel Subdued mediates thermal nociception in Drosophila. J Biol Chem. PubMed ID: 25505177

    Kim, S. Y., Kim, J. Y., Malik, S., Son, W., Kwon, K. S. and Kim, C. (2012). Negative regulation of EGFR/MAPK pathway by Pumilio in Drosophila melanogaster. PLoS One 7: e34016. PubMed ID: 22514614

  • Eun-Young Kim Northwestern University Feinberg School of Medicine, Chicago
    Lee, S. H. and Kim, E. Y. (2021). Short-term maintenance on a high-sucrose diet alleviates aging-induced sleep fragmentation in drosophila. Anim Cells Syst (Seoul) 25(6): 377-386. PubMed ID: 35059137

    Jeong, E. B., Jeong, S. S., Cho, E. and Kim, E. Y. (2019). Makorin 1 is required for Drosophila oogenesis by regulating insulin/Tor signaling. PLoS One 14(4): e0215688. PubMed ID: 31009498 Cho, E., Kwon, M., Jung, J., Hyun Kang, D., Jin, S., Choi, S. E., Kang, Y. and Kim, E. Y. (2019). AMP-activated protein kinase regulates circadian rhythm by affecting CLOCK in Drosophila. J Neurosci. PubMed ID: 30819799

    Tran, H. T., Cho, E., Jeong, S., Jeong, E. B., Lee, H. S., Jeong, S. Y., Hwang, J. S. and Kim, E. Y. (2018). Makorin 1 regulates developmental timing in Drosophila. Mol Cells. PubMed ID: 30396233

    Cho, E., Lee, E. and Kim, E. Y. (2016). Diversification of molecular clockwork for tissue specific function: insight from novel Drosophila Clock mutant homologous to mouse Clock allele. BMB Rep [Epub ahead of print]. PubMed ID: 27756446

    Lee, E., Cho, E., Kang, D. H., Jeong, E. H., Chen, Z., Yoo, S. H. and Kim, E. Y. (2016). Pacemaker-neuron-dependent disturbance of the molecular clockwork by a Drosophila CLOCK mutant homologous to the mouse Clock mutation. Proc Natl Acad Sci U S A 113: E4904-4913. PubMed ID: 27489346

    Kang, S. W., Lee, E., Cho, E., Seo, J. H., Ko, H. W. and Kim, E. Y. (2015). Drosophila peptidyl-prolyl isomerase Pin1 modulates circadian rhythms via regulating levels of PERIOD. Biochem Biophys Res Commun 463: 235-240. PubMed ID: 25998391

  • Michael Kim Department of Molecular and Cellular Pharmacology, University of Miami
    Kamiyama, D., McGorty, R., Kamiyama, R., Kim, M. D., Chiba, A. and Huang, B. (2015). Specification of Dendritogenesis Site in Drosophila aCC Motoneuron by Membrane Enrichment of Pak1 through Dscam1. Dev Cell 35: 93-106. PubMed ID: 26460947

    Chang, X., Kim, M. D., Stephens, R., Qu, T., Chiba, A. and Tsechpenakis, G. (2014). Part-based motor neuron recognition in the Drosophila ventral nerve cord. Neuroimage 90: 33-42. PubMed ID: 24373882

    Boulina, M., Samarajeewa, H., Baker, J. D., Kim, M. D. and Chiba, A. (2013). Live imaging of multicolor-labeled cells in Drosophila. Development 140: 1605-1613. PubMed ID: 23482495

  • Seung Kim Stanford University School of Medicine
    Alfa, R.W., Park, S., Skelly, K.R., Poffenberger, G., Jain, N., Gu, X., Kockel, L., Wang, J., Liu, Y., Powers, A.C. and Kim, S.K. (2015). Suppression of insulin production and secretion by a Decretin hormone. Cell Metab 21: 323-333. PubMed ID: 25651184

    Park, S., Alfa, R. W., Topper, S. M., Kim, G. E., Kockel, L. and Kim, S. K. (2014). A genetic strategy to measure circulating Drosophila insulin reveals genes regulating insulin production and secretion. PLoS Genet 10: e1004555. PubMed ID: 25101872

    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 197: 175-192. PubMed ID: 24558258

    Park, S., Bustamante, E. L., Antonova, J., McLean, G. W. and Kim, S. K. (2011). Specification of Drosophila corpora cardiaca neuroendocrine cells from mesoderm is regulated by Notch signaling. PLoS Genet 7: e1002241. PubMed ID: 21901108

  • Young Joon Kim Department of Biology, Yonsei University, Seoul
    Kim, Y. J. (2022). Activity-induced synaptic structural modifications by Akt. Biochem Biophys Res Commun 621: 94-100. PubMed ID: 35820284

    Zhang, C., Kim, A. J., Rivera-Perez, C., Noriega, F. G. and Kim, Y. J. (2022). The insect somatostatin pathway gates vitellogenesis progression during reproductive maturation and the post-mating response. Nat Commun 13(1): 969. PubMed ID: 35181671

    Zhang, C., Daubnerova, I., Jang, Y. H., Kondo, S., Zitnan, D. and Kim, Y. J. (2021). The neuropeptide allatostatin C from clock-associated DN1p neurons generates the circadian rhythm for oogenesis. Proc Natl Acad Sci U S A 118(4). PubMed ID: 33479181

    Lee, J. H., Lee, N. R., Kim, D. H. and Kim, Y. J. (2020). Molecular characterization of ligand selectivity of the sex peptide receptors of Drosophila melanogaster and Aedes aegypti. Insect Biochem Mol Biol: 103472. PubMed ID: 32971207

    Jang, Y. H., Chae, H. S. and Kim, Y. J. (2017). Female-specific myoinhibitory peptide neurons regulate mating receptivity in Drosophila melanogaster. Nat Commun 8(1): 1630. PubMed ID: 29158481

    Lee, H., Choi, H.W., Zhang, C., Park, Z.Y. and Kim, Y.J. (2016). A pair of oviduct-born Pickpocket neurons important for egg-laying in Drosophila melanogaster. Mol Cells [Epub ahead of print]. PubMed ID: 27378227

    Lee, K. M., Daubnerova, I., Isaac, R. E., Zhang, C., Choi, S., Chung, J. and Kim, Y. J. (2015). A Neuronal Pathway that Controls Sperm Ejection and Storage in Female Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 25702579

    Chen, J., Choi, M. S., Mizoguchi, A., Veenstra, J. A., Kang, K., Kim, Y. J. and Kwon, J. Y. (2015). Isoform-specific expression of the neuropeptide orcokinin in Drosophila melanogaster. Peptides. PubMed ID: 25600008

    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

    Isaac, R. E., Kim, Y. J. and Audsley, N. (2014). The degradome and the evolution of Drosophila sex peptide as a ligand for the MIP receptor. Peptides 53: 258-264. PubMed ID: 24398368

  • Elizabeth King Division of Biological Sciences, University of Missouri-Columbia
    Ng'oma, E., Williams-Simon, P. A., Rahman, A. and King, E. G. (2020). Diverse biological processes coordinate the transcriptional response to nutritional changes in a Drosophila melanogaster multiparent population. BMC Genomics 21(1): 84. PubMed ID: 31992183

    Ng'oma, E., Fidelis, W., Middleton, K. M. and King, E. G. (2019). The evolutionary potential of diet-dependent effects on lifespan and fecundity in a multi-parental population of Drosophila melanogaster. Heredity (Edinb) 122(5): 582-594. PubMed ID: 30356225

    Stanley, P. D., Ng'oma, E., O'Day, S. and King, E. G. (2017). Genetic Dissection of Nutrition-Induced Plasticity in Insulin/Insulin-Like Growth Factor Signaling and Median Life Span in a Drosophila Multiparent Population. Genetics 206(2): 587-602. PubMed ID: 28592498

    King, E. G. and Long, A. D. (2017). The Beavis Effect in Next-Generation Mapping Panels in Drosophila melanogaster. G3 (Bethesda) 7(6): 1643-1652. PubMed ID: 28592647

    Najarro, M. A., Hackett, J. L., Smith, B. R., Highfill, C. A., King, E. G., Long, A. D. and Macdonald, S. J. (2015). Identifying Loci Contributing to Natural Variation in Xenobiotic Resistance in Drosophila. PLoS Genet 11(11): e1005663. PubMed ID: 26619284

  • Kirst King-Jones University of Alberta
    Zeng, J., Huynh, N., Phelps, B. and King-Jones, K. (2020). Snail synchronizes endocycling in a TOR-dependent manner to coordinate entry and escape from endoreplication pausing during the Drosophila critical weight checkpoint. PLoS Biol 18(2): e3000609. PubMed ID: 32097403

    Huynh, N., Ou, Q., Cox, P., Lill, R. and King-Jones, K. (2019). Glycogen branching enzyme controls cellular iron homeostasis via Iron Regulatory Protein 1 and mitoNEET. Nat Commun 10(1): 5463. PubMed ID: 31784520

    Huynh, N., Zeng, J., Liu, W. and King-Jones, K. (2018). A Drosophila CRISPR/Cas9 toolkit for conditionally manipulating gene expression in the prothoracic gland as a test case for polytene tissues. G3 (Bethesda). PubMed ID: 30213867

    Zeng, J., Kamiyama, T., Niwa, R. and King-Jones, K. (2018). The Drosophila CCR4-NOT complex is required for cholesterol homeostasis and steroid hormone synthesis. Dev Biol. PubMed ID: 30149007

    Di Cara, F. and King-Jones, K. (2016). The Circadian clock is a key driver of steroid hormone production in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 27546572

    Ou, Q., Zeng, J., Yamanaka, N., Brakken-Thal, C., O'Connor, M. B. and King-Jones, K. (2016). The insect prothoracic gland as a model for steroid hormone biosynthesis and regulation. Cell Rep 16: 247-262. PubMed ID: 27320926

    Di Cara, F., Maile, T. M., Parsons, B. D., Magico, A., Basu, S., Tapon, N. and King-Jones, K. (2015). The Hippo pathway promotes cell survival in response to chemical stress. Cell Death Differ [Epub ahead of print]. PubMed ID: 26021298

    Di Cara, F. and King-Jones, K. (2013). How clocks and hormones act in concert to control the timing of insect development. Curr Top Dev Biol 105: 1-36. PubMed ID: 23962837

    Ou, Q., King-Jones, K. (2013) What goes up must come down: transcription factors have their say in making ecdysone pulses. Curr Top Dev Biol 103: 35-71. PubMed ID: 23347515

    Ou, Q., Magico, A. and King-Jones, K. (2011). Nuclear receptor DHR4 controls the timing of steroid hormone pulses during Drosophila development. PLoS Biol 9: e1001160. PubMed ID: 21980261

  • Toshihiro Kitamoto Department of Neuroscience, University of Iowa, Iowa City
    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

    Kaas, G. A., Kasuya, J., Lansdon, P., Ueda, A., Iyengar, A., Wu, C. F. and Kitamoto, T. (2016). Lithium-responsive seizure-like hyperexcitability is caused by a mutation in the Drosophila boltage-gated Sodium channel gene paralytic. eNeuro 3. PubMed ID: 27844061

    Petruccelli, E., Li, Q., Rao, Y. and Kitamoto, T. (2016). The unique dopamine/ecdysteroid receptor modulates ethanol-induced sedation in Drosophila. J Neurosci 36: 4647-4657. PubMed ID: 27098705

    Petruccelli, E., Lansdon, P. and Kitamoto, T. (2015). Exaggerated nighttime sleep and defective sleep homeostasis in a Drosophila knock-in model of human epilepsy. PLoS One 10: e0137758. PubMed ID: 26361221

    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

    Ishimoto, H., Wang, Z., Rao, Y., Wu, C. F. and Kitamoto, T. (2013). A Novel Role for Ecdysone in Drosophila Conditioned Behavior: Linking GPCR-Mediated Non-canonical Steroid Action to cAMP Signaling in the Adult Brain. PLoS Genet 9: e1003843. PubMed ID: 24130506

    Sakai, T., Sato, S., Ishimoto, H. and Kitamoto, T. (2012). Significance of the centrally expressed TRP channel painless in Drosophila courtship memory. Learn Mem 20: 34-40. PubMed ID: 23247253

    Sakai, T., Inami, S., Sato, S. and Kitamoto, T. (2012). Fan-shaped body neurons are involved in period-dependent regulation of long-term courtship memory in Drosophila. Learn Mem 19: 571-574. PubMed ID: 23154928

    Ishimoto, H., Lark, A. and Kitamoto, T. (2012). Factors that Differentially Affect Daytime and Nighttime Sleep in Drosophila melanogaster. Front Neurol 3: 24. PubMed ID: 22375135

  • Robert Kittel Institute of Physiology, Dept. of Neurophysiology, University of Wuerzburg
    Dannhäuser, S., Lux, T. J., Hu, C., Selcho, M., Chen, J. T., Ehmann, N., Sachidanandan, D., Stopp, S., Pauls, D., Pawlak, M., Langenhan, T., Soba, P., Rittner, H. L. and Kittel, R. J. (2020). Antinociceptive modulation by the adhesion GPCR CIRL promotes mechanosensory signal discrimination. Elife 9. PubMed ID: 32996461

    Scholz, N., Ehmann, N., Sachidanandan, D., Imig, C., Cooper, B. H., Jahn, O., Reim, K., Brose, N., Meyer, J., Lamberty, M., Altrichter, S., Bormann, A., Hallermann, S., Pauli, M., Heckmann, M., Stigloher, C., Langenhan, T. and Kittel, R. J. (2019). Complexin cooperates with Bruchpilot to tether synaptic vesicles to the active zone cytomatrix. J Cell Biol 218(3): 1011-1026. PubMed ID: 30782781

    Scholz, N., Guan, C., Nieberler, M., Grotemeyer, A., Maiellaro, I., Gao, S., Beck, S., Pawlak, M., Sauer, M., Asan, E., Rothemund, S., Winkler, J., Promel, S., Nagel, G., Langenhan, T. and Kittel, R. J. (2017). Mechano-dependent signaling by Latrophilin/CIRL quenches cAMP in proprioceptive neurons. Elife 6. PubMed ID: 28784204

    Dawydow, A., Gueta, R., Ljaschenko, D., Ullrich, S., Hermann, M., Ehmann, N., Gao, S., Fiala, A., Langenhan, T., Nagel, G. and Kittel, R. J. (2014). Channelrhodopsin-2-XXL, a powerful optogenetic tool for low-light applications. Proc Natl Acad Sci U S A. PubMed ID: 25201989

    Ljaschenko, D., Ehmann, N. and Kittel, R. J. (2013). Hebbian Plasticity Guides Maturation of Glutamate Receptor Fields In Vivo. Cell Rep. PubMed ID: 23643532

    Hallermann, S., Kittel, R. J., Wichmann, C., Weyhersmuller, A., Fouquet, W., Mertel, S., Owald, D., Eimer, S., Depner, H., Schwarzel, M., Sigrist, S. J. and Heckmann, M. (2010). Naked dense bodies provoke depression. J Neurosci 30: 14340-14345. PubMed ID: 20980589

    Hallermann, S., Heckmann, M. and Kittel, R. J. (2010). Mechanisms of short-term plasticity at neuromuscular active zones of Drosophila. HFSP J 4: 72-84. PubMed ID: 20811513

    Bachmann, A., Kobler, O., Kittel, R. J., Wichmann, C., Sierralta, J., Sigrist, S. J., Gundelfinger, E. D., Knust, E. and Thomas, U. (2010). A perisynaptic menage a trois between Dlg, DLin-7, and Metro controls proper organization of Drosophila synaptic junctions. J Neurosci 30: 5811-5824. PubMed ID: 20427642

  • Ole Kjaerulff Molecular Neuropharmacology and Genetics Laboratory, University of Copenhaven
    Lund, V. K., Lycas, M. D., Schack, A., Andersen, R. C., Gether, U. and Kjaerulff, O. (2021). Rab2 drives axonal transport of dense core vesicles and lysosomal organelles. Cell Rep 35(2): 108973. PubMed ID: 33852866

    Lund, V. K., Madsen, K. L. and Kjaerulff, O. (2018). Drosophila Rab2 controls endosome-lysosome fusion and LAMP delivery to late endosomes. Autophagy. PubMed ID: 29940804

    Gerdoe-Kristensen, S., Lund, V. K., Wandall, H. H. and Kjaerulff, O. (2016). Mactosylceramide prevents glial cell overgrowth by inhibiting insulin and fibroblast growth factor receptor signaling. J Cell Physiol [Epub ahead of print]. PubMed ID: 28019653

    Dahlgaard, K., Jung, A., Qvortrup, K., Clausen, H., Kjaerulff, O. and Wandall, H. H. (2012). Neurofibromatosis-like phenotype in Drosophila caused by lack of glucosylceramide extension. Proc Natl Acad Sci U S A 109(18): 6987-6992. PubMed ID: 22493273

    Jansen, A. M., Nassel, D. R., Madsen, K. L., Jung, A. G., Gether, U. and Kjaerulff, O. (2009). PICK1 expression in the Drosophila central nervous system primarily occurs in the neuroendocrine system. J Comp Neurol 517(3): 313-332. PubMed ID: 19757495

  • Louis Bernard Klaczko Departamento de Genetica e Evolucao, Instituto de Biologia, Universidade Estadual de Campinas, Brasil
    Brianti, M. T., Ananina, G., Klaczko, L. B. (2013) Differential occurrence of chromosome inversion polymorphisms among Muller's elements in three species of the tripunctata group of Drosophila, including a species with fast chromosomal evolution. Genome 56: 17-26. PubMed ID: 23379335

    Rocha, F. B. and Klaczko, L. B. (2012). Connecting the dots of nonlinear reaction norms unravels the threads of genotype-environment interaction in Drosophila. Evolution 66: 3404-3416. PubMed ID: 23106706

    Ventura, I. M., Martins, A. B., Lyra, M. L., Andrade, C. A., Carvalho, K. A. and Klaczko, L. B. (2012). Spiroplasma in Drosophila melanogaster populations: prevalence, male-killing, molecular identification, and no association with Wolbachia. Microb Ecol 64: 794-801. PubMed ID: 22562106

  • Christian Klämbt Universität Münster
    Kanel, P., Noll, G. A., Schroedter, K., Naffin, E., Kronenberg, J., Busswinkel, F., Twyman, R. M., Klambt, C. and Prufer, D. (2022). The tobacco phosphatidylethanolamine-binding protein NtFT4 increases the lifespan of Drosophila melanogaster by interacting with the proteostasis network. Aging (Albany NY) 14(undefined). PubMed ID: 35396341

    Yildirim, K., Winkler, B., Pogodalla, N., Mackensen, S., Baldenius, M., Garcia, L., Naffin, E., Rodrigues, S. and Klambt, C. (2022). Redundant functions of the SLC5A transporters Rumpel, Bumpel, and Kumpel in ensheathing glial cells. Biol Open 11(1). PubMed ID: 34897385

    Pogodalla, N., Kranenburg, H., Rey, S., Rodrigues, S., Cardona, A. and Klambt, C. (2021). Drosophila ß(Heavy)-Spectrin is required in polarized ensheathing glia that form a diffusion-barrier around the neuropil. Nat Commun 12(1): 6357. PubMed ID: 34737284

    Kottmeier, R., Bittern, J., Schoofs, A., Scheiwe, F., Matzat, T., Pankratz, M. and Klambt, C. (2020). Wrapping glia regulates neuronal signaling speed and precision in the peripheral nervous system of Drosophila. Nat Commun 11(1): 4491. PubMed ID: 32901033

    Neuert, H., Deing, P., Krukkert, K., Naffin, E., Steffes, G., Risse, B., Silies, M. and Klambt, C. (2019). The Drosophila NCAM homolog Fas2 signals independent of adhesion. Development. PubMed ID: 31862845

    Dlugos, C. P., Picciotto, C., Lepa, C., Krakow, M., Stober, A., Eddy, M. L., Weide, T., Jeibmann, A., Krahn, M. P., Van Marck, V., Klingauf, J., Ricker, A., Wedlich-Soldner, R., Pavenstadt, H., Klambt, C. and George, B. (2019). Nephrin Signaling Results in Integrin beta1 Activation. J Am Soc Nephrol. PubMed ID: 31097607

    Petri, J., Syed, M. H., Rey, S. and Klambt, C. (2019). Non-cell-autonomous function of the GPI-anchored protein Undicht during septate junction assembly. Cell Rep 26(6): 1641-1653.e1644. PubMed ID: 30726744

    Babatz, F., Naffin, E. and Klambt, C. (2018). The Drosophila blood-brain barrier adapts to cell growth by unfolding of pre-existing septate junctions. Dev Cell. PubMed ID: 30482667

    Kastl, P., Manikowski, D., Steffes, G., Schurmann, S., Bandari, S., Klambt, C. and Grobe, K. (2018). Disrupting Hedgehog Cardin-Weintraub sequence and positioning changes cellular differentiation and compartmentalization in vivo. Development 145(18). PubMed ID: 30242104

    Otto, N., Marelja, Z., Schoofs, A., Kranenburg, H., Bittern, J., Yildirim, K., Berh, D., Bethke, M., Thomas, S., Rode, S., Risse, B., Jiang, X., Pankratz, M., Leimkuhler, S. and Klambt, C. (2018). The sulfite oxidase Shopper controls neuronal activity by regulating glutamate homeostasis in Drosophila ensheathing glia. Nat Commun 9(1): 3514. PubMed ID: 30158546

  • Thomas Klein Institiut für Genetik der Heinrich Heine Universität Düsseldorf
    Schnute, B., Shimizu, H., Lyga, M., Baron, M. and Klein, T. (2022). Ubiquitylation is required for the incorporation of the Notch receptor into intraluminal vesicles to prevent prolonged and ligand-independent activation of the pathway. BMC Biol 20(1): 65. PubMed ID: 35264151

    Pannen, H., Rapp, T. and Klein, T. (2020). The ESCRT Machinery regulates Retromer dependent Transcytosis of Septate Junction Components in Drosophila. Elife 9. PubMed ID: 33377869

    Baeumers, M., Ruhnau, K., Breuer, T., Pannen, H., Goerlich, B., Kniebel, A., Haensch, S., Weidtkamp-Peters, S., Schmitt, L. and Klein, T. (2020). Lethal (2) giant discs (Lgd)/CC2D1 is required for the full activity of the ESCRT machinery. BMC Biol 18(1): 200. PubMed ID: 33349255

    Baumers, M., Klose, S., Bruser, C., Haag, C., Hansch, S., Pannen, H., Weidtkamp-Peters, S., Feldbrugge, M. and Klein, T. (2019). The auxiliary ESCRT complexes provide robustness to cold in poikilothermic organisms. Biol Open 8(9). PubMed ID: 31412999

    Morawa, K.S., Schneider, M. and Klein, T. (2015). Lgd regulates the activity of the BMP/Dpp signalling pathway during Drosophila oogenesis. Development 142: 1325-1335. PubMed ID: 25804739

    Troost, T., Schneider, M. and Klein, T. (2015). A Re-examination of the Selection of the Sensory Organ Precursor of the Bristle Sensilla of Drosophila melanogaster. PLoS Genet 11: e1004911. PubMed ID: 25569355

    Yousefian, J., Troost, T., Grawe, F., Sasamura, T., Fortini, M., Klein, T. (2013) Dmon1 controls recruitment of Rab7 to maturing endosomes in Drosophila. J Cell Sci. PubMed ID: 23418349

    Schneider, M., Troost, T., Grawe, F., Martinez-Arias, A. and Klein, T. (2012). Activation of Notch in lgd mutant cells requires the fusion of late endosomes with the lysosome. J Cell Sci. PubMed ID: 23178945

    Troost, T. and Klein, T. (2012). Sequential Notch signalling at the boundary of fringe expressing and non-expressing cells. PLoS One 7: e49007. PubMed ID: 23152840

    Troost, T., Jaeckel, S., Ohlenhard, N. and Klein, T. (2012). The tumour suppressor Lethal (2) giant discs is required for the function of the ESCRT-III component Shrub/CHMP4. J Cell Sci 125: 763-776. PubMed ID: 22389409

  • Mikhail Klenov Institute of Molecular Genetics Russian Academy of Sciences, Moskow
    Fefelova, E. A., Pleshakova, I. M., Mikhaleva, E. A., Pirogov, S. A., Poltorachenko, V. A., Abramov, Y. A., Romashin, D. D., Shatskikh, A. S., Blokh, R. S., Gvozdev, V. A. and Klenov, M. S. (2022). Impaired function of rDNA transcription initiation machinery leads to derepression of ribosomal genes with insertions of R2 retrotransposon. Nucleic Acids Res 50(2): 867-884. PubMed ID: 35037046

    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. PubMed ID: 32500264

    Sokolova, O. A., Mikhaleva, E. A., Kharitonov, S. L., Abramov, Y. A., Gvozdev, V. A. and Klenov, M. S. (2020). Special vulnerability of somatic niche cells to transposable element activation in Drosophila larval ovaries. Sci Rep 10(1): 1076. PubMed ID: 31974416

    Ryazansky, S. S., Stolyarenko, A. D., Klenov, M. S. and Gvozdev, V. A. (2017). Induction of Transposon Silencing in the Drosophila Germline. Biochemistry (Mosc) 82(5): 565-571. PubMed ID: 28601066

    Ilyin, A. A., Ryazansky, S. S., Doronin, S. A., Olenkina, O. M., Mikhaleva, E. A., Yakushev, E. Y., Abramov, Y. A., Belyakin, S. N., Ivankin, A. V., Pindyurin, A. V., Gvozdev, V. A., Klenov, M. S. and Shevelyov, Y. Y. (2017). Piwi interacts with chromatin at nuclear pores and promiscuously binds nuclear transcripts in Drosophila ovarian somatic cells. Nucleic Acids Res 45(13): 7666-7680. PubMed ID: 28472469

    Klenov, M. S., Lavrov, S. A., Korbut, A. P., Stolyarenko, A. D., Yakushev, E. Y., Reuter, M., Pillai, R. S. and Gvozdev, V. A. (2014). Impact of nuclear Piwi elimination on chromatin state in Drosophila melanogaster ovaries. Nucleic Acids Res 42(10): 6208-6218. PubMed ID: 24782529

  • Peter Klepsatel Institute of Zoology, Bratislava, Slovakia
    Galikova, M. and Klepsatel, P. (2022). Ion transport peptide regulates energy intake, expenditure, and metabolic homeostasis in Drosophila. Genetics 222(4). PubMed ID: 36190340

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

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

  • Markus Knaden Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena
    Khallaf, M. A., Cui, R., Weissflog, J., Erdogmus, M., Svatos, A., Dweck, H. K. M., Valenzano, D. R., Hansson, B. S. and Knaden, M. (2021). Large-scale characterization of sex pheromone communication systems in Drosophila. Nat Commun 12(1): 4165. PubMed ID: 34230464

    Khallaf, M. A., Auer, T. O., Grabe, V., Depetris-Chauvin, A., Ammagarahalli, B., Zhang, D. D., Lavista-Llanos, S., Kaftan, F., Weissflog, J., Matzkin, L. M., Rollmann, S. M., Lofstedt, C., Svatos, A., Dweck, H. K. M., Sachse, S., Benton, R., Hansson, B. S. and Knaden, M. (2020). Mate discrimination among subspecies through a conserved olfactory pathway. Sci Adv 6(25): eaba5279. PubMed ID: 32704542

    Qiao, H., Keesey, I. W., Hansson, B. S. and Knaden, M. (2019). Gut microbiota affects development and olfactory behavior in Drosophila melanogaster. J Exp Biol. PubMed ID: 30679242

    Koerte, S., Keesey, I. W., Khallaf, M. A., Cortes Llorca, L., Grosse-Wilde, E., Hansson, B. S. and Knaden, M. (2018). Evaluation of the DREAM Technique for a High-Throughput Deorphanization of Chemosensory Receptors in Drosophila. Front Mol Neurosci 11: 366. PubMed ID: 30356801

    Dweck, H. K. M., Ebrahim, S. A. M., Retzke, T., Grabe, V., Weissflog, J., Svatos, A., Hansson, B. S. and Knaden, M. (2018). The olfactory logic behind fruit odor preferences in larval and adult Drosophila. Cell Rep 23(8): 2524-2531. PubMed ID: 29791860

    Keesey, I. W., Koerte, S., Khallaf, M. A., Retzke, T., Guillou, A., Grosse-Wilde, E., Buchon, N., Knaden, M. and Hansson, B. S. (2017). Pathogenic bacteria enhance dispersal through alteration of Drosophila social communication. Nat Commun 8(1): 265. PubMed ID: 28814724

    Retzke, T., Thoma, M., Hansson, B. S. and Knaden, M. (2017). Potencies of effector genes in silencing odor-guided behavior in Drosophila melanogaster. J Exp Biol. PubMed ID: 28235908

    Keesey, I. W., Koerte, S., Retzke, T., Haverkamp, A., Hansson, B. S. and Knaden, M. (2016). Adult frass provides a pheromone signature for Drosophila feeding and aggregation. J Chem Ecol [Epub ahead of print]. PubMed ID: 27539589

    Ebrahim, S. A., Dweck, H. K., Stokl, J., Hofferberth, J. E., Trona, F., Weniger, K., Rybak, J., Seki, Y., Stensmyr, M. C., Sachse, S., Hansson, B. S. and Knaden, M. (2015). Drosophila Avoids Parasitoids by Sensing Their Semiochemicals via a Dedicated Olfactory Circuit. PLoS Biol 13: e1002318. PubMed ID: 26674493

    Thoma, M., Hansson, B. S. and Knaden, M. (2015). High-resolution quantification of odor-guided behavior in Drosophila melanogaster using the flywalk paradigm. J Vis Exp [Epub ahead of print]. PubMed ID: 26709624

    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

  • Jürgen Knoblich Institute of Molecular Biotechnology, Vienna
    Bonnay, F., Veloso, A., Steinmann, V., Köcher, T., Abdusselamoglu, M. D., Bajaj, S., Rivelles, E., Landskron, L., Esterbauer, H., Zinzen, R. P. and Knoblich, J. A. (2020). Oxidative Metabolism Drives Immortalization of Neural Stem Cells during Tumorigenesis. Cell 182(6): 1490-1507.e1419. PubMed ID: 32916131

    Abdusselamoglu, M. D., Landskron, L., Bowman, S. K., Eroglu, E., Burkard, T., Kingston, R. E. and Knoblich, J. A. (2019). Dynamics of activating and repressive histone modifications in Drosophila neural stem cell lineages and brain tumors. Development. PubMed ID: 31748204

    Abdusselamoglu, M. D., Eroglu, E., Burkard, T. R. and Knoblich, J. A. (2019). The transcription factor odd-paired regulates temporal identity in transit-amplifying neural progenitors via an incoherent feed-forward loop. Elife 8. PubMed ID: 31329099

    Wissel, S., Harzer, H., Bonnay, F., Burkard, T. R., Neumuller, R. A. and Knoblich, J. A. (2018). Time-resolved transcriptomics in neural stem cells identifies a v-ATPase/Notch regulatory loop. J Cell Biol. PubMed ID: 29959232

    Wissel, S., Harzer, H., Bonnay, F., Burkard, T. R., Neumuller, R. A. and Knoblich, J. A. (2018). Time-resolved transcriptomics in neural stem cells identifies a v-ATPase/Notch regulatory loop. J Cell Biol. PubMed ID: 29959232

    Landskron, L., Steinmann, V., Bonnay, F., Burkard, T. R., Steinmann, J., Reichardt, I., Harzer, H., Laurenson, A. S., Reichert, H. and Knoblich, J. A. (2018). The asymmetrically segregating lncRNA cherub is required for transforming stem cells into malignant cells. Elife 7. PubMed ID: 29580384

    Reichardt, I., Bonnay, F., Steinmann, V., Loedige, I., Burkard, T. R., Meister, G. and Knoblich, J. A. (2017). The tumor suppressor Brat controls neuronal stem cell lineages by inhibiting Deadpan and Zelda. EMBO Rep. PubMed ID: 29191977

    Abramczuk, M. K., Burkard, T. R., Rolland, V., Steinmann, V., Duchek, P., Jiang, Y., Wissel, S., Reichert, H. and Knoblich, J. A. (2017). The splicing co-factor Barricade/Tat-SF1, is required for cell cycle and lineage progression in Drosophila neural stem cells. Development. PubMed ID: 28935704

    Wissel, S., Kieser, A., Yasugi, T., Duchek, P., Roitinger, E., Gokcezade, J. F., Steinmann, V., Gaul, U., Mechtler, K., Forstemann, K., Knoblich, J. A. and Neumuller, R. A. (2016). A combination of CRISPR/Cas9 and standardized RNAi as a versatile platform for the characterization of gene function. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27280787

    Marchetti, G., Reichardt, I., Knoblich, J. A. and Besse, F. (2014). The TRIM-NHL Protein Brat Promotes Axon Maintenance by Repressing src64B Expression. J Neurosci 34: 13855-13864. PubMed ID: 25297111

    Homem, C. C., Steinmann, V., Burkard, T. R., Jais, A., Esterbauer, H. and Knoblich, J. A. (2014). Ecdysone and mediator change energy metabolism to terminate proliferation in Drosophila neural stem cells. Cell 158: 874-888. PubMed ID: 25126791

    Mauri, F., Reichardt, I., Mummery-Widmer, J. L., Yamazaki, M., Knoblich, J. A. (2014) The conserved Discs-large binding partner Banderuola regulates asymmetric cell division in Drosophila. Curr Biol. PubMed ID: 25088559

  • Elizabeth Knust Max Planck Institute of Molecular Cell Biology and Genetics, Dresden
    Hebbar, S., Lehmann, M., Behrens, S., Halsig, C., Leng, W., Yuan, M., Winkler, S. and Knust, E. (2021). Mutations in the splicing regulator Prp31 lead to retinal degeneration in Drosophila. Biol Open 10(1). PubMed ID: 33495354

    Bhagavatula, S. and Knust, E. (2020). A putative stem-loop structure in Drosophila crumbs is required for mRNA localisation in epithelia and germline cells. J Cell Sci. PubMed ID: 33310910

    Hebbar, S., Schuhmann, K., Shevchenko, A. and Knust, E. (2020). Hydroxylated sphingolipid biosynthesis regulates photoreceptor apical domain morphogenesis. J Cell Biol 219(12). PubMed ID: 33048164

    Raghuraman, B. K., Hebbar, S., Kumar, M., Moon, H., Henry, I., Knust, E. and Shevchenko, A. (2020). Absolute Quantification of Proteins in the Eye of Drosophila melanogaster. Proteomics: e1900049. PubMed ID: 32663363

    Kraut, R. S. and Knust, E. (2019). Changes in endolysosomal organization define a pre-degenerative state in the crumbs mutant Drosophila retina. PLoS One 14(12): e0220220. PubMed ID: 31834921

    Lattner, J., Leng, W., Knust, E., Brankatschk, M. and Flores-Benitez, D. (2019). Crumbs organizes the transport machinery by regulating apical levels of PI(4,5)P2 in Drosophila. Elife 8. PubMed ID: 31697234

    Skouloudaki, K., Christodoulou, I., Khalili, D., Tsarouhas, V., Samakovlis, C., Tomancak, P., Knust, E. and Papadopoulos, D. K. (2019). Yorkie controls tube length and apical barrier integrity during airway development. J Cell Biol. PubMed ID: 31315941

    Bajur, A. T., Iyer, K. V. and Knust, E. (2019). Cytocortex-dependent dynamics of Drosophila Crumbs controls junctional stability and tension during germ band retraction. J Cell Sci. PubMed ID: 31300472

    Hartenstein, V., Yuan, M., Younossi-Hartenstein, A., Karandikar, A., Bernardo-Garcia, F. J., Sprecher, S. and Knust, E. (2019). Serial electron microscopic reconstruction of the Drosophila larval eye: Photoreceptors with a rudimentary rhabdomere of microvillar-like processes. Dev Biol. PubMed ID: 31158364

    Skouloudaki, K., Papadopoulos, D. K., Tomancak, P. and Knust, E. (2019). The apical protein Apnoia interacts with Crumbs to regulate tracheal growth and inflation. PLoS Genet 15(1): e1007852. PubMed ID: 30645584

    Tsoumpekos, G., Nemetschke, L. and Knust, E. (2018). Drosophila Big bang regulates the apical cytocortex and wing growth through junctional tension. J Cell Biol [Epub ahead of print]. PubMed ID: 29326288

    Das, S. and Knust, E. (2018). A dual role of the extracellular domain of Drosophila Crumbs for morphogenesis of the embryonic neuroectoderm. Biol Open 7(1). PubMed ID: 29374056

  • Satoru Kobayashi Okazaki Institute for Integrative Bioscience, Higashiyama, Myodaiji, Okazaki 444-8787, Japan
    Hayashi, Y., Kashio, S., Murotomi, K., Hino, S., Kang, W., Miyado, K., Nakao, M., Miura, M., Kobayashi, S. and Namihira, M. (2022). Biosynthesis of S-adenosyl-methionine enhances aging-related defects in Drosophila oogenesis. Sci Rep 12(1): 5593. PubMed ID: 35379840

    Masukawa, M., Ishizaki, Y., Miura, H., Hayashi, M., Ota, R. and Kobayashi, S. (2021). Male-biased protein expression in primordial germ cells, identified through a comparative study of UAS vectors in Drosophila. Sci Rep 11(1): 21482. PubMed ID: 34728669

    Ota, R., Hayashi, M., Morita, S., Miura, H. and Kobayashi, S. (2021). Absence of X-chromosome dosage compensation in the primordial germ cells of Drosophila embryos. Sci Rep 11(1): 4890. PubMed ID: 33649478

    Ota, R. and Kobayashi, S. (2020). Myc plays an important role in Drosophila P-M hybrid dysgenesis to eliminate germline cells with genetic damage. Commun Biol 3(1): 185. PubMed ID: 32322015

    Morita, S., Ota, R., Hayashi, M. and Kobayashi, S. (2020). Repression of G1/S Transition by Transient Inhibition of miR-10404 Expression in Drosophila Primordial Germ Cells. iScience 23(3): 100950. PubMed ID: 32179474

    Asaoka, M., Hanyu-Nakamura, K., Nakamura, A. and Kobayashi, S. (2019). Maternal Nanos inhibits Importin-alpha2/Pendulin-dependent nuclear import to prevent somatic gene expression in the Drosophila germline. PLoS Genet 15(5): e1008090. PubMed ID: 31091233

    Morita, S., Ota, R. and Kobayashi, S. (2018). Downregulation of NHP2 promotes proper cyst formation in Drosophila ovary. Dev Growth Differ. PubMed ID: 29845608

    Sugimori, S., Kumata, Y. and Kobayashi, S. (2018). Maternal Nanos-dependent RNA stabilization in the primordial germ cells of Drosophila embryos. Dev Growth Differ 60(1): 63-75. PubMed ID: 29278271

    Ota, R., Morita, S., Sato, M., Shigenobu, S., Hayashi, M. and Kobayashi, S. (2017). Transcripts immunoprecipitated with Sxl protein in primordial germ cells of Drosophila embryos. Dev Growth Differ. PubMed ID: 29124738

    Hayashi, M., Shinozuka, Y., Shigenobu, S., Sato, M., Sugimoto, M., Ito, S., Abe, K. and Kobayashi, S. (2017). Conserved role of Ovo in germline development in mouse and Drosophila. Sci Rep 7: 40056. PubMed ID: 28059165

    Mukai, M., Hira, S., Nakamura, K., Nakamura, S., Kimura, H., Sato, M. and Kobayashi, S. (2015). H3K36 trimethylation-mediated epigenetic regulation is activated by Bam and promotes germ cell differentiation during early oogenesis in Drosophila. Biol Open. PubMed ID: 25572421

    Ohhara, Y., Shimada-Niwa, Y., Niwa, R., Kayashima, Y., Hayashi, Y., Akagi, K., Ueda, H., Yamakawa-Kobayashi, K. and Kobayashi, S. (2015). Autocrine regulation of ecdysone synthesis by beta3-octopamine receptor in the prothoracic gland is essential for Drosophila metamorphosis. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 25605909

  • Dalibor Kodrík Institute of Entomology, Ceské Budêjovice, Czech Republic
    Mochanova, M., Tomcala, A., Svobodova, Z. and Kodrik, D. (2018). Role of adipokinetic hormone during starvation in Drosophila. Comp Biochem Physiol B Biochem Mol Biol 226: 26-35. PubMed ID: 30110658

    Bednarova, A., Tomcala, A., Mochanova, M., Kodrik, D. and Krishnan, N. (2018). Disruption of Adipokinetic Hormone Mediated Energy Homeostasis Has Subtle Effects on Physiology, Behavior and Lipid Status During Aging in Drosophila. Front Physiol 9: 949. PubMed ID: 29627353

  • Robert Kofler Institute fur Populationsgenetik, Wien, Austria
    Schwarz, F., Wierzbicki, F., Senti, K. A. and Kofler, R. (2020). Tirant stealthily invaded natural Drosophila melanogaster populations during the last century. Mol Biol Evol. PubMed ID: 33247725

    Weilguny, L., Vlachos, C., Selvaraju, D. and Kofler, R. (2020). Reconstructing the Invasion Route of the P-Element in Drosophila melanogaster Using Extant Population Samples. Genome Biol Evol 12(11): 2139-2152. PubMed ID: 33210145

    >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

    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

  • Kyunghee Koh Biomedical Sciences, Thomas Jefferson University, Philadelphia
    Ozturk-Çolak, A., Inami, S., Buchler, J. R., McClanahan, P. D., Cruz, A., Fang-Yen, C. and Koh, K. (2020). Sleep Induction by Mechanosensory Stimulation in Drosophila. Cell Rep 33(9): 108462. PubMed ID: 33264620

    Duhart, J. M., Baccini, V., Zhang, Y., Machado, D. R. and Koh, K. (2020). Modulation of sleep-courtship balance by nutritional status in Drosophila. Elife 9. PubMed ID: 33084567

    Lamaze, A., Jepson, J. E. C., Akpoghiran, O. and Koh, K. (2020). Antagonistic Regulation of Circadian Output and Synaptic Development by JETLAG and the DYSCHRONIC-SLOWPOKE Complex. iScience 23(2): 100845. PubMed ID: 32058958

    Lamaze, A., Öztürk-Çolak, A., Fischer, R., Peschel, N., Koh, K. and Jepson, J.E. (2017). Regulation of sleep plasticity by a thermo-sensitive circuit in Drosophila. Sci Rep 7: 40304. PubMed ID: 28084307

    Afonso, D. J., Machado, D. R. and Koh, K. (2016). Control of sleep by a network of cell cycle genes. Fly (Austin): [Epub ahead of print]. PubMed ID: 26925838

    Afonso, D.J., Liu, D., Machado, D.R., Pan, H., Jepson, J.E., Rogulja, D. and Koh, K. (2015). TARANIS functions with Cyclin A and Cdk1 in a novel arousal center to control sleep in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26096977

    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

    Kim, Y. H., Kwon, D. H., Ahn, H. M., Koh, Y. H. and Lee, S. H. (2014). Induction of soluble AChE expression via alternative splicing by chemical stress in Drosophila melanogaster. Insect Biochem Mol Biol 48: 75-82. PubMed ID: 24637386

    Kim, W., Lee, D., Choi, J., Kim, A., Han, S., Park, K., Choi, J., Kim, J., Choi, Y., Lee, S. H. and Koh, Y. H. (2011). Pharmacogenetic regulation of acetylcholinesterase activity in Drosophila reveals the regulatory mechanisms of AChE inhibitors in synaptic plasticity. Neurochem Res 36: 879-893. PubMed ID: 21305389

  • Hiroshi Kohsaka Department of Complexity Science and Engineering The University of Tokyo, Todai
    Sun, X., Liu, Y., Liu, C., Mayumi, K., Ito, K., Nose, A. and Kohsaka, H. (2022). A neuromechanical model for Drosophila larval crawling based on physical measurements. BMC Biol 20(1): 130. PubMed ID: 35701821

    Giachello, C. N. G., Hunter, I., Pettini, T., Coulson, B., Knufer, A., Cachero, S., Winding, M., Arzan Zarin, A., Kohsaka, H., Fan, Y. N., Nose, A., Landgraf, M. and Baines, R. A. (2022). Electrophysiological validation of monosynaptic connectivity between premotor interneurons and the aCC motoneuron in the Drosophila larval CNS. J Neurosci 42(35): 6724-6738. PubMed ID: 35868863

    Matsuo, Y., Nose, A. and Kohsaka, H. (2021). Interspecies variation of larval locomotion kinematics in the genus Drosophila and its relation to habitat temperature. BMC Biol 19(1): 176. PubMed ID: 34470643

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

  • Tetsuya Kojima Department of Integrated Biosciences, Evolutionary Developmental Biology, University of Tokyo
    Tajiri, R., Fujiwara, H. and Kojima, T. (2021). A corset function of exoskeletal ECM promotes body elongation in Drosophila. Commun Biol 4(1): 88. PubMed ID: 33469125

    Tajiri, R., Ogawa, N., Fujiwara, H. and Kojima, T. (2017). Mechanical Control of Whole Body Shape by a Single Cuticular Protein Obstructor-E in Drosophila melanogaster. PLoS Genet 13(1): e1006548. PubMed ID: 28076349

    Natori, K., Tajiri, R., Furukawa, S. and Kojima, T. (2012). Progressive tarsal patterning in the Drosophila by temporally dynamic regulation of transcription factor genes. Dev Biol 361: 450-462. PubMed ID: 22079694

    Miyazono, K., Zhi, Y., Takamura, Y., Nagata, K., Saigo, K., Kojima, T. and Tanokura, M. (2010). Cooperative DNA-binding and sequence-recognition mechanism of aristaless and clawless. EMBO J 29: 1613-1623. PubMed ID: 20389279

  • https://www.winthrop.edu/cas/faculty/kohl-kathryn.aspx Biology Department, Winthrop University, Rock Hill, SC
    Mitchell, C., Becker, V., DeLoach, J., Nestore, E., Bolterstein, E. and Kohl, K. P. (2022). The Drosophila Mutagen-Sensitivity Gene mus109 Encodes DmDNA2. Genes (Basel) 13(2). PubMed ID: 35205357

    Hartmann, M., Sekelsky, J. and Hatkevich, T. (2019). Meiotic MCM Proteins Promote and Inhibit Crossovers During Meiotic Recombination. Genetics 212(2): 461-468. PubMed ID: 31028111

    Kohl, K. P. and Singh, N. D. (2018). Experimental evolution across different thermal regimes yields genetic divergence in recombination fraction but no divergence in temperature associated plastic recombination. Evolution 72(4): 989-999. PubMed ID: 29468654

    Hatkevich, T., Kohl, K. P., McMahan, S., Hartmann, M. A., Williams, A. M. and Sekelsky, J. (2017). Bloom Syndrome Helicase Promotes Meiotic Crossover Patterning and Homolog Disjunction. Curr Biol 27(1): 96-102. PubMed ID: 27989672

  • Alex Kolodkin Johns Hopkins University School of Medicine
    Xie, X., Tabuchi, M., Corver, A., Duan, G., Wu, M. N. and Kolodkin, A. L. (2019). Semaphorin 2b regulates sleep-circuit formation in the Drosophila central brain. Neuron. PubMed ID: 31564592

    Jeong, S., Yang, D. S., Hong, Y. G., Mitchell, S. P., Brown, M. P. and Kolodkin, A. L. (2017). Varicose and cheerio collaborate with pebble to mediate semaphorin-1a reverse signaling in Drosophila. Proc Natl Acad Sci U S A 114(39): E8254-e8263. PubMed ID: 28894005

    Wu, Z., Andreone, B. J. and Kolodkin, A. L. (2012). The coordinate regulation of sensory afferent CNS targeting and CNS longitudinal tract organization in Drosophila during neural development. J Peripher Nerv Syst 17 Suppl 3: 34-37. PubMed ID: 23279430

    Jeong, S., Juhaszova, K. and Kolodkin, A. L. (2012). The Control of Semaphorin-1a-Mediated Reverse Signaling by Opposing Pebble and RhoGAPp190 Functions in Drosophila. Neuron 76: 721-734. PubMed ID: 23177958

    Cho, J. Y., Chak, K., Andreone, B. J., Wooley, J. R. and Kolodkin, A. L. (2012). The extracellular matrix proteoglycan perlecan facilitates transmembrane semaphorin-mediated repulsive guidance. Genes Dev 26: 2222-2235. PubMed ID: 23028146

  • Alexey Kondrashov Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor
    Denisov, S. V., Bazykin, G. A., Sutormin, R., Favorov, A. V., Mironov, A. A., Gelfand, M. S. and Kondrashov, A. S. (2014). Weak negative and positive selection and the drift load at splice sites. Genome Biol Evol 6: 1437-1447. PubMed ID: 24966225

    Leushkin, E. V., Bazykin, G. A. and Kondrashov, A. S. (2013). Strong mutational bias towards deletions in the Drosophila melanogaster genome is compensated by selection. Genome Biol Evol. PubMed ID: 23395983

    Bazykin, G. A. and Kondrashov, A. S. (2012). Major role of positive selection in the evolution of conservative segments of Drosophila proteins. Proc Biol Sci 279: 3409-3417. PubMed ID: 22673359

    Leushkin, E. V., Bazykin, G. A. and Kondrashov, A. S. (2012). Insertions and deletions trigger adaptive walks in Drosophila proteins. Proc Biol Sci 279: 3075-3082. PubMed ID: 22456880

    Assis, R. and Kondrashov, A. S. (2012). A strong deletion bias in nonallelic gene conversion. PLoS Genet 8: e1002508. PubMed ID: 22359514

  • Mary Konsolaki Department of Genetics, Rutgers University, Piscataway, NJ
    Konsolaki, M. (2013). Fruitful research: drug target discovery for neurodegenerative diseases in Drosophila. Expert Opin Drug Discov. PubMed ID: 24151920

    Sanokawa-Akakura, R., Cao, W., Allan, K., Patel, K., Ganesh, A., Heiman, G., Burke, R., Kemp, F. W., Bogden, J. D., Camakaris, J., Birge, R. B. and Konsolaki, M. (2010). Control of Alzheimer's amyloid beta toxicity by the high molecular weight immunophilin FKBP52 and copper homeostasis in Drosophila. PLoS One 5: e8626. PubMed ID: 20084280

    van de Hoef, D. L., Hughes, J., Livne-Bar, I., Garza, D., Konsolaki, M. and Boulianne, G. L. (2009). Identifying genes that interact with Drosophila presenilin and amyloid precursor protein. Genesis 47: 246-260. PubMed ID: 19241393

  • Dimitrios Kontoyiannis Department of Infectious Diseases, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
    Zanette, R. A., Santurio, J. M., Loreto, E. S., Alves, S. H. and Kontoyiannis, D. P. (2013). Toll-deficient Drosophila is susceptible to Pythium insidiosum infection. Microbiol Immunol. PubMed ID: 23865688

    Zanette, R. A. and Kontoyiannis, D. P. (2013). Paradoxical effect to caspofungin in Candida species does not confer survival advantage in a Drosophila model of candidiasis. Virulence 4. PubMed ID: 23863608

    Diaz, L., Kontoyiannis, D. P., Panesso, D., Albert, N. D., Singh, K. V., Tran, T. T., Munita, J. M., Murray, B. E. and Arias, C. A. (2013). Dissecting the mechanisms of linezolid resistance in a Drosophila melanogaster infection model of Staphylococcus aureus. J Infect Dis 208: 83-91. PubMed ID: 23547139

    Ben-Ami, R., Watson, C. C., Lewis, R. E., Albert, N. D., Arias, C. A., Raad, II and Kontoyiannis, D. P. (2013). Drosophila melanogaster as a model to explore the effects of methicillin-resistant Staphylococcus aureus strain type on virulence and response to linezolid treatment. Microb Pathog 55: 16-20. PubMed ID: 23232438

  • Artyom Kopp Department of Evolution & Ecology, University of California Davis
    Gao, J. J., Barmina, O., Thompson, A., Kim, B. Y., Suvorov, A., Tanaka, K., Watabe, H., Toda, M. J., Chen, J. M., Katoh, T. K. and Kopp, A. (2022). Secondary reversion to sexual monomorphism associated with tissue-specific loss of doublesex expression. Evolution 76(9): 2089-2104. PubMed ID: 35841603

    Tanaka, K., Barmina, O., Thompson, A., Massey, J. H., Kim, B. Y., Suvorov, A. and Kopp, A. (2022). Evolution and development of male-specific leg brushes in Drosophilidae. Dev Genes Evol. PubMed ID: 35939093

    Luecke, D., Rice, G. and Kopp, A. (2022). Sex-specific evolution of a Drosophila sensory system via interacting cis- and trans-regulatory changes. Evol Dev 24(1-2): 37-60. PubMed ID: 35239254

    Luo, Y., Zhang, Y., Farine, J. P., Ferveur, J. F., Ramirez, S. and Kopp, A. (2019). Evolution of sexually dimorphic pheromone profiles coincides with increased number of male-specific chemosensory organs in Drosophila prolongata. Ecol Evol 9(23): 13608-13618. PubMed ID: 31871670

    Luecke, D. M. and Kopp, A. (2019). Sex-specific evolution of relative leg size in Drosophila prolongata results from changes in the intersegmental coordination of tissue growth. Evolution. PubMed ID: 31595502

    Rice, G. R., Barmina, O., Luecke, D., Hu, K., Arbeitman, M. and Kopp, A. (2019). Modular tissue-specific regulation of doublesex underpins sexually dimorphic development in Drosophila. Development 146(14). PubMed ID: 31285355

    Rice, G., Barmina, O., Hu, K. and Kopp, A. (2018). Evolving doublesex expression correlates with the origin and diversification of male sexual ornaments in the Drosophila immigrans species group. Evol Dev. PubMed ID: 29372584

    Yassin, A., Delaney, E. K., Reddiex, A. J., Seher, T. D., Bastide, H., Appleton, N. C., Lack, J. B., David, J. R., Chenoweth, S. F., Pool, J. E. and Kopp, A. (2016). The pdm3 locus is a hotspot for recurrent evolution of female-limited color dimorphism in Drosophila. Curr Biol. PubMed ID: 27546577

    Signor, S. A., Liu, Y., Rebeiz, M. and Kopp, A. (2016). Genetic convergence in the evolution of male-specific color patterns in Drosophila. Curr Biol. PubMed ID: 27546578

    Atallah, J., Teixeira, L., Salazar, R., Zaragoza, G. and Kopp, A. (2014). The making of a pest: the evolution of a fruit-penetrating ovipositor in Drosophila suzukii and related species. Proc Biol Sci 281: 20132840. PubMed ID: 24573846

    Atallah, J., Vurens, G., Mavong, S., Mutti, A., Hoang, D. and Kopp, A. (2013). Sex-specific repression of dachshund is required for Drosophila sex comb development. Dev Biol. PubMed ID: 24361261

  • Christopher Korey Department of Biology, College of Charleston, North Carolina
    Aby, E., Roth, A., Gumps, K., Sigmon, S., Jenkins, S. E., Kim, J. J., Kramer, N. J., Parfitt, K. D. and Korey, C. A. (2013). Mutations in palmitoyl-protein thioesterase 1 alter exocytosis and endocytosis at synapses in Drosophila larvae. Fly (Austin) 7. PubMed ID: 24091420

    Saja, S., Buff, H., Smith, A. C., Williams, T. S. and Korey, C. A. (2010). Identifying cellular pathways modulated by Drosophila palmitoyl-protein thioesterase 1 function. Neurobiol Dis 40: 135-145. PubMed ID: 20206262

  • Thomas Kornberg University of California San Francisco
    Barbosa, G. O. and Kornberg, T. B. (2022). Protocol for ex vivo time lapse imaging of Drosophila melanogaster cytonemes. STAR Protoc 3(1): 101138. PubMed ID: 35141564

    Hatori, R., Wood, B. M., Oliveira Barbosa, G. and Kornberg, T. B. (2021). Regulated delivery controls Drosophila Hedgehog, Wingless and Decapentaplegic signaling. Elife 10. PubMed ID: 34292155

    Wood, B. M., Baena, V., Huang, H., Jorgens, D. M., Terasaki, M. and Kornberg, T. B. (2021). Cytonemes with complex geometries and composition extend into invaginations of target cells. J Cell Biol 220(5). PubMed ID: 33734293

    Ali-Murthy, Z., Fetter, R. D., Wang, W., Yang, B., Royer, L. A. and Kornberg, T. B. (2021). Elimination of nurse cell nuclei that shuttle into oocytes during oogenesis. J Cell Biol 220(7). PubMed ID: 33950159

    Zhang, Z., Luo, S., Barbosa, G. O., Bai, M., Kornberg, T. B. and Ma, D. K. (2021). The conserved transmembrane protein TMEM-39 coordinates with COPII to promote collagen secretion and regulate ER stress response. PLoS Genet 17(2): e1009317. PubMed ID: 33524011

    Hatori, R. and Kornberg, T. B. (2020). Hedgehog produced by the Drosophila wing imaginal disc induces distinct responses in three target tissues. Development 147(22). PubMed ID: 33028613

    Zhang, Z., Bai, M., Barbosa, G. O., Chen, A., Wei, Y., Luo, S., Wang, X., Wang, B., Tsukui, T., Li, H., Sheppard, D., Kornberg, T. B. and Ma, D. K. (2020). Broadly conserved roles of TMEM131 family proteins in intracellular collagen assembly and secretory cargo trafficking. Sci Adv 6(7): eaay7667. PubMed ID: 32095531

    Fereres, S., Hatori, R., Hatori, M. and Kornberg, T. B. (2019). Cytoneme-mediated signaling essential for tumorigenesis. PLoS Genet 15(9): e1008415. PubMed ID: 31568500

    Huang, H., Liu, S. and Kornberg, T. B. (2019). Glutamate signaling at cytoneme synapses. Science 363(6430): 948-955. PubMed ID: 30819957

    Chen, W., Huang, H., Hatori, R. and Kornberg, T. B. (2017). Essential basal cytonemes take up Hedgehog in the Drosophila wing imaginal disc. Development. PubMed ID: 28743798

    Kashima, R., Redmond, P. L., Ghatpande, P., Roy, S., Kornberg, T. B., Hanke, T., Knapp, S., Lagna, G. and Hata, A. (2017). Hyperactive locomotion in a Drosophila model is a functional readout for the synaptic abnormalities underlying fragile X syndrome. Sci Signal 10(477). PubMed ID: 28465421

    Huang, H. and Kornberg, T. B. (2016). Cells must express components of the planar cell polarity system and extracellular matrix to support cytonemes. Elife 5 [Epub ahead of print]. PubMed ID: 27591355

  • Sally Kornbluth Pharmacology and Cancer Biology, Duke University School of Medicine, Durham
    Yang, C. S., Sinenko, S. A., Thomenius, M. J., Robeson, A. C., Freel, C. D., Horn, S. R. and Kornbluth, S. (2013). The deubiquitinating enzyme DUBAI stabilizes DIAP1 to suppress Drosophila apoptosis. Cell Death Differ. PubMed ID: 24362437

    Thomenius, M., Freel, C. D., Horn, S., Krieser, R., Abdelwahid, E., Cannon, R., Balasundaram, S., White, K. and Kornbluth, S. (2011). Mitochondrial fusion is regulated by Reaper to modulate Drosophila programmed cell death. Cell Death Differ 18: 1640-1650. PubMed ID: 21475305

    Yang, C. S., Thomenius, M. J., Gan, E. C., Tang, W., Freel, C. D., Merritt, T. J., Nutt, L. K. and Kornbluth, S. (2010). Metabolic regulation of Drosophila apoptosis through inhibitory phosphorylation of Dronc. EMBO J 29: 3196-3207. PubMed ID: 20700104

  • Iryna Kozeretska Department of General and Molecular Genetics, National Taras Shevchenko University of Kyiv, Kiev, Ukraine
    Bergman, C. M., Han, S., Nelson, M. G., Bondarenko, V. and Kozeretska, I. (2017). Genomic analysis of P elements in natural populations of Drosophila melanogaster. PeerJ 5: e3824. PubMed ID: 28929030

    Bilousov, O. O., Katanaev, V. L., Demydov, S. V. and Kozeretska, I. A. (2013). The downregulation of the miniature gene does not replicate miniature loss-of-function phenotypes in Drosophila melanogaster wing to the full extent. Tsitol Genet 47: 77-81. PubMed ID: 23745366

    Vaiserman, A. M., Zabuga, O. G., Kolyada, A. K., Pisaruk, A. V. and Kozeretska, I. A. (2013). Reciprocal cross differences in Drosophila melanogaster longevity: an evidence for non-genomic effects in heterosis phenomenon? Biogerontology 14: 153-163. PubMed ID: 23529279

    Rozhok, A. I., Ievdokymenko, K. S. and Kozeretska, I. A. (2012). On the persistence of P element in cultured lineages of Drosophila melanogaster. Tsitol Genet 46: 55-58. PubMed ID: 23074963

  • Michael Krahn Institute for Molecular and Cellular Anatomy, University of Regensburg
    Gass, M. M., Borkowsky, S., Lotz, M. L., Siwek, R., Schroter, R., Nedvetsky, P., Luschnig, S., Rohlmann, A., Missler, M. and Krahn, M. P. (2022). PI(4,5)P2 controls slit diaphragm formation and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci 79(5): 248. PubMed ID: 35437696

    Heiden, S., Siwek, R., Lotz, M. L., Borkowsky, S., Schröter, R., Nedvetsky, P., Rohlmann, A., Missler, M. and Krahn, M. P. (2021). Apical-basal polarity regulators are essential for slit diaphragm assembly and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci. PubMed ID: 33651172

    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

    Kullmann, L. and Krahn, M. P. (2018). Redundant regulation of localization and protein stability of DmPar3. Cell Mol Life Sci. PubMed ID: 29523893

    Hochapfel, F., Denk, L., Mendl, G., Schulze, U., Maassen, C., Zaytseva, Y., Pavenstadt, H., Weide, T., Rachel, R., Witzgall, R. and Krahn, M. P. (2017). Distinct functions of Crumbs regulating slit diaphragms and endocytosis in Drosophila nephrocytes. Cell Mol Life Sci. PubMed ID: 28717874

    Dogliotti, G., Kullmann, L., Dhumale, P., Thiele, C., Panichkina, O., Mendl, G., Houben, R., Haferkamp, S., Puschel, A. W. and Krahn, M. P. (2017). Membrane-binding and activation of LKB1 by phosphatidic acid is essential for development and tumour suppression. Nat Commun 8: 15747. PubMed ID: 28649994

    Koch, L., Feicht, S., Sun, R., Sen, A. and Krahn, M. P. (2016). Domain-specific functions of Stardust in Drosophila embryonic development. R Soc Open Sci 3(11): 160776. PubMed ID: 28018665

    Loedige, I., Jakob, L., Treiber, T., Ray, D., Stotz, M., Treiber, N., Hennig, J., Cook, K. B., Morris, Q., Hughes, T. R., Engelmann, J. C., Krahn, M. P. and Meister, G. (2015). The Crystal Structure of the NHL Domain in Complex with RNA Reveals the Molecular Basis of Drosophila Brain-Tumor-Mediated Gene Regulation. Cell Rep 13(6): 1206-1220. PubMed ID: 26527002

    Sen, A., Sun, R. and Krahn, M. P. (2015). Localization and Function of Pals1-associated Tight Junction Protein in Drosophila Is Regulated by Two Distinct Apical Complexes. J Biol Chem 290(21): 13224-13233. PubMed ID: 25847234

    Shahab, J., Tiwari, M. D., Honemann-Capito, M., Krahn, M. P. and Wodarz, A. (2015). Bazooka/PAR3 is dispensable for polarity in Drosophila follicular epithelial cells. Biol Open 4(4): 528-541. PubMed ID: 25770183

  • Achim Kramer Universitãtsmedizin Berlin
    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

    Klemz, R., Reischl, S., Wallach, T., Witte, N., Jurchott, K., Klemz, S., Lang, V., Lorenzen, S., Knauer, M., Heidenreich, S., Xu, M., Ripperger, J. A., Schupp, M., Stanewsky, R. and Kramer, A. (2017). Reciprocal regulation of carbon monoxide metabolism and the circadian clock. Nat Struct Mol Biol 24(1): 15-22. PubMed ID: 27892932

    Lee, E., Jeong, E. H., Jeong, H. J., Yildirim, E., Vanselow, J. T., Ng, F., Liu, Y., Mahesh, G., Kramer, A., Hardin, P. E., Edery, I. and Kim, E. Y. (2014). Phosphorylation of a central clock transcription factor is required for thermal but not photic entrainment. PLoS Genet 10(8): e1004545. PubMed ID: 25121504

  • Helmut Krämer University of Texas Southwestern Medical Center, Dallas
    Casey, A. K., Gray, H. F., Chimalapati, S., Hernandez, G., Moehlman, A. T., Stewart, N., Fields, H. A., Gulen, B., Servage, K. A., Stefanius, K., Blevins, A., Evers, B. M., Kramer, H. and Orth, K. (2022). Fic-mediated AMPylation tempers the unfolded protein response during physiological stress. Proc Natl Acad Sci U S A 119(32): e2208317119. PubMed ID: 35914137

    Nandi, N., Zaidi, Z., Tracy, C. and Kramer, H. (2022). A phospho-switch at Acinus-Serine(437) controls autophagic responses to Cadmium exposure and neurodegenerative stress. Elife 11. PubMed ID: 35037620

    Kurtz, P., Jones, A. E., Tiwari, B., Link, N., Wylie, A., Tracy, C., Kramer, H. and Abrams, J. M. (2019). Drosophila p53 directs non-apoptotic programs in postmitotic tissue. Mol Biol Cell: mbcE18120791. PubMed ID: 30892991

    Moehlman, A. T., Casey, A. K., Servage, K., Orth, K. and Kramer, H. (2018). Adaptation to constant light requires Fic-mediated AMPylation of BiP to protect against reversible photoreceptor degeneration. Elife 7. PubMed ID: 30015618

    Nandi, N. and Kramer, H. (2018). Cdk5-mediated Acn/Acinus phosphorylation regulates basal autophagy independently of metabolic stress. Autophagy: 1-2. PubMed ID: 29782227

    Casey, A. K., Moehlman, A. T., Zhang, J., Servage, K. A., Kramer, H. and Orth, K. (2017). Fic-mediated deAMPylation is not dependent on homo-dimerization and rescues toxic AMPylation in flies. J Biol Chem [Epub ahead of print]. PubMed ID: 29089387

    Akbar, M. A., Mandraju, R., Tracy, C., Hu, W., Pasare, C. and Kramer, H. (2016). ARC syndrome-linked Vps33B protein is required for inflammatory endosomal maturation and signal termination. Immunity 45: 267-279. PubMed ID: 27496733

    Ham, H., Woolery, A. R., Tracy, C., Stenesen, D., Kramer, H. and Orth, K. (2014). Unfolded protein response-regulated dFic reversibly AMPylates BiP during endoplasmic reticulum homeostasis. J Biol Chem. PubMed ID: 25395623

    Nandi, N., Tyra, L. K., Stenesen, D. and Kramer, H. (2014). Acinus integrates AKT1 and subapoptotic caspase activities to regulate basal autophagy. J Cell Biol 207: 253-268. PubMed ID: 25332163

    Takats, S., Pircs, K., Nagy, P., Varga, A., Karpati, M., Hegedus, K., Kramer, H., Kovacs, A. L., Sass, M. and Juhasz, G. (2014). Interaction of the HOPS complex with Syntaxin 17 mediates autophagosome clearance in Drosophila. Mol Biol Cell 25: 1338-1354. PubMed ID: 24554766

    Kowalewski-Nimmerfall, E., Schahs, P., Maresch, D., Rendic, D., Kramer, H. and Mach, L. (2014). Drosophila melanogaster cellular repressor of E1A-stimulated genes is a lysosomal protein essential for fly development. Biochim Biophys Acta 1843: 2900-2912. PubMed ID: 25173815

  • Sunita Gupta Kramer Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, NJ
    King, T. R., Kramer, J., Cheng, Y. S., Swope, D. and Kramer, S. G. (2021). Enabled/VASP is required to mediate proper sealing of opposing cardioblasts during Drosophila dorsal vessel formation. Dev Dyn. PubMed ID: 33587326

    Swope, D., Kramer, J., King, T. R., Cheng, Y. S. and Kramer, S. G. (2014). Cdc42 is required in a genetically distinct subset of cardiac cells during drosophila dorsal vessel closure. Dev Biol. PubMed ID: 24949939

    Macabenta, F. D., Jensen, A. G., Cheng, Y. S., Kramer, J. J. and Kramer, S. G. (2013). Frazzled/DCC facilitates cardiac cell outgrowth and attachment during Drosophila dorsal vessel formation. Dev Biol 380: 233-242. PubMed ID: 23685255

    Soplop, N. H., Cheng, Y. S. and Kramer, S. G. (2012). Roundabout is required in the visceral mesoderm for proper microvillus length in the hindgut epithelium. Dev Dyn 241: 759-769. PubMed ID: 22334475

  • David Krantz Neuroscience Program, UCLA
    Knapp, E. M., Kaiser, A., Arnold, R. C., Sampson, M. M., Ruppert, M., Xu, L., Anderson, M. I., Bonanno, S. L., Scholz, H., Donlea, J. M. and Krantz, D. E. (2022). Mutation of the Drosophila melanogaster serotonin transporter dSERT impacts sleep, courtship, and feeding behaviors. PLoS Genet 18(11): e1010289. PubMed ID: 36409783

    Sampson, M. M., Myers Gschweng, K. M., Hardcastle, B. J., Bonanno, S. L., Sizemore, T. R., Arnold, R. C., Gao, F., Dacks, A. M., Frye, M. A. and Krantz, D. E. (2020). Serotonergic modulation of visual neurons in Drosophila melanogaster. PLoS Genet 16(8): e1009003. PubMed ID: 32866139

    Majdi, S., Berglund, E. C., Dunevall, J., Oleinick, A. I., Amatore, C., Krantz, D. E. and Ewing, A. G. (2015). Electrochemical Measurements of Optogenetically Stimulated Quantal Amine Release from Single Nerve Cell Varicosities in Drosophila Larvae. Angew Chem Int Ed Engl. PubMed ID: 26387683

    Martin, C. A., Barajas, A., Lawless, G., Lawal, H. O., Assani, K., Lumintang, Y. P., Nunez, V. and Krantz, D. E. (2014). Synergistic effects on dopamine cell death in a Drosophila model of chronic toxin exposure. Neurotoxicology. PubMed ID: 25160001

    Grygoruk, A., Chen, A., Martin, C. A., Lawal, H. O., Fei, H., Gutierrez, G., Biedermann, T., Najibi, R., Hadi, R., Chouhan, A. K., Murphy, N. P., Schweizer, F. E., Macleod, G. T., Maidment, N. T. and Krantz, D. E. (2014). The redistribution of Drosophila vesicular monoamine transporter mutants from synaptic vesicles to large dense-core vesicles impairs amine-dependent behaviors. J Neurosci 34: 6924-6937. PubMed ID: 24828646

    Martin, C. A. and Krantz, D. E. (2014). Drosophila melanogaster as a genetic model system to study neurotransmitter transporters. Neurochem Int. PubMed ID: 24704795

    Lawal, H. O., Terrell, A., Lam, H. A., Djapri, C., Jang, J., Hadi, R., Roberts, L., Shahi, V., Chou, M. T., Biedermann, T., Huang, B., Lawless, G. M., Maidment, N. T. and Krantz, D. E. (2014). Drosophila modifier screens to identify novel neuropsychiatric drugs including aminergic agents for the possible treatment of Parkinson's disease and depression. Mol Psychiatry 19: 235-242. PubMed ID: 23229049

    Wu, T. H., Lu, Y. N., Chuang, C. L., Wu, C. L., Chiang, A. S., Krantz, D. E. and Chang, H. Y. (2013). Loss of vesicular dopamine release precedes tauopathy in degenerative dopaminergic neurons in a Drosophila model expressing human tau. Acta Neuropathol 125: 711-725. PubMed ID: 23494099

    Chen, A., Ng, F., Lebestky, T., Grygoruk, A., Djapri, C., Lawal, H. O., Zaveri, H. A., Mehanzel, F., Najibi, R., Seidman, G., Murphy, N. P., Kelly, R. L., Ackerson, L. C., Maidment, N. T., Jackson, F. R. and Krantz, D. E. (2013). Dispensable, redundant, complementary, and cooperative roles of dopamine, octopamine, and serotonin in Drosophila melanogaster. Genetics 193: 159-176. PubMed ID: 23086220

  • Alexey N Krasnov Institute of Gene Biology, Russian Academy of Sciences, Moscow
    Vorobyeva, N. E., Erokhin, M., Chetverina, D., Krasnov, A. N. and Mazina, M. Y. (2021). Su(Hw) primes 66D and 7F Drosophila chorion genes loci for amplification through chromatin decondensation. Sci Rep 11(1): 16963. PubMed ID: 34417521

    Nikolenko, J. V., Kurshakova, M. M., Krasnov, A. N. and Georgieva, S. G. (2021). MLE Helicase Is a New Participant in the Transcription Regulation of the ftz-f1 Gene Encoding Nuclear Receptor in Higher Eukaryotes. Dokl Biochem Biophys 496(1): 1-4. PubMed ID: 33689064

    Fursova, N. A., Mazina, M. Y., Nikolenko, J. V., Vorobyova, N. E. and Krasnov, A. N. (2020). Drosophila Zinc Finger Protein CG9890 Is Colocalized with Chromatin Modifying and Remodeling Complexes on Gene Promoters and Involved in Transcription Regulation. Acta Naturae 12(4): 114-119. PubMed ID: 33456983

    Nikolenko, J. V., Kurshakova, M. M. and Krasnov, A. N. (2019). Multifunctional ENY2 Protein Interacts with RNA Helicase MLE. Dokl Biochem Biophys 489(1): 407-410. PubMed ID: 32130612

  • Mark Krasnow Department of Biochemistry, Stanford
    Peterson, S.J. and Krasnow, M.A. (2015). Subcellular trafficking of FGF controls tracheal invasion of Drosophila flight muscle. Cell 160: 313-323. PubMed ID: 25557078

    Chen, F. and Krasnow, M. A. (2014). Progenitor outgrowth from the niche in Drosophila trachea is guided by FGF from decaying branches. Science 343: 186-189. PubMed ID: 24408434

    Tan, F. E., Vladar, E. K., Ma, L., Fuentealba, L. C., Hoh, R., Espinoza, F. H., Axelrod, J. D., Alvarez-Buylla, A., Stearns, T., Kintner, C. and Krasnow, M. A. (2013). Myb promotes centriole amplification and later steps of the multiciliogenesis program. Development 140: 4277-4286. PubMed ID: 24048590

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

  • Ed Kravitz Department of Neurobiology, Harvard Medical School, Boston
    Sengupta, S., Chan, Y. B., Palavicino-Maggio, C. B. and Kravitz, E. A. (2022). GABA transmission from mAL interneurons regulates aggression in Drosophila males. Proc Natl Acad Sci U S A 119(5). PubMed ID: 35082150

    Monyak, R. E., Golbari, N. M., Chan, Y. B., Pranevicius, A., Tang, G., Fernandez, M. P. and Kravitz, E. A. (2021). Masculinized Drosophila females adapt their fighting strategies to their opponent. J Exp Biol. PubMed ID: 33568440

    Palavicino-Maggio, C. B., Chan, Y. B., McKellar, C. and Kravitz, E. A. (2019). A small number of cholinergic neurons mediate hyperaggression in female Drosophila. Proc Natl Acad Sci U S A 116(34): 17029-17038. PubMed ID: 31391301

    Alekseyenko, O. V., Chan, Y. B., Okaty, B. W., Chang, Y., Dymecki, S. M. and Kravitz, E. A. (2019). Serotonergic modulation of aggression in Drosophila involves GABAergic and cholinergic opposing pathways. Curr Biol. PubMed ID: 31231050

    Chowdhury, B., Chan, Y.B. and Kravitz, E.A. (2017). Putative transmembrane transporter modulates higher-level aggression in Drosophila. Proc Natl Acad Sci U S A 114: 2373-2378. PubMed ID: 28193893

    Trannoy, S. and Kravitz, E. A. (2016). Strategy changes in subsequent fights as consequences of winning and losing in fruit fly fights. Fly (Austin): [Epub ahead of print]. PubMed ID: 27834611

    Trannoy, S., Penn, J., Lucey, K., Popovic, D. and Kravitz, E.A. (2016). Short and long-lasting behavioral consequences of agonistic encounters between male Drosophila melanogaster. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27071097

    Alekseyenko, O. V. and Kravitz, E. A. (2014). Serotonin and the search for the anatomical substrate of aggression. Fly (Austin) 8: 200-205. PubMed ID: 25923771

    Trannoy, S., Chowdhury, B. and Kravitz, E. A. (2015). A new approach that eliminates handling for studying aggression and the "loser" effect in Drosophila melanogaster. J Vis Exp. PubMed ID: 26780386

    Chan, Y. B., Alekseyenko, O. V. and Kravitz, E. A. (2015). Optogenetic Control of Gene Expression in Drosophila. PLoS One 10: e0138181. PubMed ID: 26383635

    Trannoy, S. and Kravitz, E. A. (2015). Learning and memory during aggression in Drosophila: handling affects aggression and the formation of a "loser" effect. J Nat Sci 1: e56. PubMed ID: 25789340

    Trannoy, S., Chowdhury, B. and Kravitz, E. A. (2015). Handling alters aggression and "loser" effect formation in Drosophila melanogaster. Learn Mem 22: 64-68. PubMed ID: 25593291

  • Henry Krause Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto
    Witt, E., Shao, Z., Hu, C., Krause, H. M. and Zhao, L. (2021). Single-cell RNA-sequencing reveals pre-meiotic X-chromosome dosage compensation in Drosophila testis. PLoS Genet 17(8): e1009728. PubMed ID: 34403408

    Umegawachi, T., Yoshida, H., Koshida, H., Yamada, M., Ohkawa, Y., Sato, T., Suyama, M., Krause, H. M. and Yamaguchi, M. (2017). Control of tissue size and development by a regulatory element in the yorkie 3'UTR. Am J Cancer Res 7(3): 673-687. PubMed ID: 28401020

    Lee, D. M., Wilk, R., Hu, J., Krause, H. M. and Harris, T. J. (2015). Germ Cell Segregation from the Drosophila Soma Is Controlled by an Inhibitory Threshold Set by the Arf-GEF Steppke. Genetics. PubMed ID: 25971667

    Wilk, R., Hu, J., Krause, H. M. (2013) Spatial Profiling of Nuclear Receptor Transcription Patterns Over the Course of Drosophila Development. G3 (Bethesda). PubMed ID: 23665880

    Knox, J., Moyer, K., Yacoub, N., Soldaat, C., Komosa, M., Vassilieva, K., Wilk, R., Hu, J., Vazquez Paz Lde, L., Syed, Q., Krause, H. M., Georgescu, M. and Jacobs, J. R. (2011). Syndecan contributes to heart cell specification and lumen formation during Drosophila cardiogenesis. Dev Biol 356: 279-290. PubMed ID: 21565181

    Zou, S., Chang, J., LaFever, L., Tang, W., Johnson, E. L., Hu, J., Wilk, R., Krause, H. M., Drummond-Barbosa, D. and Irusta, P. M. (2011). Identification of dAven, a Drosophila melanogaster ortholog of the cell cycle regulator Aven. Cell Cycle 10: 989-998. PubMed ID: 21368576

  • Rachel Kraut Division of Molecular Genetics & Cell Biology School of Biological Sciences, Nanyang Technological University
    Hebbar, S., Sahoo, I., Matysik, A., Argudo Garcia, I., Osborne, K. A., Papan, C., Torta, F., Narayanaswamy, P., Fun, X. H., Wenk, M. R., Shevchenko, A., Schwudke, D. and Kraut, R. (2015). Ceramides and stress signalling intersect with autophagic defects in neurodegenerative Drosophila blue cheese (bchs) mutants. Sci Rep 5: 15926. PubMed ID: 26639035

    Kraut, R. (2011). Roles of sphingolipids in Drosophila development and disease. J Neurochem 116: 764-778. PubMed ID: 21214556

    Hortsch, R., Lee, E., Erathodiyil, N., Hebbar, S., Steinert, S., Lee, J. Y., Chua, D. S. and Kraut, R. (2010). Glycolipid trafficking in Drosophila undergoes pathway switching in response to aberrant cholesterol levels. Mol Biol Cell 21: 778-790. PubMed ID: 20053687

  • Edward Kravitz Harvard Medical School
    Alekseyenko, O. V., Chan, Y. B., Li, R., Kravitz, E. A. (2013) Single dopaminergic neurons that modulate aggression in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 23530210

    Rezaval, C., Pavlou, H. J., Dornan, A. J., Chan, Y. B., Kravitz, E. A. and Goodwin, S. F. (2012). Neural circuitry underlying Drosophila female postmating behavioral responses. Curr Biol 22: 1155-1165. PubMed ID: 22658598

    Fernandez, M. P., Chan, Y. B., Yew, J. Y., Billeter, J. C., Dreisewerd, K., Levine, J. D. and Kravitz, E. A. (2010). Pheromonal and behavioral cues trigger male-to-female aggression in Drosophila. PLoS Biol 8: e1000541. PubMed ID: 21124886

    Certel, S. J., Leung, A., Lin, C. Y., Perez, P., Chiang, A. S. and Kravitz, E. A. (2010). Octopamine neuromodulatory effects on a social behavior decision-making network in Drosophila males. PLoS One 5: e13248. PubMed ID: 20967276

  • Martin Kreitman Department of Ecology and Evolution, University of Chicago
    Jha, A. R., Zhou, D., Brown, C. D., Kreitman, M., Haddad, G. G. and White, K. P. (2015). Shared Genetic Signals of Hypoxia Adaptation in Drosophila and in High-Altitude Human Populations. Mol Biol Evol [Epub ahead of print]. PubMed ID: 26576852

    Jiang, P., Ludwig, M. Z., Kreitman, M. and Reinitz, J. (2015). Natural variation of the expression pattern of the segmentation gene even-skipped in melanogaster. Dev Biol [Epub ahead of print]. PubMed ID: 26129990

    Jha, A. R., Miles, C. M., Lippert, N. R., Brown, C. D., White, K. P. and Kreitman, M. (2015). Whole genome resequencing of experimental populations reveals polygenic basis of egg size variation in Drosophila melanogaster. Mol Biol Evol. PubMed ID: 26044351

    Palsson, A., Wesolowska, N., Reynisdottir, S., Ludwig, M. Z. and Kreitman, M. (2014). Naturally Occurring Deletions of Hunchback Binding Sites in the Even-Skipped Stripe 3+7 Enhancer. PLoS One 9: e91924. PubMed ID: 24786295

    Martinez, C., Rest, J. S., Kim, A. R., Ludwig, M., Kreitman, M., White, K. and Reinitz, J. (2014). Ancestral resurrection of the Drosophila S2E enhancer reveals accessible evolutionary paths through compensatory change. Mol Biol Evol. PubMed ID: 24408913

    He, B. Z., Ludwig, M. Z., Dickerson, D. A., Barse, L., Arun, B., Vilhjalmsson, B. J., Park, S. Y., Tamarina, N. A., Selleck, S. B., Wittkopp, P. J., Bell, G. I. and Kreitman, M. (2013). Effect of Genetic Variation in a Drosophila Model of Diabetes-Associated Misfolded Human Proinsulin. Genetics. PubMed ID: 24281155

    Park, S. Y., Ludwig, M. Z., Tamarina, N. A., He, B. Z., Carl, S. H., Dickerson, D. A., Barse, L., Arun, B., Williams, C. L., Miles, C. M., Philipson, L. H., Steiner, D. F., Bell, G. I. and Kreitman, M. (2013). Genetic Complexity in a Drosophila Model of Diabetes-Associated Misfolded Human Proinsulin. Genetics. PubMed ID: 24281154

    Cheung, D., Miles, C., Kreitman, M. and Ma, J. (2013). Adaptation of the length scale and amplitude of the Bicoid gradient profile to achieve robust patterning in abnormally large Drosophila melanogaster embryos. Development. PubMed ID: 24284208

    Manu, M., Ludwig, M. Z., Kreitman, M. (2013) Sex-specific Pattern Formation During Early Drosophila Development. Genetics. PubMed ID: 23410834

  • Doris Kretszchmar Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland
    Cassar, M., Law, A. D., Chow, E. S., Giebultowicz, J. M. and Kretzschmar, D. (2020). Disease-Associated Mutant Tau Prevents Circadian Changes in the Cytoskeleton of Central Pacemaker Neurons. Front Neurosci 14: 232. PubMed ID: 32292325

    Sunderhaus, E. R., Law, A. D. and Kretzschmar, D. (2019). ER responses play a key role in Swiss-Cheese/Neuropathy Target Esterase-associated neurodegeneration. Neurobiol Dis: 104520. PubMed ID: 31233884

    Cassar, M., Sunderhaus, E., Wentzell, J. S., Kuntz, S., Strauss, R. and Kretzschmar, D. (2018). The PKA-C3 catalytic subunit is required in two pairs of interneurons for successful mating of Drosophila. Sci Rep 8(1): 2458. PubMed ID: 29410515

    Sunderhaus, E. R. and Kretzschmar, D. (2016). Mass Histology to Quantify Neurodegeneration in Drosophila. J Vis Exp(118) [Epub ahead of print] PubMed ID: 28060320

    Ramaker, J. M., Cargill, R. S., Swanson, T. L., Quirindongo, H., Cassar, M., Kretzschmar, D. and Copenhaver, P. F. (2016). Amyloid precursor proteins are dynamically trafficked and processed during neuronal development. Front Mol Neurosci 9: 130. PubMed ID: 27932950

    Cassar, M. and Kretzschmar, D. (2016). Analysis of Amyloid precursor protein function in Drosophila melanogaster. Front Mol Neurosci 9: 61. PubMed ID: 27507933

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

    Cook, M., Bolkan, B. J. and Kretzschmar, D. (2014). Increased actin polymerization and stabilization interferes with neuronal function and survival in the AMPKgamma mutant Loechrig. PLoS One 9: e89847. PubMed ID: 24587072

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

    Bolkan, B. J. and Kretzschmar, D. (2014). Loss of Tau results in defects in photoreceptor development and progressive neuronal degeneration in Drosophila. Dev Neurobiol. PubMed ID: 24909306

    Wentzell, J. S., Cassar, M. and Kretzschmar, D. (2014). Organophosphate-Induced Changes in the PKA Regulatory Function of Swiss Cheese/NTE Lead to Behavioral Deficits and Neurodegeneration. PLoS One 9: e87526. PubMed ID: 24558370

    Bolkan, B. J., Triphan, T. and Kretzschmar, D. (2012). beta-secretase cleavage of the fly amyloid precursor protein is required for glial survival. J Neurosci 32: 16181-16192. PubMed ID: 23152602

  • Torsten Kristensen Section of Biology and Environmental Science, Aalborg, Denmark
    Jorgensen, D. B., Orsted, M. and Kristensen, T. N. (2022). Sustained positive consequences of genetic rescue of fitness and behavioural traits in inbred populations of Drosophila melanogaster. J Evol Biol 35(6): 868-878. PubMed ID: 35532930

    Davies, L. R., Loeschcke, V., Schou, M. F., Schramm, A. and Kristensen, T. N. (2021). The importance of environmental microbes for Drosophila melanogaster during seasonal macronutrient variability. Sci Rep 11(1): 18850. PubMed ID: 34552121

    Rohde, P. D., Jensen, I. R., Sarup, P. M., Orsted, M., Demontis, D., Sorensen, P. and Kristensen, T. N. (2019). Genetic signatures of drug response variability in Drosophila melanogaster. Genetics. PubMed ID: 31455722

    Orsted, M., Hoffmann, A. A., Sverrisdottir, E., Nielsen, K. L. and Kristensen, T. N. (2019). Genomic variation predicts adaptive evolutionary responses better than population bottleneck history. PLoS Genet 15(6): e1008205. PubMed ID: 31188830

    Orsted, M., Hoffmann, A. A., Rohde, P. D., Sorensen, P. and Kristensen, T. N. (2018). Strong impact of thermal environment on the quantitative genetic basis of a key stress tolerance trait. Heredity (Edinb). PubMed ID: 30050062

    Schou, M. F., Loeschcke, V., Bechsgaard, J., Schlotterer, C. and Kristensen, T. N. (2017). Unexpected high genetic diversity in small populations suggests maintenance by associative overdominance. Mol Ecol. PubMed ID: 28746770

    Orsted, M., Schou, M. F. and Kristensen, T. N. (2017). Biotic and abiotic factors investigated in two Drosophila species - evidence of both negative and positive effects of interactions on performance. Sci Rep 7: 40132. PubMed ID: 28059144

    Schou, M. F., Loeschcke, V. and Kristensen, T. N. (2015). Inbreeding depression across a nutritional stress continuum. Heredity (Edinb) 115: 56-62. PubMed ID: 26059969

    Schou, M.F., Loeschcke, V. and Kristensen, T.N. (2015). Strong costs and benefits of winter acclimatization in Drosophila melanogaster. PLoS One 10: e0130307. PubMed ID: 26075607

    Jensen, P., Overgaard, J., Loeschcke, V., Schou, M. F., Malte, H. and Kristensen, T. N. (2014). Inbreeding effects on standard metabolic rate investigated at cold, benign and hot temperatures in Drosophila melanogaster. J Insect Physiol. PubMed ID: 24456661

  • Dan Kuebler Franciscan University of Steubenville, Steubenville, Ohio
    Stone, B., Burke, B., Pathakamuri, J., Coleman, J. and Kuebler, D. (2014). A Low-cost Method for Analyzing Seizure-like Activity and Movement in Drosophila. J Vis Exp. PubMed ID: 24637378

    Stone, B., Evans, L., Coleman, J. and Kuebler, D. (2013). Genetic and pharmacological manipulations that alter metabolism suppress seizure-like activity in Drosophila. Brain Res 1496: 94-103. PubMed ID: 23247062

    Whelan, J., Burke, B., Rice, A., Tong, M. and Kuebler, D. (2010). Sensitivity to seizure-like activity in Drosophila following acute hypoxia and hypercapnia. Brain Res 1316: 120-128. PubMed ID: 20034480

  • Gustavo Kuhn Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
    Silva, B., Heringer, P., Dias, G. B., Svartman, M. and Kuhn, G. C. S. (2019). De novo identification of satellite DNAs in the sequenced genomes of Drosophila virilis and D. americana using the RepeatExplorer and TAREAN pipelines. PLoS One 14(12): e0223466. PubMed ID: 31856171

    Teixeira, J. R., Dias, G. B., Svartman, M., Ruiz, A. and Kuhn, G. C. S. (2018). Concurrent Duplication of Drosophila Cid and Cenp-C Genes Resulted in Accelerated Evolution and Male Germline-Biased Expression of the New Copies. J Mol Evol. PubMed ID: 29934734

    de Lima, L. G., Svartman, M. and Kuhn, G. C. S. (2017). Dissecting the satellite DNA landscape in three cactophilic Drosophila sequenced genomes. G3 (Bethesda). PubMed ID: 28659292

    Dias, G. B., Heringer, P., Svartman, M. and Kuhn, G. C. (2015). Helitrons shaping the genomic architecture of Drosophila: enrichment of DINE-TR1 in alpha- and beta-heterochromatin, satellite DNA emergence, and piRNA expression. Chromosome Res. PubMed ID: 26408292

    Dias, G. B., Svartman, M., Delprat, A., Ruiz, A. and Kuhn, G. C. (2014). Tetris is a foldback transposon that provided the building blocks for an emerging satellite DNA of Drosophila virilis. Genome Biol Evol. PubMed ID: 24858539

    Kuhn, G. C., Kuttler, H., Moreira-Filho, O. and Heslop-Harrison, J. S. (2012). The 1.688 repetitive DNA of Drosophila: concerted evolution at different genomic scales and association with genes. Mol Biol Evol 29: 7-11. PubMed ID: 21712468

    Kuhn, G. C. and Heslop-Harrison, J. S. (2011). Characterization and genomic organization of PERI, a repetitive DNA in the Drosophila buzzatii cluster related to DINE-1 transposable elements and highly abundant in the sex chromosomes. Cytogenet Genome Res 132: 79-88. PubMed ID: 20938165

  • Rob Kulathinal Molecular and Fly Laboratory, Temple University, Philadelphia
    Stanley, C.E. Jr. and Kulathinal, R.J. (2016). Genomic signatures of domestication on neurogenetic genes in Drosophila melanogaster. BMC Evol Biol 16: 6. PubMed ID: 26728183Hansen, M. E., Kulathinal, R. J. (2013) Sex-biased networks and nodes of sexually antagonistic conflict in Drosophila. Int J Evol Biol 2013: 545392. PubMed ID: 23431497

    Renn, S. C., Machado, H. E., Jones, A., Soneji, K., Kulathinal, R. J. and Hofmann, H. A. (2010). Using comparative genomic hybridization to survey genomic sequence divergence across species: a proof-of-concept from Drosophila. BMC Genomics 11: 271. PubMed ID: 20429934

    Kulathinal, R. J., Stevison, L. S. and Noor, M. A. (2009). The genomics of speciation in Drosophila: diversity, divergence, and introgression estimated using low-coverage genome sequencing. PLoS Genet 5: e1000550. PubMed ID: 19578407

  • Ronald Kühnlein Max Planck Institute for Biophysical Chemistry, Dept. of Molecular Developmental Biology, Gõttingen
    Pauls, D., Selcho, M., Räderscheidt, J., Amatobi, K. M., Fekete, A., Krischke, M., Hermann-Luibl, C., Ozbek-Unal, A. G., Ehmann, N., Itskov, P. M., Kittel, R. J., Helfrich-Forster, C., Kuhnlein, R. P., Mueller, M. J. and Wegener, C. (2021). Endocrine signals fine-tune daily activity patterns in Drosophila. Curr Biol. PubMed ID: 34329588

    Hofbauer, H. F., Heier, C., Sen Saji, A. K. and Kuhnlein, R. P. (2020). Lipidome remodeling in aging normal and genetically obese Drosophila males. Insect Biochem Mol Biol: 103498. PubMed ID: 33221388

    Hehlert, P., Hofferek, V., Heier, C., Eichmann, T. O., Riedel, D., Rosenberg, J., Takacs, A., Nagy, H. M., Oberer, M., Zimmermann, R. and Kuhnlein, R. P. (2019). The alpha/beta-hydrolase domain-containing 4 and 5 (ABHD4/5)-related phospholipase Pummelig controls energy storage in Drosophila. J Lipid Res. PubMed ID: 31164391

    Xu, Y., Borcherding, A. F., Heier, C., Tian, G., Roeder, T. and Kuhnlein, R. P. (2019). Chronic dysfunction of Stromal interaction molecule by pulsed RNAi induction in fat tissue impairs organismal energy homeostasis in Drosophila. Sci Rep 9(1): 6989. PubMed ID: 31061470

    Heier, C. and Kuhnlein, R. P. (2018). Triacylglycerol metabolism in Drosophila melanogaster. Genetics 210(4): 1163-1184. PubMed ID: 30523167

    Korandova, M., Krucek, T., Szakosova, K., Kodrik, D., Kuhnlein, R. P., Tomaskova, J. and Capkova Frydrychova, R. (2017). Chronic low-dose pro-oxidant treatment stimulates transcriptional activity of telomeric retroelements and increases telomere length in Drosophila. J Insect Physiol. PubMed ID: 29122549

    Galikova, M., Klepsatel, P., Munch, J. and Kuhnlein, R. P. (2017). Spastic paraplegia-linked phospholipase PAPLA1 is necessary for development, reproduction, and energy metabolism in Drosophila. Sci Rep 7: 46516. PubMed ID: 28422159

    Klepsatel, P., Galikova, M., Xu, Y. and Kuhnlein, R. P. (2016). Thermal stress depletes energy reserves in Drosophila. Sci Rep 6: 33667. PubMed ID: 27641694

    Galikova, M., Diesner, M., Klepsatel, P., Hehlert, P., Xu, Y., Bickmeyer, I., Predel, R. and Kuhnlein, R. P. (2015). Energy Homeostasis Control in Drosophila Adipokinetic Hormone Mutants. Genetics. PubMed ID: 26275422

    Gáliková, M., Diesner, M., Klepsatel, P., Hehlert, P., Xu, Y., Bickmeyer, I., Predel, R. and Kühnlein, R.P. (2015). Energy homeostasis control in Drosophila Adipokinetic hormone mutants. Genetics [Epub ahead of print]. PubMed ID: 26275422

  • Justin Kumar Department of Biology, Indiana University
    Weasner, B. M. and Kumar, J. P. (2022). The timing of cell fate decisions is crucial for initiating pattern formation in the Drosophila eye. Development 149(2). PubMed ID: 35072208

    Ordway, A. J., Teeters, G. M., Weasner, B. M., Weasner, B. P., Policastro, R. and Kumar, J. P. (2021). A multi-gene knockdown approach reveals a new role for Pax6 in controlling organ number in Drosophila. Development 148(9). PubMed ID: 33982759

    Baker, L. R., Weasner, B. M., Nagel, A., Neuman, S. D., Bashirullah, A. and Kumar, J. P. (2018). Eyeless/Pax6 initiates eye formation non-autonomously from the peripodial epithelium. Development. PubMed ID: 29980566

    Palliyil, S., Zhu, J., Baker, L. R., Neuman, S. D., Bashirullah, A. and Kumar, J. P. (2018). Allocation of distinct organ fates from a precursor field requires a shift in expression and function of gene regulatory networks. PLoS Genet 14(1): e1007185. PubMed ID: 29351292

    Zhu, J., Palliyil, S., Ran, C. and Kumar, J. P. (2017). Drosophila Pax6 promotes development of the entire eye-antennal disc, thereby ensuring proper adult head formation. Proc Natl Acad Sci U S A 114(23): 5846-5853. PubMed ID: 28584125

    Weasner, B. M., Weasner, B. P., Neuman, S. D., Bashirullah, A. and Kumar, J. P. (2016). Retinal expression of the Drosophila eyes absent gene is controlled by several cooperatively acting cis-regulatory elements. PLoS Genet 12(12): e1006462. PubMed ID: 27930646

    Iyer, J., Wang, Q., Le, T., Pizzo, L., Gronke, S., Ambegaokar, S., Imai, Y., Srivastava, A., Llamusi Troisi, B., Mardon, G., Artero, R., Jackson, G. R., Isaacs, A. M., Partridge, L., Kumar, J. P. and Girirajan, S. (2016). Quantitative Assessment of Eye Phenotypes for Functional Genetic Studies Using Drosophila melanogaster. G3 (Bethesda). PubMed ID: 26994292

    Spratford, C. M. and Kumar, J. P. (2015). Extramacrochaetae functions in dorsal-ventral patterning of Drosophila imaginal discs. Development 142: 1006-1015. PubMed ID: 25715400

    Spratford, C. M. and Kumar, J. P. (2014). Dissection and Immunostaining of Imaginal Discs from Drosophila melanogaster. J Vis Exp. PubMed ID: 25285379

    Anderson, A. M., Weasner, B. P., Weasner, B. M. and Kumar, J. P. (2014). The Drosophila Wilms' Tumor 1-Associating Protein (WTAP) Homolog is Required for Eye Development. Dev Biol. PubMed ID: 24690230

  • Sudhir Kumar Temple University
    Kumar, S., Konikoff, C., Sanderford, M., Liu, L., Newfeld, S., Ye, J. and Kulathinal, R. J. (2017). FlyExpress 7: An Integrated Discovery Platform To Study Co-expressed Genes Using In Situ Hybridization Images in Drosophila. G3 (Bethesda). PubMed ID: 28667017

    Katsura, Y., Stanley, C. E., Jr., Kumar, S. and Nei, M. (2017). The Reliability and Stability of an Inferred Phylogenetic Tree from Empirical Data. Mol Biol Evol 34(3): 718-723. PubMed ID: 28100791

    Chen, Z. X., Golovnina, K., Sultana, H., Kumar, S. and Oliver, B. (2014). Transcriptional effects of gene dose reduction. Biol Sex Differ 5: 5. PubMed ID: 24581086

    Montiel, I., Konikoff, C., Braun, B., Packard, M., Gramates, S. L., Sun, Q., Ye, J. and Kumar, S. (2014). myFX: A turn-key software for laboratory desktops to analyze spatial patterns of gene expression in Drosophila embryos. Bioinformatics. PubMed ID: 24413523

    Zhang, W., Feng, D., Li, R., Chernikov, A., Chrisochoides, N., Osgood, C., Konikoff, C., Newfeld, S., Kumar, S. and Ji, S. (2013). A mesh generation and machine learning framework for Drosophila gene expression pattern image analysis. BMC Bioinformatics 14: 372. PubMed ID: 24373308

    Sun, Q., Muckatira, S., Yuan, L., Ji, S., Newfeld, S., Kumar, S. and Ye, J. (2013). Image-level and group-level models for Drosophila gene expression pattern annotation. BMC Bioinformatics 14: 350. PubMed ID: 24299119

    Yuan, L., Pan, C., Ji, S., McCutchan, M., Zhou, Z. H., Newfeld, S. J., Kumar, S. and Ye, J. (2013). Automated Annotation of Developmental Stages of Drosophila Embryos in Images Containing Spatial Patterns of Expression. Bioinformatics. PubMed ID: 24300439

    Gilbert, J. D., Acquisti, C., Martinson, H. M., Elser, J. J., Kumar, S. and Fagan, W. F. (2013). GRASP Genomic Resource Access for Stoichioproteomics: comparative explorations of the atomic content of 12 Drosophila proteomes. BMC Genomics 14: 599. PubMed ID: 24007337

    Yuan, L., Woodard, A., Ji, S., Jiang, Y., Zhou, Z. H., Kumar, S., Ye, J. (2012) Learning sparse representations for fruit-fly gene expression pattern image annotation and retrieval. BMC Bioinformatics 13: 107. PubMed ID: 22621237

    Kumar, S., Konikoff, C., Van Emden, B., Busick, C., Davis, K. T., Ji, S., Wu, L. W., Ramos, H., Brody, T., Panchanathan, S., Ye, J., Karr, T. L., Gerold, K., McCutchan, M. and Newfeld, S. J. (2011). FlyExpress: visual mining of spatiotemporal patterns for genes and publications in Drosophila embryogenesis. Bioinformatics 27: 3319-3320. PubMed ID: 21994220

  • Vimlesh Kumar Biological Sciences, Indian Institute of Science Education and Research Bhopal
    Choudhury, S. D., Dwivedi, M. K., Pippadpally, S., Patnaik, A., Mishra, S., Padinjat, R. and Kumar, V. (2022). AP2 Regulates Thickveins Trafficking to Attenuate NMJ Growth Signaling in Drosophila. eNeuro 9(5). PubMed ID: 36180220

    Khyati, Malik, I., Agrawal, N. and Kumar, V. (2020). Melatonin and curcumin reestablish disturbed circadian gene expressions and restore locomotion ability and eclosion behavior in Drosophila model of Huntington's disease. Chronobiol Int: 1-18. PubMed ID: 33334207

    Raut, S., Mallik, B., Parichha, A., V, A., Sahi, C. and Kumar, V. (2017). RNAi-Mediated Reverse Genetic Screen Identified Drosophila Chaperones Regulating Eye and Neuromuscular Junction Morphology. G3 (Bethesda). PubMed ID: 28500055

    Mallik, B., Dwivedi, M. K., Mushtaq, Z., Kumari, M., Verma, P. K. and Kumar, V. (2017). Regulation of neuromuscular junction organization by Rab2 and its effector ICA69 in Drosophila. Development. PubMed ID: 28455372

    Mushtaq, Z., Choudhury, S. D., Gangwar, S. K., Orso, G. and Kumar, V. (2016). Human Senataxin Modulates Structural Plasticity of the Neuromuscular Junction in Drosophila through a Neuronally Conserved TGFbeta Signalling Pathway. Neurodegener Dis 16(5-6): 324-336. PubMed ID: 27197982

    Choudhury, S. D., Mushtaq, Z., Reddy-Alla, S., Balakrishnan, S. S., Thakur, R. S., Krishnan, K. S., Raghu, P., Ramaswami, M. and Kumar, V. (2016). sigma2-Adaptin Facilitates Basal Synaptic Transmission and Is Required for Regenerating Endo-Exo Cycling Pool Under High-Frequency Nerve Stimulation in Drosophila. Genetics 203(1): 369-385. PubMed ID: 26920756

  • Kazuhiko Kume Kumamoto University, Kumamoto, Japan
    Kato, Y. S., Tomita, J. and Kume, K. (2022). Interneurons of fan-shaped body promote arousal in Drosophila. PLoS One 17(11): e0277918. PubMed ID: 36409701

    Yamaguchi, S. T., Tomita, J. and Kume, K. (2022). Insulin signaling in clock neurons regulates sleep in Drosophila. Biochem Biophys Res Commun 591: 44-49. PubMed ID: 34998032

    Tomita, J., Ban, G., Kato, Y. S. and Kume, K. (2021). Protocerebral Bridge Neurons That Regulate Sleep in Drosophila melanogaster. Front Neurosci 15: 647117. PubMed ID: 34720844

    Hasegawa, T., Tomita, J., Hashimoto, R., Ueno, T., Kume, S. and Kume, K. (2017). Sweetness induces sleep through gustatory signalling independent of nutritional value in a starved fruit fly. Sci Rep 7(1): 14355. PubMed ID: 29084998

    Sano, H., Nakamura, A., Texada, M. J., Truman, J. W., Ishimoto, H., Kamikouchi, A., Nibu, Y., Kume, K., Ida, T. and Kojima, M. (2015). The nutrient-responsive hormone CCHamide-2 controls growth by regulating Insulin-like peptides in the brain of Drosophila melanogaster. PLoS Genet 11: e1005209. PubMed ID: 26020940

    Tomita, J., Ueno, T., Mitsuyoshi, M., Kume, S. and Kume, K. (2015). The NMDA receptor promotes sleep in the fruit fly, Drosophila melanogaster. PLoS One 10: e0128101. PubMed ID: 26023770

    Ueno, T. and Kume, K. (2014). Functional characterization of dopamine transporter in vivo using Drosophila melanogaster behavioral assays. Front Behav Neurosci 8: 303. PubMed ID: 25232310

    Ueno, T., et al. (2012). Identification of a dopamine pathway that regulates sleep and arousal in Drosophila. Nat. Neurosci. 15(11): 1516-23. PubMed ID: 23064381

    Ueno, T., Masuda, N., Kume, S. and Kume, K. (2012). Dopamine modulates the rest period length without perturbation of its power law distribution in Drosophila melanogaster. PLoS One 7: e32007. PubMed ID: 22359653

  • Sam Kunes Harvard Neurosciences
    Daniele, J. R., Chu, T. and Kunes, S. (2017). A novel proteolytic event controls Hedgehog intracellular sorting and distribution to receptive fields. Biol Open [Epub ahead of print]. PubMed ID: 28298318

    Daniele, J. R., Baqri, R. M. and Kunes, S. (2017). Analysis of axonal trafficking via a novel live imaging technique reveals distinct Hedgehog transport kinetics. Biol Open [Epub ahead of print]. PubMed ID: 28298319

    Baqri, R. M., Pietron, A. V., Gokhale, R. H., Turner, B. A., Kaguni, L. S., Shingleton, A. W., Kunes, S. and Miller, K. E. (2014). Mitochondrial chaperone TRAP1 activates the mitochondrial UPR and extends healthspan in Drosophila. Mech Ageing Dev. PubMed ID: 25265088

    Dearborn, R. E., Jr., Dai, Y., Reed, B., Karian, T., Gray, J. and Kunes, S. (2012). Reph, a regulator of Eph receptor expression in the Drosophila melanogaster optic lobe. PLoS One 7: e37303. PubMed ID: 22615969

    Song, E., de Bivort, B., Dan, C. and Kunes, S. (2012). Determinants of the Drosophila odorant receptor pattern. Dev Cell 22: 363-376. PubMed ID: 22340498

    Huang, H. R., Chen, Z. J., Kunes, S., Chang, G. D. and Maniatis, T. (2010). Endocytic pathway is required for Drosophila Toll innate immune signaling. Proc Natl Acad Sci U S A 107: 8322-8327. PubMed ID: 20404143

  • Chay Kuo Department of Cell Biology, Duke University
    Lyons, G. R., Andersen, R. O., Abdi, K., Song, W. S. and Kuo, C. T. (2014). Cysteine Proteinase-1 and Cut Protein Isoform Control Dendritic Innervation of Two Distinct Sensory Fields by a Single Neuron. Cell Rep. PubMed ID: 24582961

  • Erina Kuranaga Laboratory for Histgenetic Dynamics, Tohoku University
    Uechi, H. and Kuranaga, E. (2019). The tricellular junction protein Sidekick regulates vertex dynamics to promote bicellular junction extension. Dev Cell. PubMed ID: 31353316

    Ming, M., Obata, F., Kuranaga, E. and Miura, M. (2014). Persephone/Spatzle pathogen sensors mediate the activation of Toll receptor signaling in response to endogenous danger signals in apoptosis-deficient Drosophila. J Biol Chem 289(11): 7558-7568. PubMed ID: 24492611

    Ihara, R., Matsukawa, K., Nagata, Y., Kunugi, H., Tsuji, S., Chihara, T., Kuranaga, E., Miura, M., Wakabayashi, T., Hashimoto, T. and Iwatsubo, T. (2013). RNA binding mediates neurotoxicity in the transgenic Drosophila model of TDP-43 proteinopathy. Hum Mol Genet 22(22): 4474-4484. PubMed ID: 23804749

  • Estee Kurant Rappaport Faculty of Medicine, Technion, Haifa
    Zohar-Fux, M., Ben-Hamo-Arad, A., Arad, T., Volin, M., Shklyar, B., Hakim-Mishnaevski, K., Porat-Kuperstein, L., Kurant, E. and Toledano, H. (2022). The phagocytic cyst cells in Drosophila testis eliminate germ cell progenitors via phagoptosis. Sci Adv 8(24): eabm4937. PubMed ID: 35714186

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

    Dabool, L., Hakim-Mishnaevski, K., Juravlev, L., Flint-Brodsly, N., Mandel, S. and Kurant, E. (2020). Drosophila Skp1 Homologue SkpA Plays a Neuroprotective Role in Adult Brain. iScience 23(8): 101375. PubMed ID: 32739834

    Hakim-Mishnaevski, K., Flint-Brodsly, N., Shklyar, B., Levy-Adam, F. and Kurant, E. (2019). Glial phagocytic receptors promote neuronal loss in adult Drosophila brain. Cell Rep 29(6): 1438-1448.e1433. PubMed ID: 31693886

    Shlyakhover, E., Shklyar, B., Hakim-Mishnaevski, K., Levy-Adam, F. and Kurant, E. (2018). Drosophila GATA factor serpent establishes phagocytic ability of embryonic macrophages. Front Immunol 9: 266. PubMed ID: 29568295

    Hilu-Dadia, R., Hakim-Mishnaevski, K., Levy-Adam, F. and Kurant, E. (2018). Draper-mediated JNK signaling is required for glial phagocytosis of apoptotic neurons during Drosophila metamorphosis. Glia [Epub ahead of print]. PubMed ID: 29520845

    Shklover, J., Levy-Adam, F. and Kurant, E. (2015). The role of Drosophila TNF Eiger in developmental and damage-induced neuronal apoptosis. FEBS Lett [Epub ahead of print]. PubMed ID: 25754009

    Shklover, J., Mishnaevski, K., Levy-Adam, F. and Kurant, E. (2015). JNK pathway activation is able to synchronize neuronal death and glial phagocytosis in Drosophila. Cell Death Dis 6: e1649. PubMed ID: 25695602

    Shklyar, B., Levy-Adam, F. and Kurant, E. (2015). Drosophila Drosophila Model for Studying PhagocytosisPhagocytosis Following Neuronal Cell DeathNeuronal Cell Death. Methods Mol Biol 1254: 359-368. PubMed ID: 25431078

    Shklyar, B., Sellman, Y., Shklover, J., Mishnaevski, K., Levy-Adam, F. and Kurant, E. (2014). Developmental regulation of glial cell phagocytic function during Drosophila embryogenesis. Dev Biol. PubMed ID: 25046770

  • Shoichiro Kurata Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, Sendai
    Tang, C., Kurata, S. and Fuse, N. (2022). Genetic dissection of innate immune memory in Drosophila melanogaster. Front Immunol 13: 857707. PubMed ID: 35990631

    Fuse, N., Okamori, C., Okaji, R., Tang, C., Hirai, K. and Kurata, S. (2022). Transcriptome features of innate immune memory in Drosophila. PLoS Genet 18(10): e1010005. PubMed ID: 36252180

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

    Kumada, K., Fuse, N., Tamura, T., Okamori, C. and Kurata, S. (2019). HSP70/DNAJA3 chaperone/cochaperone regulates NF-kappaB activity in immune responses. Biochem Biophys Res Commun. PubMed ID: 31005254

    Kanoh, H., Kato, H., Suda, Y., Hori, A., Kurata, S. and Kuraishi, T. (2018). Dual comprehensive approach to decipher the Drosophila Toll pathway, ex vivo RNAi screenings and immunoprecipitation-mass spectrometry. Biochem Biophys Res Commun. PubMed ID: 30497778

    Masuko, K., Furuhashi, H., Komaba, K., Numao, E., Nakajima, R., Fuse, N. and Kurata, S. (2018). Nuclear Lamin is required for Winged Eye-mediated transdetermination of Drosophila imaginal disc. Genes Cells. PubMed ID: 29968323

    Masuko, K., Fuse, N., Komaba, K., Katsuyama, T., Nakajima, R., Furuhashi, H. and Kurata, S. (2018). winged eye induces transdetermination of Drosophila imaginal disc by acting in concert with a histone methyltransferase, Su(var)3-9. Cell Rep 22(1): 206-217. PubMed ID: 29298422

    Hori, A., Kurata, S and Kuraishi, T. (2018). Unexpected role of the IMD pathway in Drosophila gut defense against Staphylococcus aureus. Biochem Biophys Res Commun. 495(1): 395-400. PubMed ID: 29108998

    Kenmoku, H., Hori, A., Kuraishi, T. and Kurata, S. (2017). A novel mode of induction of the humoral innate immune response in Drosophila larvae. Dis Model Mech 10(3): 271-281. PubMed ID: 28250052

    Kenmoku, H., Ishikawa, H., Ote, M., Kuraishi, T. and Kurata, S. (2016). A subset of neurons controls the permeability of the peritrophic matrix and midgut structure in Drosophila adults. J Exp Biol. PubMed ID: 27229474

    Ozawa, N., Furuhashi, H., Masuko, K., Numao, E., Makino, T., Yano, T. and Kurata, S. (2016). Organ identity specification factor WGE localizes to the histone locus body and regulates histone expression to ensure genomic stability in Drosophila. Genes Cells 21: 442-456. PubMed ID: 27145109

  • Mitzi Kuroda Department of Genetics, Harvard Medical School
    Kang, H., Cabrera, J. R., Zee, B. M., Kang, H. A., Jobe, J. M., Hegarty, M. B., Barry, A. E., Glotov, A., Schwartz, Y. B. and Kuroda, M. I. (2022). Variant Polycomb complexes in Drosophila consistent with ancient functional diversity. Sci Adv 8(36): eadd0103. PubMed ID: 36070387

    Copur, O., Gorchakov, A., Finkl, K., Kuroda, M. I. and Muller, J. (2018). Sex-specific phenotypes of histone H4 point mutants establish dosage compensation as the critical function of H4K16 acetylation in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 30530664

    Kang, H., Jung, Y. L., McElroy, K. A., Zee, B. M., Wallace, H. A., Woolnough, J. L., Park, P. J. and Kuroda, M. I. (2017). Bivalent complexes of PRC1 with orthologs of BRD4 and MOZ/MORF target developmental genes in Drosophila. Genes Dev 31(19): 1988-2002. PubMed ID: 29070704

    McElroy, K. A., Jung, Y. L., Zee, B. M., Wang, C. I., Park, P. J. and Kuroda, M. I. (2017). upSET, the Drosophila homologue of SET3, Is required for viability and the proper balance of active and repressive chromatin marks. G3 (Bethesda). PubMed ID: 28064188

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

    Zee, B. M., Alekseyenko, A. A., McElroy, K. A. and Kuroda, M. I. (2016). Streamlined discovery of cross-linked chromatin complexes and associated histone modifications by mass spectrometry. Proc Natl Acad Sci U S A 113: 1784-1789. PubMed ID: 26831069

    Jung, Y.L., Kang, H., Park, P.J. and Kuroda, M.I. (2016). Correspondence of Drosophila Polycomb Group proteins with broad H3K27me3 silent domains. Fly (Austin) [Epub ahead of print]. PubMed ID: 26940990

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

    Jung, Y. L., Luquette, L. J., Ho, J. W., Ferrari, F., Tolstorukov, M., Minoda, A., Issner, R., Epstein, C. B., Karpen, G. H., Kuroda, M. I. and Park, P. J. (2014). Impact of sequencing depth in ChIP-seq experiments. Nucleic Acids Res 42: e74. PubMed ID: 24598259

    Ferrari, F., Alekseyenko, A. A., Park, P. J. and Kuroda, M. I. (2014). Transcriptional control of a whole chromosome: emerging models for dosage compensation. Nat Struct Mol Biol 21: 118-125. PubMed ID: 24500429

    McElroy, K. A., Kang, H. and Kuroda, M. I. (2014). Are we there yet? Initial targeting of the Male-Specific Lethal and Polycomb group chromatin complexes in Drosophila. Open Biol 4: 140006. PubMed ID: 24671948

  • Mitzi Kuroda Biological and Biomedical Sciences, Harvard University
    Kang, H., McElroy, K.A., Jung, Y.L., Alekseyenko, A.A., Zee, B.M., Park, P.J. and Kuroda, M.I. (2015). Sex comb on midleg (Scm) is a functional link between PcG-repressive complexes in Drosophila. Genes Dev 29: 1136-1150. PubMed ID: 26063573

    Alekseyenko, A. A., Ellison, C. E., Gorchakov, A. A., Zhou, Q., Kaiser, V. B., Toda, N., Walton, Z., Peng, S., Park, P. J., Bachtrog, D., Kuroda, M. I. (2013) Conservation and de novo acquisition of dosage compensation on newly evolved sex chromosomes in Drosophila. Genes Dev 27: 853-858. PubMed ID: 23630075

    É, A. A., Ho, J. W., Peng, S., Gelbart, M., Tolstorukov, M. Y., Plachetka, A., Kharchenko, P. V., Jung, Y. L., Gorchakov, A. A., Larschan, E., Gu, T., Minoda, A., Riddle, N. C., Schwartz, Y. B., Elgin, S. C., Karpen, G. H., Pirrotta, V., Kuroda, M. I. and Park, P. J. (2012). Sequence-specific targeting of dosage compensation in Drosophila favors an active chromatin context. PLoS Genet 8: e1002646. PubMed ID: 22570616

    Wang, C. I., Alekseyenko, A. A., LeRoy, G., Elia, A. E., Gorchakov, A. A., Britton, L. M., Elledge, S. J., Kharchenko, P. V., Garcia, B. A. and Kuroda, M. I. (2013). Chromatin proteins captured by ChIP-mass spectrometry are linked to dosage compensation in Drosophila. Nat Struct Mol Biol 20: 202-209. PubMed ID: 23295261

    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

  • Éva Kurucz Biological Research Centre, Hungarian Academy of Sciences, Szeged
    Cinege, G., Zsamboki, J., Vidal-Quadras, M., Uv, A., Csordas, G., Honti, V., Gabor, E., Hegedus, Z., Varga, G. I. B., Kovacs, A. L., Juhasz, G., Williams, M. J., Ando, I. and Kurucz, E. (2017). Genes encoding cuticular proteins are components of the Nimrod gene cluster in Drosophila. Insect Biochem Mol Biol 87: 45-54. PubMed ID: 28633893

    Kari, B., Csordas, G., Honti, V., Cinege, G., Williams, M. J., Ando, I. and Kurucz, E. (2016). The raspberry Gene Is Involved in the Regulation of the Cellular Immune Response in Drosophila melanogaster. PLoS One 11: e0150910. PubMed ID: 26942456

    Bretscher, A. J., Honti, V., Binggeli, O., Burri, O., Poidevin, M., Kurucz, E., Zsamboki, J., Ando, I. and Lemaitre, B. (2015). The Nimrod transmembrane receptor Eater is required for hemocyte attachment to the sessile compartment in Drosophila melanogaster. Biol Open 4: 355-363. PubMed ID: 25681394

    Markus, R., Lerner, Z., Honti, V., Csordas, G., Zsamboki, J., Cinege, G., Parducz, A., Lukacsovich, T., Kurucz, E. and Ando, I. (2015). Multinucleated Giant Hemocytes Are Effector Cells in Cell-Mediated Immune Responses of Drosophila. J Innate Immun 7: 340-353. PubMed ID: 25659341

    Csordas, G., Varga, G. I., Honti, V., Jankovics, F., Kurucz, E. and Ando, I. (2014). In vivo immunostaining of hemocyte compartments in Drosophila for live imaging. PLoS One 9: e98191. PubMed ID: 24892745

  • Charalambos Kyriacou University of Leicester
    Pegoraro, M., Fishman, B., Zonato, V., Zouganelis, G., Francis, A., Kyriacou, C. P. and Tauber, E. (2022). Photoperiod-Dependent Expression of MicroRNA in Drosophila. Int J Mol Sci 23(9). PubMed ID: 35563325

    Maddison, D. C., Alfonso-Nunez, M., Swaih, A. M., Breda, C., Campesan, S., Allcock, N., Straatman-Iwanowska, A., Kyriacou, C. P. and Giorgini, F. (2020). A novel role for kynurenine 3-monooxygenase in mitochondrial dynamics. PLoS Genet 16(11): e1009129. PubMed ID: 33170836

    de Azevedo, R. V. M., Hansen, C., Chen, K. F., Rosato, E. and Kyriacou, C. P. (2020). Disrupted Glutamate Signaling in Drosophila Generates Locomotor Rhythms in Constant Light. Front Physiol 11: 145. PubMed ID: 32210832

    Nagy, D., Cusumano, P., Andreatta, G., Anduaga, A. M., Hermann-Luibl, C., Reinhard, N., Gesto, J., Wegener, C., Mazzotta, G., Rosato, E., Kyriacou, C. P., Helfrich-Forster, C. and Costa, R. (2019). Peptidergic signaling from clock neurons regulates reproductive dormancy in Drosophila melanogaster. PLoS Genet 15(6): e1008158. PubMed ID: 31194738

    Noreen, S., Pegoraro, M., Nouroz, F., Tauber, E. and Kyriacou, C. P. (2018). Interspecific studies of circadian genes period and timeless in Drosophila. Gene [Epub ahead of print]. PubMed ID: 29353056

    Zonato, V., Vanin, S., Costa, R., Tauber, E. and Kyriacou, C. P. (2017). Inverse European Latitudinal Cline at the timeless Locus of Drosophila melanogaster Reveals Selection on a Clock Gene: Population Genetics of ls-tim. J Biol Rhythms: 748730417742309. PubMed ID: 29183263

    Schiesari, L., Andreatta, G., Kyriacou, C. P., O'Connor, M. B. and Costa, R. (2016). The Insulin-Like Proteins dILPs-2/5 Determine Diapause Inducibility in Drosophila. PLoS One 11: e0163680. PubMed ID: 27689881

    Adewoye, A.B., Kyriacou, C.P. and Tauber, E. (2015). Identification and functional analysis of early gene expression induced by circadian light-resetting in Drosophila. BMC Genomics 16: 570. PubMed ID: 26231660

    Green, E. W., O'Callaghan, E. K., Hansen, C. N., Bastianello, S., Bhutani, S., Vanin, S., Armstrong, J. D., Costa, R. and Kyriacou, C. P. (2015). Drosophila circadian rhythms in seminatural environments: Summer afternoon component is not an artifact and requires TrpA1 channels. Proc Natl Acad Sci U S A 112: 8702-8707. PubMed ID: 26124142

    Pegoraro, M., Picot, E., Hansen, C. N., Kyriacou, C. P., Rosato, E. and Tauber, E. (2015). Gene Expression Associated with Early and Late Chronotypes in Drosophila melanogaster. Front Neurol 6: 100. PubMed ID: 26097463

    Montelli, S., Mazzotta, G., Vanin, S., Caccin, L., Corra, S., De Pitta, C., Boothroyd, C., Green, E. W., Kyriacou, C. P. and Costa, R. (2015). period and timeless mRNA Splicing Profiles under Natural Conditions in Drosophila melanogaster. J Biol Rhythms 30: 217-227. PubMed ID: 25994101

    Green, E. W., O'Callaghan, E. K., Pegoraro, M., Armstrong, J. D., Costa, R. and Kyriacou, C. P. (2015). Genetic analysis of Drosophila circadian behavior in seminatural conditions. Methods Enzymol 551: 121-133. PubMed ID: 25662454


  • Return to The Interactive Fly