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


  • Daniel Babcock Biological Sciences, Lehigh University, Bethlehem, PA
    Sidisky, J. M., de Paula Moreira, D., Okumus, M., Caratenuto, R., Drost, C., Connors, B., Hussain, S., Alkhatib, S. and Babcock, D. T. (2023). Genome-wide analysis reveals novel regulators of synaptic maintenance in Drosophila. Genetics 223(4). PubMed ID: 36799927

    Davis, J., Kolaski, E. and Babcock, D. T. (2022). Vexed mutations promote degeneration of dopaminergic neurons through excessive activation of the innate immune response. NPJ Parkinsons Dis 8(1): 147. PubMed ID: 36323700

    Sidisky, J. M., Weaver, D., Hussain, S., Okumus, M., Caratenuto, R. and Babcock, D. (2021). Mayday sustains trans-synaptic BMP signaling required for synaptic maintenance with age. Elife 10. PubMed ID: 33667157

    Sidisky, J. M. and Babcock, D. T. (2020). Visualizing Synaptic Degeneration in Adult Drosophila in Association with Neurodegeneration. J Vis Exp(159). PubMed ID: 32478750

    Cunningham, P. C., Waldeck, K., Ganetzky, B. and Babcock, D. T. (2018). Neurodegeneration and locomotor dysfunction in Drosophila scarlet mutants. J Cell Sci 131(18). PubMed ID: 30154211

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

  • Erika Bach New York University School of Medicine
    Herrera, S. C., Sainz de la Maza, D., Grmai, L., Margolis, S., Plessel, R., Burel, M., O'Connor, M., Amoyel, M. and Bach, E. A. (2021). Proliferative stem cells maintain quiescence of their niche by secreting the Activin inhibitor Follistatin. Dev Cell 56(16): 2284-2294.e2286. PubMed ID: 34363758

    Grmai, L., Harsh, S., Lu, S., Korman, A., Deb, I. B. and Bach, E. A. (2021). Transcriptomic analysis of feminizing somatic stem cells in the Drosophila testis reveals putative downstream effectors of the transcription factor Chinmo. G3 (Bethesda) 11(4). PubMed ID: 33751104

    Sitaram, P., Lu, S., Harsh, S., Herrera, S. C. and Bach, E. A. (2019). Next-generation sequencing reveals increased anti-oxidant response and ecdysone signaling in STAT supercompetitors in Drosophila. G3 (Bethesda). PubMed ID: 31227525

    Bailetti, A. A., Negron-Pineiro, L. J., Dhruva, V., Harsh, S., Lu, S., Bosula, A. and Bach, E. A. (2019). Enhancer of Polycomb/Tip60 represses hematological tumor initiation by negatively regulating JAK/STAT pathway activity. Dis Model Mech. PubMed ID: 31072879

    Herrera, S. C. and Bach, E. A. (2018). JNK signaling triggers spermatogonial dedifferentiation during chronic stress to maintain the germline stem cell pool in the Drosophila testis. Elife 7. PubMed ID: 29985130

    Grmai, L., Hudry, B., Miguel-Aliaga, I. and Bach, E. A. (2018). Chinmo prevents transformer alternative splicing to maintain male sex identity. PLoS Genet 14(2): e1007203. PubMed ID: 29389999

    Court, H., Ahearn, I. M., Amoyel, M., Bach, E. A. and Philips, M. R. (2017). Regulation of NOTCH signaling by RAB7 and RAB8 requires carboxyl methylation by ICMT. J Cell Biol 216(12): 4165-4182. PubMed ID: 29051265

    Anderson, A. M., Bailetti, A. A., Rodkin, E., De, A. and Bach, E. A. (2017). A genetic screen reveals an unexpected role for Yorkie signaling in JAK/STAT-dependent hematopoietic malignancies in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 28620086

    Cook, M. S., Cazin, C., Amoyel, M., Yamamoto, S., Bach, E. and Nystul, T. (2017). Neutral competition for Drosophila follicle and cyst stem cell niches requires vesicle trafficking genes. Genetics [Epub ahead of print]. PubMed ID: 28512187

    Amoyel, M., Hillion, K. H., Margolis, S. R. and Bach, E. A. (2016). Somatic stem cell differentiation is regulated by PI3K/Tor signaling in response to local cues. Development 143(21): 3914-3925. PubMed ID: 27633989

  • Doris Bachtrog Department of Integrative Biology, Center for Theoretical Evolutionary Genomics, Berkeley
    Wei, K. H., Mai, D., Chatla, K. and Bachtrog, D. (2022). Dynamics and Impacts of Transposable Element Proliferation in the Drosophila nasuta Species Group Radiation. Mol Biol Evol 39(5). PubMed ID: 35485457

    Nguyen, A. H., Wang, W., Chong, E., Chatla, K. and Bachtrog, D. (2022). Transposable element accumulation drives size differences among polymorphic Y Chromosomes in Drosophila. Genome Res 32(6): 1074-1088. PubMed ID: 35501131

    Wang, S., Nalley, M. J., Chatla, K., Aldaimalani, R., MacPherson, A., Wei, K. H., Corbett-Detig, R. B., Mai, D. and Bachtrog, D. (2022). Neo-sex chromosome evolution shapes sex-dependent asymmetrical introgression barrier. Proc Natl Acad Sci U S A 119(19): e2119382119. PubMed ID: 35512091

    Wang, S., Nalley, M. J., Chatla, K., Aldaimalani, R., MacPherson, A., Wei, K. H., Corbett-Detig, R. B., Mai, D. and Bachtrog, D. (2022). Neo-sex chromosome evolution shapes sex-dependent asymmetrical introgression barrier. Proc Natl Acad Sci U S A 119(19): e2119382119. PubMed ID: 35512091

    Wei, K. H., Chan, C. and Bachtrog, D. (2021). Establishment of H3K9me3-dependent heterochromatin during embryogenesis in Drosophila miranda. Elife 10. PubMed ID: 34128466

    Nguyen, A. H. and Bachtrog, D. (2021). Toxic Y chromosome: Increased repeat expression and age-associated heterochromatin loss in male Drosophila with a young Y chromosome. PLoS Genet 17(4): e1009438. PubMed ID: 33886541

    Wei, K. H., Gibilisco, L. and Bachtrog, D. (2020). Epigenetic conflict on a degenerating Y chromosome increases mutational burden in Drosophila males. Nat Commun 11(1): 5537. PubMed ID: 33139741

    Brown, E. J., Nguyen, A. H. and Bachtrog, D. (2020). The Drosophila Y chromosome affects heterochromatin integrity genome-wide. Mol Biol Evol. PubMed ID: 32211857

    Brown, E. J., Nguyen, A. H. and Bachtrog, D. (2020). The Y chromosome may contribute to sex-specific ageing in Drosophila. Nat Ecol Evol. PubMed ID: 32313175

    Bracewell, R. and Bachtrog, D. (2020). Complex evolutionary history of the Y chromosome in flies of the Drosophila obscura species group. Genome Biol Evol. PubMed ID: 32176296

    Wei, K. H. and Bachtrog, D. (2019). Ancestral male recombination in Drosophila albomicans produced geographically restricted neo-Y chromosome haplotypes varying in age and onset of decay. PLoS Genet 15(11): e1008502. PubMed ID: 31738748

    Mai, D., Nalley, M. J. and Bachtrog, D. (2019). Patterns of genomic differentiation in the Drosophila nasuta species complex. Mol Biol Evol. PubMed ID: 31556453

  • Juan Domingo Bacigalupo Doctorado en Biología Molecular Celular y Neurociencias, Universidad de Chile
    Delgado, R., Munoz, Y., Pena-Cortes, H., Giavalisco, P. and Bacigalupo, J. (2014). Diacylglycerol Activates the Light-Dependent Channel TRP in the Photosensitive Microvilli of Drosophila melanogaster Photoreceptors. J Neurosci 34: 6679-6686. PubMed ID: 24806693

    Munoz, Y., Fuenzalida, K., Bronfman, M., Gatica, A., Sepulveda, M., Bacigalupo, J., Roth, A. D. and Delgado, R. (2013). Fatty acid composition of Drosophila photoreceptor light-sensitive microvilli. Biol Res 46: 289-294. PubMed ID: 24346077

    Astorga, G., Hartel, S., Sanhueza, M. and Bacigalupo, J. (2012). TRP, TRPL and cacophony channels mediate Ca2+ influx and exocytosis in photoreceptors axons in Drosophila. PLoS One 7: e44182. PubMed ID: 22952921

  • Paul Badenhorst Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham
    Kwon, S. Y., Jang, B. and Badenhorst, P. (2021). The ISWI chromatin remodelling factor NURF is not required for mitotic male X chromosome organisation. MicroPubl Biol 2021. PubMed ID: 33537560

    Kwon, S. Y., Massey, K., Watson, M. A., Hussain, T., Volpe, G., Buckley, C. D., Nicolaou, A. and Badenhorst, P. (2020). Oxidised metabolites of the omega-6 fatty acid linoleic acid activate dFOXO. Life Sci Alliance 3(2). PubMed ID: 31992650

    Choudhury, S.R., Singh, A.K., McLeod, T., Blanchette, M., Jang, B., Badenhorst, P., Kanhere, A. and Brogna, S. (2016). Exon Junction Complex proteins bind nascent transcripts independently of pre-mRNA splicing in Drosophila melanogaster. Elife [Epub ahead of print]. PubMed ID: 27879206

    Kwon, S. Y., Grisan, V., Jang, B., Herbert, J. and Badenhorst, P. (2016). Genome-wide mapping targets of the metazoan chromatin remodeling factor NURF reveals nucleosome remodeling at enhancers, core promoters and gene insulators. PLoS Genet 12: e1005969. PubMed ID: 27046080

    Walker, J., Kwon, S. Y., Badenhorst, P., East, P., McNeill, H. and Svejstrup, J. Q. (2011). Role of elongator subunit Elp3 in Drosophila melanogaster larval development and immunity. Genetics 187: 1067-1075. PubMed ID: 21288872

    Stofanko, M., Kwon, S. Y. and Badenhorst, P. (2010). Lineage tracing of lamellocytes demonstrates Drosophila macrophage plasticity. PLoS One 5: e14051. PubMed ID: 21124962

  • Gyeong Hun Baeg Yong Loo Lin School of Medicine, Singapore
    Tan, S. W. S., Yip, G. W., Suda, T. and Baeg, G. H. (2017). Small Maf functions in the maintenance of germline stem cells in the Drosophila testis. Redox Biol 15: 125-134. PubMed ID: 29245136

    Tan, S. W. S., Lee, Q. Y., Wong, B. S. E., Cai, Y. and Baeg, G. H. (2017). Redox Homeostasis Plays Important Roles in the Maintenance of the Drosophila Testis Germline Stem Cells. Stem Cell Reports 9(1): 342-354. PubMed ID: 28669604

    Ng, C. T., Yong, L. Q., Hande, M. P., Ong, C. N., Yu, L. E., Bay, B. H. and Baeg, G. H. (2017). Zinc oxide nanoparticles exhibit cytotoxicity and genotoxicity through oxidative stress responses in human lung fibroblasts and Drosophila melanogaster. Int J Nanomedicine 12: 1621-1637. PubMed ID: 28280330

    Ong, C., Lee, Q. Y., Cai, Y., Liu, X., Ding, J., Yung, L. Y., Bay, B. H. and Baeg, G. H. (2016). Silver nanoparticles disrupt germline stem cell maintenance in the Drosophila testis. Sci Rep 6: 20632. PubMed ID: 26847594

    Liu, Z., Zhong, G., Chai, P. C., Luo, L., Liu, S., Yang, Y., Baeg, G. H. and Cai, Y. (2015). Coordinated niche-associated signals promote germline homeostasis in the Drosophila ovary. J Cell Biol 211(2): 469-484. PubMed ID: 26504174

  • Eric Baehrecke University of Massachusetts Medical School
    Shen, J. L., Doherty, J., Allen, E., Fortier, T. M. and Baehrecke, E. H. (2022). Atg6 promotes organismal health by suppression of cell stress and inflammation. Cell Death Differ. PubMed ID: 35523956

    Wang, R., Miao, G., Shen, J. L., Fortier, T. M. and Baehrecke, E. H. (2022). ESCRT dysfunction compromises endoplasmic reticulum maturation and autophagosome biogenesis in Drosophila. Curr Biol 32(6): 1262-1274.e1264. PubMed ID: 35134326

    Lam, G., Nam, H. J., Velentzas, P. D., Baehrecke, E. H. and Thummel, C. S. (2021). Drosophila E93 promotes adult development and suppresses larval responses to ecdysone during metamorphosis. Dev Biol. PubMed ID: 34648816

    Allen, E. A., Amato, C., Fortier, T. M., Velentzas, P., Wood, W. and Baehrecke, E. H. (2020). A conserved myotubularin-related phosphatase regulates autophagy by maintaining autophagic flux. J Cell Biol 219(11). PubMed ID: 32915229

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

    Zhao, S., Fortier, T. M. and Baehrecke, E. H. (2018). Autophagy promotes tumor-like stem cell niche occupancy. Curr Biol 28(19): 3056-3064.e3053. PubMed ID: 30270184

    Anding, A. L., Wang, C., Chang, T. K., Sliter, D. A., Powers, C. M., Hofmann, K., Youle, R. J. and Baehrecke, E. H. (2018). Vps13D encodes a ubiquitin-binding protein that is required for the regulation of mitochondrial size and clearance. Curr Biol 28(2): 287-295.e286. PubMed ID: 29307555

    Lin, L., Rodrigues, F., Kary, C., Contet, A., Logan, M., Baxter, R. H. G., Wood, W. and Baehrecke, E. H. (2017). Complement-Related Regulates Autophagy in Neighboring Cells. Cell 170(1): 158-171.e158. PubMed ID: 28666117

    Lee, T. V., Kamber Kaya, H. E., Simin, R., Baehrecke, E. H. and Bergmann, A. (2016). The initiator caspase Dronc is subject of enhanced autophagy upon proteasome impairment in Drosophila. Cell Death Differ. PubMed ID: 27104928

    Tracy, K., Velentzas, P. D. and Baehrecke, E. H. (2015). Ral GTPase and the exocyst regulate autophagy in a tissue-specific manner. EMBO Rep [Epub ahead of print]. PubMed ID: 26598552

    Zhang, H. and Baehrecke, E. H. (2015). Eaten alive: novel insights into autophagy from multicellular model systems. Trends Cell Biol. PubMed ID: 25862458

    Nelson, C. and Baehrecke, E. H. (2014). Eaten to death. FEBS J 281: 5411-5417. PubMed ID: 25323556

    Anding, A. L. and Baehrecke, E. H. (2014). Vps15 is required for stress induced and developmentally triggered autophagy and salivary gland protein secretion in Drosophila. Cell Death Differ. PubMed ID: 25342466

  • Luis Alberto Baena-Lopez Sir William Dunn School of Pathology, University of Oxford
    Galasso, A., Xu, D. C., Hill, C., Iakovleva, D., Stefana, M. I. and Baena-Lopez, L. A. (2023). Non-apoptotic caspase activation ensures the homeostasis of ovarian somatic stem cells. EMBO Rep: e51716. PubMed ID: 37039000

    Xu, D. C., Wang, L., Yamada, K. M. and Baena-Lopez, L. A. (2022). Non-apoptotic activation of Drosophila caspase-2/9 modulates JNK signaling, the tumor microenvironment, and growth of wound-like tumors. Cell Rep 39(3): 110718. PubMed ID: 35443185

    Arthurton, L., Nahotko, D. A., Alonso, J., Wendler, F. and Baena-Lopez, L. A. (2020). Non-apoptotic caspase activation preserves Drosophila intestinal progenitor cells in quiescence. EMBO Rep: e48892. PubMed ID: 33135280

    Baena-Lopez, L. A., Alexandre, C., Mitchell, A., Pasakarnis, L. and Vincent, J. P. (2013). Accelerated homologous recombination and subsequent genome modification in Drosophila. Development 140(23): 4818-4825. PubMed ID: 24154526

    Baena-Lopez, L. A., Alonso, J., Rodriguez, J. and Santaren, J. F. (2008). The expression of heat shock protein HSP60A reveals a dynamic mitochondrial pattern in Drosophila melanogaster embryos. J Proteome Res 7(7): 2780-2788. PubMed ID: 18549261

    Baena-Lopez, L. A. and Garcia-Bellido, A. (2006). Control of growth and positional information by the graded vestigial expression pattern in the wing of Drosophila melanogaster. Proc Natl Acad Sci U S A 103(37): 13734-13739. PubMed ID: 16950871

  • Claudia Bagni
    Mariano, V., Kanellopoulos, A. K., Aiello, G., Lo, A. C., Legius, E., Achsel, T. and Bagni, C. (2023). SREBP modulates the NADP(+)/NADPH cycle to control night sleep in Drosophila. Nat Commun 14(1): 763. PubMed ID: 36808152

    Mariano, V., Achsel, T., Bagni, C. and Kanellopoulos, A. K. (2020). Modelling Learning and Memory in Drosophila to Understand Intellectual Disabilities. Neuroscience 445: 12-30. PubMed ID: 32730949

    Kanellopoulos, A. K., Mariano, V., Spinazzi, M., Woo, Y. J., McLean, C., Pech, U., Li, K. W., Armstrong, J. D., Giangrande, A., Callaerts, P., Smit, A. B., Abrahams, B. S., Fiala, A., Achsel, T. and Bagni, C. (2020). Aralar Sequesters GABA into Hyperactive Mitochondria, Causing Social Behavior Deficits. Cell 180(6): 1178-1197 e1120. PubMed ID: 32200800

  • Hua Bai Genetics, Development, and Cell Biology, Iowa State University, Ames
    Bouska, M. J. and Bai, H. (2021). Loxl2 is a mediator of cardiac aging in Drosophila melanogaster; genetically examining the role of aging clock genes. G3 (Bethesda). PubMed ID: 34734976

    Bouska, M. J. and Bai, H. (2021). Long noncoding RNA regulation of spermatogenesis via the spectrin cytoskeleton in Drosophila. G3 (Bethesda) 11(5). PubMed ID: 33720346

    Birnbaum, A., Sodders, M., Bouska, M., Chang, K., Kang, P., McNeill, E. and Bai, H. (2020). FOXO Regulates Neuromuscular Junction Homeostasis During Drosophila Aging. Front Aging Neurosci 12: 567861. PubMed ID: 33584240

    Huang, K., Miao, T., Chang, K., Kim, J., Kang, P., Jiang, Q., Simmonds, A. J., Di Cara, F. and Bai, H. (2020). Impaired peroxisomal import in Drosophila oenocytes causes cardiac dysfunction by inducing upd3 as a peroxikine. Nat Commun 11(1): 2943. PubMed ID: 32523050

    Birnbaum, A., Wu, X., Tatar, M., Liu, N. and Bai, H. (2019). Age-Dependent Changes in Transcription Factor FOXO Targeting in Female Drosophila. Front Genet 10: 312. PubMed ID: 31134124

    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

    Kang, P., Chang, K., Liu, Y., Bouska, M., Birnbaum, A., Karashchuk, G., Thakore, R., Zheng, W., Post, S., Brent, C. S., Li, S., Tatar, M. and Bai, H. (2017). Drosophila Kruppel homolog 1 represses lipolysis through interaction with dFOXO. Sci Rep 7(1): 16369. PubMed ID: 29180716

  • Richard Baines University of Manchester
    Bradlaugh, A. A., Fedele, G., Munro, A. L., Hansen, C. N., Hares, J. M., Patel, S., Kyriacou, C. P., Jones, A. R., Rosato, E. and Baines, R. A. (2023). Essential elements of radical pair magnetosensitivity in Drosophila. Nature 615(7950): 111-116. PubMed ID: 36813962

    Dyson, A., Ryan, M., Garg, S., Evans, D. G. and Baines, R. A. (2022). Loss of NF1 in Drosophila Larvae Causes Tactile Hypersensitivity and Impaired Synaptic Transmission at the Neuromuscular Junction. J Neurosci 42(50): 9450-9472. PubMed ID: 36344265

    Mulroe, F., Lin, W. H., Mackenzie-Gray Scott, C., Aourz, N., Fan, Y. N., Coutts, G., Parrish, R. R., Smolders, I., Trevelyan, A., Wykes, R., Allan, S., Freeman, S. and Baines, R. A. (2022). Targeting firing rate neuronal homeostasis can prevent seizures. Dis Model Mech. PubMed ID: 36073607

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

    Mituzaite, J., Petersen, R., Claridge-Chang, A. and Baines, R. A. (2021). Characterization of Seizure Induction Methods in Drosophila. eNeuro 8(4). PubMed ID: 34330816

    Hodge, S. H., Watts, A., Marley, R., Baines, R. A., Hafen, E. and MacDougall, L. K. (2021). Twitchy, the Drosophila orthologue of the ciliary gating protein FBF1/dyf-19, is required for coordinated locomotion and male fertility. Biol Open 10(8). PubMed ID: 34357392

    Hunter, I., Coulson, B., Zarin, A. A. and Baines, R. A. (2021). The Drosophila Larval Locomotor Circuit Provides a Model to Understand Neural Circuit Development and Function. Front Neural Circuits 15: 684969. PubMed ID: 34276315

    Vernon, S. W., Goodchild, J. and Baines, R. A. (2019). A poly-glutamine region in the Drosophila VAChT dictates fill-level of cholinergic synaptic vesicles. eNeuro 6(1). PubMed ID: 30847389

    Lin, W. H. and Baines, R. A. (2019). Myocyte enhancer factor-2 and p300 interact to regulate expression of the homeostatic regulator Pumilio in Drosophila. Eur J Neurosci. PubMed ID: 30687963

    Lin, W. H., Giachello, C. N. and Baines, R. A. (2016). Seizure control through genetic and pharmacological manipulation of Pumilio: a key component of neuronal homeostasis. Dis Model Mech [Epub ahead of print]. PubMed ID: 28067623

  • Chris Bakal Institute of Cancer Research, Chester Beatty Laboratories, London
    Sailem, H., Bousgouni, V., Cooper, S. and Bakal, C. (2014). Cross-talk between Rho and Rac GTPases drives deterministic exploration of cellular shape space and morphological heterogeneity. Open Biol 4: 130132. PubMed ID: 24451547

    Yin, Z., Sadok, A., Sailem, H., McCarthy, A., Xia, X., Li, F., Garcia, M. A., Evans, L., Barr, A. R., Perrimon, N., Marshall, C. J., Wong, S. T. and Bakal, C. (2013). A screen for morphological complexity identifies regulators of switch-like transitions between discrete cell shapes. Nat Cell Biol. PubMed ID: 23748611

    Garcia, M. A., Alvarez, M. S., Sailem, H., Bousgouni, V., Sero, J. and Bakal, C. (2012). Differential RNAi screening provides insights into the rewiring of signalling networks during oxidative stress. Mol Biosyst 8: 2605-2613. PubMed ID: 22790786

    Bakal, C. (2011). Drosophila RNAi screening in a postgenomic world. Brief Funct Genomics 10: 197-205. PubMed ID: 21752787

  • Bruce Baker HHNI, Janelia Farm
    Chen, D., Sitaraman, D., Chen, N., Jin, X., Han, C., Chen, J., Sun, M., Baker, B. S., Nitabach, M. N. and Pan, Y. (2017). Genetic and neuronal mechanisms governing the sex-specific interaction between sleep and sexual behaviors in Drosophila. Nat Commun 8(1): 154. PubMed ID: 28754889

    Montague, S. A. and Baker, B. S. (2016). Memory elicited by courtship conditioning requires mushroom body neuronal subsets similar to those utilized in appetitive memory. PLoS One 11: e0164516. PubMed ID: 27764141

    Meissner, G. W., Luo, S. D., Dias, B. G., Texada, M. J. and Baker, B. S. (2016). Sex-specific regulation of Lgr3 in Drosophila neurons. Proc Natl Acad Sci U S A. PubMed ID: 26884206

    Zhou, C., Franconville, R., Vaughan, A. G., Robinett, C. C., Jayaraman, V. and Baker, B. S. (2015). Central neural circuitry mediating courtship song perception in male Drosophila. Elife 4. PubMed ID: 26390382

    Luo, S. D. and Baker, B. S. (2015). Constraints on the evolution of a doublesex target gene arising from doublesex's pleiotropic deployment. Proc Natl Acad Sci U S A. PubMed ID: 25675536

    Vaughan, A. G., Zhou, C., Manoli, D. S. and Baker, B. S. (2014). Neural Pathways for the Detection and Discrimination of Conspecific Song in D. melanogaster. Curr Biol. PubMed ID: 24794294

    Tran, D. H., Meissner, G. W., French, R. L. and Baker, B. S. (2014). A small subset of fruitless subesophageal neurons modulate early courtship in Drosophila. PLoS One 9: e95472. PubMed ID: 24740138

    Pan, Y. and Baker, B. S. (2014). Genetic identification and separation of innate and experience-dependent courtship behaviors in Drosophila. Cell 156: 236-248. PubMed ID: 24439379

    Fan, P., Manoli, D. S., Ahmed, O. M., Chen, Y., Agarwal, N., Kwong, S., Cai, A. G., Neitz, J., Renslo, A., Baker, B. S. and Shah, N. M. (2013). Genetic and Neural Mechanisms that Inhibit Drosophila from Mating with Other Species. Cell 154: 89-102. PubMed ID: 23810192

    Mellert, D. J., Robinett, C. C. and Baker, B. S. (2012). doublesex Functions Early and Late in Gustatory Sense Organ Development. PLoS One 7: e51489. PubMed ID: 23240029

  • Keith Baker Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond
    Padmanabha, D. and Baker, K. D. (2014). Drosophila gains traction as a repurposed tool to investigate metabolism. Trends Endocrinol Metab. PubMed ID: 24768030

    Li, Y., Padmanabha, D., Gentile, L. B., Dumur, C. I., Beckstead, R. B., Baker, K. D. (2013) HIF- and Non-HIF-Regulated Hypoxic Responses Require the Estrogen-Related Receptor in Drosophila melanogaster. PLoS Genet 9: e1003230. PubMed ID: 23382692

    Tennessen, J. M., Baker, K. D., Lam, G., Evans, J. and Thummel, C. S. (2011). The Drosophila estrogen-related receptor directs a metabolic switch that supports developmental growth. Cell Metab 13: 139-148. PubMed ID: 21284981

  • Nick Baker Albert Einstein College of Medicine, Bronx
    Kiparaki, M. and Baker, N. E. (2023). Ribosomal protein mutations and cell competition: autonomous and nonautonomous effects on a stress response. Genetics 224(3). PubMed ID: 37267156

    Kumar, A. and Baker, N. E. (2022). The CRL4 E3 ligase Mahjong/DCAF1 controls cell competition through the transcription factor Xrp1, independently of polarity genes. Development 149(22). PubMed ID: 36278853

    Ji, Z., Chuen, J., Kiparaki, M. and Baker, N. (2021). Cell competition removes segmental aneuploid cells from Drosophila imaginal disc-derived tissues based on ribosomal protein gene dose. Elife 10. PubMed ID: 33847264

    Quiquand, M., Rimesso, G., Qiao, N., Suo, S., Zhao, C., Slattery, M., White, K. P., Han, J. J. and Baker, N. E. (2021). New regulators of Drosophila eye development identified from temporal transcriptome changes. Genetics. PubMed ID: 33681970

    Blanco, J., Cooper, J. C. and Baker, N. E. (2020). Roles of C/EBP class bZip proteins in the growth and cell competition of Rp ('Minute') mutants in Drosophila. Elife 9. PubMed ID: 31909714

    Ji, Z., Kiparaki, M., Folgado, V., Kumar, A., Blanco, J., Rimesso, G., Chuen, J., Liu, Y., Zheng, D. and Baker, N. E. (2019). Drosophila RpS12 controls translation, growth, and cell competition through Xrp1. PLoS Genet 15(12): e1008513. PubMed ID: 31841522

    Li, K. and Baker, N. E. (2019). Transcriptional and post-transcriptional regulation of extra macrochaetae during Drosophila adult peripheral neurogenesis. Dev Biol. PubMed ID: 30771303

    Wang, L. H. and Baker, N. E. (2018). Spatial regulation of expanded transcription in the Drosophila wing imaginal disc. PLoS One 13(7): e0201317. PubMed ID: 30063727

    Li, K. and Baker, N. E. (2018). Regulation of the Drosophila ID protein Extra macrochaetae by proneural dimerization partners. Elife 7. PubMed ID: 29687780

    Kale, A., Ji, Z., Kiparaki, M., Blanco, J., Rimesso, G., Flibotte, S. and Baker, N. E. (2018). Ribosomal Protein S12e Has a Distinct Function in Cell Competition. Dev Cell 44(1): 42-55.e44. PubMed ID: 29316439

    Bhattacharya, A., Li, K., Quiquand, M., Rimesso, G. and Baker, N. E. (2017). The Notch pathway regulates the Second Mitotic Wave cell cycle independently of bHLH proteins. Dev Biol [Epub ahead of print]. PubMed ID: 28919436

  • Bill Ballard Biotechnology and Biomolecular Sciences. University of New South Wales, Sydney, Australia
    Towarnicki, S. G., Youngson, N. A., Corley, S. M., St John, J. C., Melvin, R. G., Turner, N., Morris, M. J. and Ballard, J. W. O. (2022). Ancestral dietary change alters the development of Drosophila larvae through MAPK signalling. Fly (Austin) 16(1): 299-311. PubMed ID: 35765944

    Towarnicki, S. G. and Ballard, J. W. O. (2018). Mitotype interacts With diet to influence longevity, fitness, and mitochondrial functions in adult female Drosophila. Front Genet 9: 593. PubMed ID: 30555517

    Aw, W. C., Towarnicki, S. G., Melvin, R. G., Youngson, N. A., Garvin, M. R., Hu, Y., Nielsen, S., Thomas, T., Pickford, R., Bustamante, S., Vila-Sanjurjo, A., Smyth, G. K. and Ballard, J. W. O. (2018). Genotype to phenotype: Diet-by-mitochondrial DNA haplotype interactions drive metabolic flexibility and organismal fitness. PLoS Genet 14(11): e1007735. PubMed ID: 30399141

    Aw, W. C., Garvin, M. R., Melvin, R. G. and Ballard, J. W. O. (2017). Sex-specific influences of mtDNA mitotype and diet on mitochondrial functions and physiological traits in Drosophila melanogaster. PLoS One 12(11): e0187554. PubMed ID: 29166659

    Aw, W. C. and Ballard, J. W. (2013). The effects of temperature and diet on age grading and population age structure determination in Drosophila. J Insect Physiol. PubMed ID: 23892055

    Pichaud, N., Messmer, M., Correa, C. C., Ballard, J. W. (2013) Diet influences the intake target and mitochondrial functions of Drosophila melanogaster males. Mitochondrion. PubMed ID: 23707480

    Wolff, J. N., Sutovsky, P. and Ballard, J. W. (2013). Mitochondrial DNA content of mature spermatozoa and oocytes in the genetic model Drosophila. Cell Tissue Res. PubMed ID: 23686567

    Wolff, J. N., Nafisinia, M., Sutovsky, P. and Ballard, J. W. (2013). Paternal transmission of mitochondrial DNA as an integral part of mitochondrial inheritance in metapopulations of Drosophila simulans. Heredity (Edinb) 110: 57-62. PubMed ID: 23010820

    Aw, W. C., Dowell, F. E. and Ballard, J. W. (2012). Using near-infrared spectroscopy to resolve the species, gender, age, and the presence of Wolbachia infection in laboratory-reared Drosophila. G3 (Bethesda) 2: 1057-1065. PubMed ID: 22973543

  • Swati Banerjee Cellular and Integrative Physiology, University of Texas, San Antonio
    Xie, J., Chen, S., Bopassa, J. C. and Banerjee, S. (2021). Drosophila tubulin polymerization promoting protein mutants reveal pathological correlates relevant to human Parkinson's disease. Sci Rep 11(1): 13614. PubMed ID: 34193896

    Shi, Q., Lin, Y. Q., Saliba, A., Xie, J., Neely, G. G. and Banerjee, S. (2019). Tubulin Polymerization Promoting Protein, Ringmaker, and MAP1B Homolog Futsch Coordinate Microtubule Organization and Synaptic Growth. Front Cell Neurosci 13: 192. PubMed ID: 31156389

    Banerjee, S., Mino, R. E., Fisher, E. S. and Bhat, M. A. (2017). A versatile genetic tool to study midline glia function in the Drosophila CNS. Dev Biol 429(1): 35-43. PubMed ID: 28602954

    Subramanian, A., Siefert, M., Banerjee, S., Vishal, K., Bergmann, K. A., Curts, C. C. M., Dorr, M., Molina, C. and Fernandes, J. (2017). Remodeling of peripheral nerve ensheathment during the larval-to-adult transition in Drosophila. Dev Neurobiol 77(10): 1144-1160. PubMed ID: 28388016

  • Utpal Banerjee University of California, Los Angeles
    Evans, C. J., Liu, T., Girard, J. R. and Banerjee, U. (2022). Injury-induced inflammatory signaling and hematopoiesis in Drosophila. Proc Natl Acad Sci U S A 119(12): e2119109119. PubMed ID: 35286208

    Spratford, C. M., Goins, L. M., Chi, F., Girard, J. R., Macias, S. N., Ho, V. W. and Banerjee, U. (2021). Intermediate progenitor cells provide a transition between hematopoietic progenitors and their differentiated descendants. Development 148(24). PubMed ID: 34918741

    Girard, J. R., Goins, L. M., Vuu, D. M., Sharpley, M. S., Spratford, C. M., Mantri, S. R. and Banerjee, U. (2021). Paths and pathways that generate cell-type heterogeneity and developmental progression in hematopoiesis. Elife 10. PubMed ID: 34713801

    Cho, B., Spratford, C. M., Yoon, S., Cha, N., Banerjee, U. and Shim, J. (2018). Systemic control of immune cell development by integrated carbon dioxide and hypoxia chemosensation in Drosophila. Nat Commun 9(1): 2679. PubMed ID: 29992947

    Wang, C. W., Purkayastha, A., Jones, K. T., Thaker, S. K. and Banerjee, U. (2016). In vivo genetic dissection of tumor growth and the Warburg effect. Elife 5. PubMed ID: 27585295
    Mondal, B. C., Shim, J., Evans, C. J. and Banerjee, U. (2014). Pvr expression regulators in equilibrium signal control and maintenance of Drosophila blood progenitors. Elife: e03626. PubMed ID: 25201876

    Evans, C. J., Liu, T. and Banerjee, U. (2014). Drosophila hematopoiesis: markers and methods for molecular genetic analysis. Methods. PubMed ID: 24613936

    Shim, J., Mukherjee, T., Mondal, B. C., Liu, T., Young, G. C., Wijewarnasuriya, D. P. and Banerjee, U. (2013). Olfactory control of blood progenitor maintenance. Cell 155: 1141-1153. PubMed ID: 24267893

    Shim, J., Gururaja-Rao, S. and Banerjee, U. (2013). Nutritional regulation of stem and progenitor cells in Drosophila. Development 140: 4647-4656. PubMed ID: 24255094

    Honti, V., Cinege, G., Csordas, G., Kurucz, E., Zsamboki, J., Evans, C. J., Banerjee, U. and Ando, I. (2013). Variation of NimC1 expression in Drosophila stocks and transgenic strains. Fly (Austin) 7. PubMed ID: 23899817

    Grigorian, M., Liu, T., Banerjee, U., Hartenstein, V. (2013) The proteoglycan Trol controls the architecture of the extracellular matrix and balances proliferation and differentiation of blood progenitors in the Drosophila lymph gland. Dev Biol. PubMed ID: 23510717

  • Erdem Bangi Department of Biological Science at Florida State University, Tallahassee,
    Datta, I. and Bangi, E. (2023). Senescent cells and macrophages cooperate through a multi-kinase signaling network to promote intestinal transformation in Drosophila. bioRxiv. PubMed ID: 37292988

    Datta, I. and Bangi, E. (2023). Senescent cells and macrophages cooperate through a multi-kinase signaling network to promote intestinal transformation in Drosophila. bioRxiv. PubMed ID: 37292988 <><>Datta, I. and Bangi, E. (2023). Senescent cells and macrophages cooperate through a multi-kinase signaling network to promote intestinal transformation in Drosophila. bioRxiv. PubMed ID: 37292988

    Datta, I., Vassel, T., Linkous, B., Odum, T., Drew, C., Taylor, A. and Bangi, E. (2023). A targeted genetic modifier screen in Drosophila uncovers vulnerabilities in a genetically complex model of colon cancer. G3 (Bethesda). PubMed ID: 36880303

    Teague, A. G., Quintero, M., Karimi Dermani, F., Cagan, R. L. and Bangi, E. (2023). A polycistronic transgene design for combinatorial genetic perturbations from a single transcript in Drosophila. PLoS Genet 19(6): e1010792. PubMed ID: 37267433

    Datta, I., Vassel, T., Linkous, B., Odum, T., Drew, C., Taylor, A. and Bangi, E. (2023). A targeted genetic modifier screen in Drosophila uncovers vulnerabilities in a genetically complex model of colon cancer. G3 (Bethesda) 13(5). PubMed ID: 36880303

    Bangi, E., Smibert, P., Uzilov, A. V., Teague, A. G., Gopinath, S., Antipin, Y., Chen, R., Hecht, C., Gruszczynski, N., Yon, W. J., Malyshev, D., Laspina, D., Selkridge, I., Wang, H., Gomez, J., Mascarenhas, J., Moe, A. S., Lau, C. Y., Taik, P., Pandya, C., Sung, M., Kim, S., Yum, K., Sebra, R., Donovan, M., Misiukiewicz, K., Ang, C., Schadt, E. E., Posner, M. R. and Cagan, R. L. (2021). A Drosophila platform identifies a novel, personalized therapy for a patient with adenoid cystic carcinoma. iScience 24(3): 102212. PubMed ID: 33733072

  • Antonio Baonza Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid
    Velarde, S. B., Quevedo, A., Estella, C. and Baonza, A. (2021). Dpp and Hedgehog promote the glial response to neuronal apoptosis in the developing Drosophila visual system. PLoS Biol 19(8): e3001367. PubMed ID: 34379617

    Ahmed-de-Prado, S., Diaz-Garcia, S. and Baonza, A. (2018). JNK and JAK/STAT signalling are required for inducing loss of cell fate specification during imaginal wing discs regeneration in Drosophila melanogaster. Dev Biol. PubMed ID: 29870691

    Diaz-Garcia, S., Ahmed, S. and Baonza, A. (2016). Analysis of the function of apoptosis during imaginal wing disc regeneration in Drosophila melanogaster. PLoS One 11(11): e0165554. PubMed ID: 27893747

    Andrade-Zapata, I. and Baonza, A. (2014). The bHLH Factors Extramacrochaetae and Daughterless Control Cell Cycle in Drosophila Imaginal Discs through the Transcriptional Regulation of the cdc25 Phosphatase string. PLoS Genet 10: e1004233. PubMed ID: 24651265

    Diaz-Garcia, S. and Baonza, A. (2013). Pattern reorganization occurs independently of cell division during Drosophila wing disc regeneration in situ. Proc Natl Acad Sci U S A. PubMed ID: 23878228

    San Juan, B. P., Andrade-Zapata, I. and Baonza, A. (2012). The bHLH factors Dpn and members of the E(spl) complex mediate the function of Notch signalling regulating cell proliferation during wing disc development. Biol Open 1: 667-676. PubMed ID: 23213460

    San-Juan, B. P. and Baonza, A. (2011). The bHLH factor deadpan is a direct target of Notch signaling and regulates neuroblast self-renewal in Drosophila. Dev. Biol. 352: 70-82. PubMed ID: 21262215

    Rodriguez, I., Baena-Lopez, L. A. and Baonza, A. (2008). Upregulation of glypicans in Hippo mutants alters the coordinated activity of morphogens. Fly (Austin) 2: 320-322. PubMed ID: 19077544

  • Thomas Baranski School of Medicine, Washington University in St. Louis
    Musselman, L. P., Fink, J. L. and Baranski, T. J. (2019). Similar effects of high-fructose and high-glucose feeding in a Drosophila model of obesity and diabetes. PLoS One 14(5): e0217096. PubMed ID: 31091299

    Musselman, L. P., Fink, J. L., Maier, E. J., Gatto, J. A., Brent, M. R. and Baranski, T. J. (2018). Seven-up is a novel regulator of insulin signaling. Genetics [Epub ahead of print]. PubMed ID: 29487137

    Musselman, L. P., Fink, J. L., Grant, A. R., Gatto, J. A., Tuthill, B. F., and Baranski, T. J. (2017). The relationship between immunity and metabolism in Drosophila diet-induced insulin resistance. Mol Cell Biol [Epub ahead of print]. PubMed ID: 29084810

    Musselman, L. P., Fink, J. L. and Baranski, T. J. (2016). CoA protects against the deleterious effects of caloric overload in Drosophila. J Lipid Res. PubMed ID: 26805007

    Nichols, A. S., Floyd, D. H., Bruinsma, S. P., Narzinski, K., Baranski, T. J. (2013) Frizzled receptors signal through G proteins. Cell Signal. PubMed ID: 23524329

    Musselman, L. P., Fink, J. L., Ramachandran, P. V., Patterson, B. W., Okunade, A. L., Maier, E., Brent, M. R., Turk, J. and Baranski, T. J. (2013). Role of fat body lipogenesis in protection against the effects of caloric overload in Drosophila. J Biol Chem. PubMed ID: 23355467

    Na, J., Musselman, L. P., Pendse, J., Baranski, T. J., Bodmer, R., Ocorr, K. and Cagan, R. (2013). A Drosophila model of high sugar diet-induced cardiomyopathy. PLoS Genet 9: e1003175. PubMed ID: 23326243

    Musselman, L. P., Fink, J. L., Narzinski, K., Ramachandran, P. V., Hathiramani, S. S., Cagan, R. L. and Baranski, T. J. (2011). A high-sugar diet produces obesity and insulin resistance in wild-type Drosophila. Dis Model Mech 4: 842-849. PubMed ID: 21719444

  • Daniel Barbash Department of Molecular Biology and Genetics, Cornell University
    McGurk, M. P., Dion-Cote, A. M. and Barbash, D. A. (2021). Rapid evolution at the Drosophila telomere: transposable element dynamics at an intrinsically unstable locus. Genetics 217(2). PubMed ID: 33724410

    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

    McGurk, M. P. and Barbash, D. A. (2018). Double insertion of transposable elements provides a substrate for the evolution of satellite DNA. Genome Res 28(5): 714-725. PubMed ID: 29588362

    Wei, K. H., Lower, S. E., Caldas, I. V., Sless, T. J., Barbash, D. A. and Clark, A. G. (2018). Variable rates of simple satellite gains across the Drosophila phylogeny. Mol Biol Evol [Epub ahead of print]. PubMed ID: 29361128

    Wei, K. H., Reddy, H. M., Rathnam, C., Lee, J., Lin, D., Ji, S., Mason, J. M., Clark, A. G. and Barbash, D. A. (2017). A Pooled Sequencing Approach Identifies a Candidate Meiotic Driver in Drosophila. Genetics. PubMed ID: 28258181

    Flores, H. A., Bubnell, J. E., Aquadro, C. F. and Barbash, D. A. (2015). The Drosophila bag of marbles gene interacts genetically with Wolbachia and shows female-specific effects of divergence. PLoS Genet 11: e1005453. PubMed ID: 26291077

    Wei, K. H., Grenier, J. K., Barbash, D. A. and Clark, A. G. (2014). Correlated variation and population differentiation in satellite DNA abundance among lines of Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 25512552

    Cuykendall, T. N., Satyaki, P., Ji, S., Clay, D. M., Edelman, N. B., Kimchy, A., Li, L. H., Nuzzo, E. A., Parekh, N., Park, S. and Barbash, D. A. (2014). A Screen for F1 Hybrid Male Rescue Reveals No Major-Effect Hybrid Lethality Loci in the Drosophila melanogaster Autosomal Genome. G3 (Bethesda). PubMed ID: 25352540

    Wei, K. H., Clark, A. G. and Barbash, D. A. (2014). Limited gene misregulation is exacerbated by allele-specific up-regulation in lethal hybrids between Drosophila melanogaster and D. simulans. Mol Biol Evol. PubMed ID: 24723419

    Satyaki, P. R., Cuykendall, T. N., Wei, K. H., Brideau, N. J., Kwak, H., Aruna, S., Ferree, P. M., Ji, S. and Barbash, D. A. (2014). The hmr and lhr hybrid incompatibility genes suppress a broad range of heterochromatic repeats. PLoS Genet 10: e1004240. PubMed ID: 24651406

    Ferree, P. M., Gomez, K., Rominger, P., Howard, D., Kornfeld, H. and Barbash, D. A. (2014). Heterochromatin Position Effects on Circularized Sex Chromosomes Cause Filicidal Embryonic Lethality in Drosophila melanogaster. Genetics. PubMed ID: 24478337

  • Scott Barbee Department of Biological Sciences, University of Denver
    Wilkinson, E. C., Starke, E. L. and Barbee, S. A. (2021). Vps54 Regulates Lifespan and Locomotor Behavior in Adult Drosophila melanogaster. Front Genet 12: 762012. PubMed ID: 34712272

    Patel, P. H., Wilkinson, E. C., Starke, E. L., McGimsey, M. R., Blankenship, J. T. and Barbee, S. A. (2020). Vps54 regulates Drosophila neuromuscular junction development and interacts genetically with Rab7 to control composition of the postsynaptic density. Biol Open 9(8). PubMed ID: 32747448

    Nesler, K. R., Starke, E. L., Boin, N. G., Ritz, M. and Barbee, S. A. (2016). Presynaptic CamKII regulates activity-dependent axon terminal growth. Mol Cell Neurosci 76: 33-41. PubMed ID: 27567686

    Patel, P. H., Barbee, S. A. and Blankenship, J. T. (2016). GW-Bodies and P-Bodies Constitute Two Separate Pools of Sequestered Non-Translating RNAs. PLoS One 11: e0150291. PubMed ID: 26930655

    Nesler, K. R., Sand, R. I., Symmes, B. A., Pradhan, S. J., Boin, N. G., Laun, A. E. and Barbee, S. A. (2013). The miRNA Pathway Controls Rapid Changes in Activity-Dependent Synaptic Structure at the Drosophila melanogaster Neuromuscular Junction. PLoS One 8: e68385. PubMed ID: 23844193

    Pradhan, S. J., Nesler, K. R., Rosen, S. F., Kato, Y., Nakamura, A., Ramaswami, M. and Barbee, S. A. (2012). The conserved P body component HPat/Pat1 negatively regulates synaptic terminal growth at the larval Drosophila neuromuscular junction. J. Cell Sci. [Epub ahead of print]. PubMed ID: 23097047

  • Allison Bardin Stem Cells and Tissue Homeostasis, Genetics and Developmental Biology, Institut Curie, Paris
    Siudeja, K., van den Beek, M., Riddiford, N., Boumard, B., Wurmser, A., Stefanutti, M., Lameiras, S. and Bardin, A. J. (2021). Unraveling the features of somatic transposition in the Drosophila intestine. Embo j: e106388. PubMed ID: 33634906

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

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

    Salle, J., Gervais, L., Boumard, B., Stefanutti, M., Siudeja, K. and Bardin, A. J. (2017). Intrinsic regulation of enteroendocrine fate by Numb. EMBO J [Epub ahead of print]. PubMed ID: 28533229

    Siudeja, K., Nassari, S., Gervais, L., Skorski, P., Lameiras, S., Stolfa, D., Zande, M., Bernard, V., Rio Frio, T. and Bardin, A. J. (2015). Frequent somatic mutation in adult intestinal stem cells drives neoplasia and genetic mosaicism during aging. Cell Stem Cell 17: 663-674. PubMed ID: 26607382

    Mathieu, J., Cauvin, C., Moch, C., Radford, S. J., Sampaio, P., Perdigoto, C. N., Schweisguth, F., Bardin, A. J., Sunkel, C. E., McKim, K., Echard, A. and Huynh, J. R. (2013). Aurora B and cyclin B have opposite effects on the timing of cytokinesis abscission in Drosophila germ cells and in vertebrate somatic cells. Dev Cell 26: 250-265. PubMed ID: 23948252

    de Navascues, J., Perdigoto, C. N., Bian, Y., Schneider, M. H., Bardin, A. J., Martinez-Arias, A. and Simons, B. D. (2012). Drosophila midgut homeostasis involves neutral competition between symmetrically dividing intestinal stem cells. EMBO J 31: 2473-2485. PubMed ID: 22522699

  • E. M. Baricheva Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk
    Fedorova, S., Dorogova, N. V., Karagodin, D. A., Oshchepkov, D. Y., Brusentsov, II, Klimova, N. V. and Baricheva, E. M. (2023). The complex role of transcription factor GAGA in germline death during Drosophila spermatogenesis: transcriptomic and bioinformatic analyses. PeerJ 11: e14063. PubMed ID: 36643636

    Fedorova, E. V., Dorogova, N. V., Bolobolova, E. U., Fedorova, S. A., Karagodin, D. A., Ogienko, A. A., Khruscheva, A. S. and Baricheva, E. M. (2018). GAGA protein is required for multiple aspects of Drosophila oogenesis and female fertility. Genesis: e23269. PubMed ID: 30537428

    Dorogova, N. V., Fedorova, E. V., Bolobolova, E., Ogienko, A. A. and Baricheva, E. M. (2014). GAGA protein is essential for male germ cell development in Drosophila. Genesis [Epub ahead of print]. PubMed ID: 24817547

    Karagodin, D. A., Omelina, E. S., Fedorova, E. V., Baricheva, E. M. (2013) Identification of functionally significant elements in the second intron of the Drosophila melanogaster Trithorax-like gene. Gene. PubMed ID: 23481306

    Ogienko, A. A., Karagodin, D. A., Lashina, V. V., Baiborodin, S. I., Omelina, E. S., Baricheva, E. M. (2013) Capping protein beta is required for actin cytoskeleton organisation and cell migration during Drosophila oogenesis. Cell Biol Int 37: 149-159. PubMed ID: 23339103

    Omelina, E. S., Baricheva, E. M., Oshchepkov, D. Y. and Merkulova, T. I. (2011). Analysis and recognition of the GAGA transcription factor binding sites in Drosophila genes. Comput Biol Chem 35: 363-370. PubMed ID: 22099633

    Omelina, E. S., Pavlova, N. V., Ogienko, A. A. and Baricheva, E. M. (2011). The GAGA protein is essential for dorsal appendage formation during Drosophila melanogaster oogenesis. Dokl Biochem Biophys 436: 32-34. PubMed ID: 21369899

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

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

    Averbukh, I., Ben-Zvi, D., Mishra, S. and Barkai, N. (2014). Scaling morphogen gradients during tissue growth by a cell division rule. Development 141: 2150-2156. PubMed ID: 24803660

    Tamari, Z., Barkai, N. (2012) Improved readout precision of the Bicoid morphogen gradient by early decoding. J Biol Phys 38: 317-329. Pubmed ID: 23449375

    Haskel-Ittah, M., Ben-Zvi, D., Branski-Arieli, M., Schejter, E. D., Shilo, B. Z. and Barkai, N. (2012). Self-organized shuttling: generating sharp dorsoventral polarity in the early Drosophila embryo. Cell 150: 1016-1028. PubMed ID: 22939625

    Barad, O., Hornstein, E. and Barkai, N. (2011). Robust selection of sensory organ precursors by the Notch-Delta pathway. Curr Opin Cell Biol 23: 663-667. PubMed ID: 21963301

  • Gilad Barnea Department of Neuroscience, Brown University, Providence, RI
    Sorkac, A., Savva, Y. A., Savas, D., Talay, M. and Barnea, G. (2022). Circuit analysis reveals a neural pathway for light avoidance in Drosophila larvae. Nat Commun 13(1): 5274. PubMed ID: 36071059

    Snell, N. J., Fisher, J. D., Hartmann, G. G., Zolyomi, B., Talay, M. and Barnea, G. (2022). Complex representation of taste quality by second-order gustatory neurons in Drosophila. Curr Biol 32(17): 3758-3772. PubMed ID: 35973432

    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

  • Scott Barolo Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor
    Gurdziel, K., Lorberbaum, D. S., Udager, A. M., Song, J. Y., Richards, N., Parker, D. S., Johnson, L. A., Allen, B. L., Barolo, S. and Gumucio, D. L. (2015). Identification and validation of novel Hedgehog-responsive enhancers predicted by computational analysis of Ci/Gli binding site density. PLoS One 10: e0145225. PubMed ID: 26710299

    Lorberbaum, D. S. and Barolo, S. (2015). Enhancers: holding out for the right promoter. Curr Biol 25: R290-293. PubMed ID: 25829016

    Ramos, A. I. and Barolo, S. (2013). Low-affinity transcription factor binding sites shape morphogen responses and enhancer evolution. Philos Trans R Soc Lond B Biol Sci 368: 20130018. PubMed ID: 24218631

    White, M. A., Parker, D. S., Barolo, S. and Cohen, B. A. (2012). A model of spatially restricted transcription in opposing gradients of activators and repressors. Mol Syst Biol 8: 614. PubMed ID: 23010997

    Evans, N. C., Swanson, C. I. and Barolo, S. (2012). Sparkling insights into enhancer structure, function, and evolution. Curr Top Dev Biol 98: 97-120. PubMed ID: 22305160

    Swanson, C. I., Schwimmer, D. B. and Barolo, S. (2011). Rapid evolutionary rewiring of a structurally constrained eye enhancer. Curr Biol 21: 1186-1196. PubMed ID: 21737276

  • Martin Baron Faculty of Life Sciences, Manchester, England
    Shimizu, H., Wilkin, M. B., Woodcock, S. A., Bonfini, A., Hung, Y., Mazaleyrat, S. and Baron, M. (2017). The Drosophila ZO-1 protein Polychaetoid suppresses Deltex-regulated Notch activity to modulate germline stem cell niche formation. Open Biol 7(4). PubMed ID: 28424321

    Bonfini, A., Wilkin, M.B. and Baron, M. (2015). Reversible regulation of stem cell niche size associated with dietary control of Notch signalling. BMC Dev Biol [Epub ahead of print]. PubMed ID: 25637382

    Shimizu, H., Woodcock, S. A., Wilkin, M. B., Trubenova, B., Monk, N. A. and Baron, M. (2014). Compensatory Flux Changes within an Endocytic Trafficking Network Maintain Thermal Robustness of Notch Signaling. Cell 157: 1160-1174. PubMed ID: 24855951

    Yamada, K., Fuwa, T. J., Ayukawa, T., Tanaka, T., Nakamura, A., Wilkin, M. B., Baron, M. and Matsuno, K. (2011). Roles of Drosophila deltex in Notch receptor endocytic trafficking and activation. Genes Cells 16: 261-272. PubMed ID: 21299753

    Djiane, A., Shimizu, H., Wilkin, M., Mazleyrat, S., Jennings, M. D., Avis, J., Bray, S. and Baron, M. (2011). Su(dx) E3 ubiquitin ligase-dependent and -independent functions of polychaetoid, the Drosophila ZO-1 homologue. J Cell Biol 192: 189-200. PubMed ID: 21200027

  • Rosa Barrio Center for Cooperative Research in Biosciences, Bilbao, Spain
    Gradilla, A. C., Gonzalez, E., Seijo, I., Andres, G., Bischoff, M., Gonzalez-Mendez, L., Sanchez, V., Callejo, A., Ibanez, C., Guerra, M., Ortigao-Farias, J. R., Sutherland, J. D., Gonzalez, M., Barrio, R., Falcon-Perez, J. M. and Guerrero, I. (2014). Exosomes as Hedgehog carriers in cytoneme-mediated transport and secretion. Nat Commun 5: 5649. PubMed ID: 25472772

    Herboso, L., Oliveira, M.M., Talamillo, A., Pérez, C., González, M., Martín, D., Sutherland, J.D., Shingleton, A.W., Mirth, C.K. and Barrio, R. (2015). Ecdysone promotes growth of imaginal discs through the regulation of Thor in D. melanogaster.Sci Rep 5: 12383. PubMed ID: 26198204

    Cantera, R. and Barrio, R. (2014). Do the Genes of the Innate Immune Response Contribute to Neuroprotection in Drosophila ? J Innate Immun. PubMed ID: 25115549

    Cantera, R., Ferreiro, M. J., Aransay, A. M. and Barrio, R. (2014). Global Gene Expression Shift during the Transition from Early Neural Development to Late Neuronal Differentiation in Drosophila melanogaster. PLoS One 9: e97703. PubMed ID: 24830291

    Talamillo, A., Herboso, L., Pirone, L., Perez, C., Gonzalez, M., Sanchez, J., Mayor, U., Lopitz-Otsoa, F., Rodriguez, M. S., Sutherland, J. D., Barrio, R. (2013) Scavenger Receptors Mediate the Role of SUMO and Ftz-f1 in Drosophila Steroidogenesis. PLoS Genet 9: e1003473. PubMed ID: 23637637

    Ferreiro, M. J., Rodriguez-Ezpeleta, N., Perez, C., Hackenberg, M., Aransay, A. M., Barrio, R. and Cantera, R. (2012). Whole transcriptome analysis of a reversible neurodegenerative process in Drosophila reveals potential neuroprotective genes. BMC Genomics 13: 483. PubMed ID: 22978642

    Gonzalez, M., Martin-Ruiz, I., Jimenez, S., Pirone, L., Barrio, R. and Sutherland, J. D. (2011). Generation of stable Drosophila cell lines using multicistronic vectors. Sci Rep 1: 75. PubMed ID: 22355594

    Herboso, L., Talamillo, A., Perez, C. and Barrio, R. (2011). Expression of the Scavenger Receptor Class B type I (SR-BI) family in Drosophila melanogaster. Int J Dev Biol 55: 603-611. PubMed ID: 21948708

  • Claudia Barros Neural Stem Cell Research, University of Plymouth, United Kingdom
    Gil-Ranedo, J., Gonzaga, E., Jaworek, K. J., Berger, C., Bossing, T. and Barros, C. S. (2019). STRIPAK Members Orchestrate Hippo and Insulin Receptor Signaling to Promote Neural Stem Cell Reactivation. Cell Rep 27(10): 2921-2933 e2925. PubMed ID: 31167138

    Raghu, S. V., Mohammad, F., Chua, J. Y., Lam, J. S. W., Loberas, M., Sahani, S., Barros, C. S. and Claridge-Chang, A. (2018). A zinc-finger fusion protein refines Gal4-defined neural circuits. Mol Brain 11(1): 46. PubMed ID: 30126464

    Ding, R., Weynans, K., Bossing, T., Barros, C. S. and Berger, C. (2016). The Hippo signalling pathway maintains quiescence in Drosophila neural stem cells. Nat Commun 7: 10510. PubMed ID: 26821647

  • David Bartel Whitehead Institute, Cambridge
    Kingston, E. R., Blodgett, L. W. and Bartel, D. P. (2022). Endogenous transcripts direct microRNA degradation in Drosophila, and this targeted degradation is required for proper embryonic development. Mol Cell. PubMed ID: 36150386

    Kingston, E. R. and Bartel, D. P. (2021). Ago2 protects Drosophila siRNAs and microRNAs from target-directed degradation, even in the absence of 2'-O-methylation. Rna. PubMed ID: 33853897

    Kwasnieski, J. C., Orr-Weaver, T. L. and Bartel, D. P. (2019). Early genome activation in Drosophila is extensive with an initial tendency for aborted transcripts and retained introns. Genome Res. PubMed ID: 31235656

    Agarwal, V., Subtelny, A. O., Thiru, P., Ulitsky, I. and Bartel, D. P. (2018). Predicting microRNA targeting efficacy in Drosophila. Genome Biol 19(1): 152. PubMed ID: 30286781

    Rissland, O. S., Subtelny, A. O., Wang, M., Lugowski, A., Nicholson, B., Laver, J. D., Sidhu, S. S., Smibert, C. A., Lipshitz, H. D. and Bartel, D. P. (2017). The influence of microRNAs and poly(A) tail length on endogenous mRNA-protein complexes. Genome Biol 18(1): 211. PubMed ID: 27474798

    Kronja, I., Yuan, B., Eichhorn, S. W., Dzeyk, K., Krijgsveld, J., Bartel, D. P. and Orr-Weaver, T. L. (2014). Widespread changes in the posttranscriptional landscape at the Drosophila oocyte-to-embryo transition. Cell Rep 7(5): 1495-1508. PubMed ID: 24882012

  • Greg Bashaw Department of Neuroscience, University of Pennsylvania
    Zang, Y., Chaudhari, K. and Bashaw, G. J. (2022). Tace/ADAM17 is a bi-directional regulator of axon guidance that coordinates distinct Frazzled and Dcc receptor signaling outputs. Cell Rep 41(10): 111785. PubMed ID: 36476876

    Russell, S. A., Laws, K. M. and Bashaw, G. J. (2021). Frazzled/Dcc acts independently of Netrin to promote germline survival during Drosophila oogenesis. Development 148(24). PubMed ID: 34910816

    Daiber, T., VanderZwan-Butler, C. J., Bashaw, G. J. and Evans, T. A. (2021). Conserved and divergent aspects of Robo receptor signaling and regulation between Drosophila Robo1 and C. elegans SAX-3. Genetics 217(3). PubMed ID: 33789352

    Chaudhari, K., Gorla, M., Chang, C., Kania, A. and Bashaw, G. J. (2021). Robo recruitment of the Wave regulatory complex plays an essential and conserved role in midline repulsion. Elife 10. PubMed ID: 33843588

    Arbeille, E. and Bashaw, G. J. (2018). Brain Tumor promotes axon growth across the midline through interactions with the microtubule stabilizing protein Apc2. PLoS Genet 14(4): e1007314. PubMed ID: 29617376

    Santiago, C. and Bashaw, G.J. (2017). Islet coordinately regulates motor axon guidance and dendrite targeting through the Frazzled/DCC receptor. Cell Rep 18: 1646-1659. PubMed ID: 28199838

    Hernandez-Fleming, M., Rohrbach, E. W. and Bashaw, G. J. (2017). Sema-1a Reverse Signaling Promotes Midline Crossing in Response to Secreted Semaphorins. Cell Rep 18(1): 174-184. PubMed ID: 28052247

    Chance, R. K. and Bashaw, G. J. (2015). Slit-dependent endocytic trafficking of the Robo receptor is required for Son of Sevenless recruitment and midline axon repulsion. PLoS Genet 11: e1005402. PubMed ID: 26335920

    Evans, T.A., Santiago, C., Arbeille, E. and Bashaw, G.J. (2015). Robo2 acts in trans to inhibit Slit-Robo1 repulsion in pre-crossing commissural axons. Elife 4. PubMed ID: 26186094

    Santiago, C., Labrador, J. P. and Bashaw, G. J. (2014). The Homeodomain Transcription Factor Hb9 Controls Axon Guidance in Drosophila through the Regulation of Robo Receptors. Cell Rep. PubMed ID: 24685136

  • Arash Bashirullah University of Wisconsin Laboratory of Genetics, Madison
    Neuman, S. D., Lee, A. R., Selegue, J. E., Cavanagh, A. T. and Bashirullah, A. (2021). A novel function for Rab1 and Rab11 during secretory granule maturation. J Cell Sci 134(15). PubMed ID: 34342349

    Neuman, S. D., Jorgensen, J. R., Cavanagh, A. T., Smyth, J. T., Selegue, J. E., Emr, S. D. and Bashirullah, A. (2022). The Hob proteins are novel and conserved lipid-binding proteins at ER-PM contact sites. J Cell Sci 135(5). PubMed ID: 34415038

    Neuman, S. D. and Bashirullah, A. (2018). Hobbit regulates intracellular trafficking to drive insulin-dependent growth during Drosophila development. Development 145(11). PubMed ID: 29891564

    Kang, Y., Neuman, S. D. and Bashirullah, A. (2017). Tango7 regulates cortical activity of caspases during reaper-triggered changes in tissue elasticity. Nat Commun 8(1): 603. PubMed ID: 28928435

    Kang, Y., Marischuk, K., Castelvecchi, G.D. and Bashirullah, A. (2017). HDAC inhibitors disrupt programmed resistance to apoptosis during Drosophila development. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28455414

    Neuman, S. D., Ihry, R. J., Gruetzmacher, K. M. and Bashirullah, A. (2014). INO80-dependent regression of ecdysone-induced transcriptional responses regulates developmental timing in Drosophila. Dev Biol. PubMed ID: 24468295

    Ihry, R. J. and Bashirullah, A. (2013). Genetic Control of Specificity to Steroid-Triggered Responses in Drosophila. Genetics. PubMed ID: 24374353

    Kang, Y. and Bashirullah, A. (2013). A steroid-controlled global switch in sensitivity to apoptosis during Drosophila development. Dev Biol. PubMed ID: 24333635

    Sapiro, A. L., Ihry, R. J., Buhr, D. L., Konieczko, K. M., Ives, S. M., Engstrom, A. K., Wleklinski, N. P., Kopish, K. J. and Bashirullah, A. (2013). Rapid recombination mapping for high-throughput genetic screens in Drosophila. G3 (Bethesda) 3: 2313-2319. PubMed ID: 24170736

    Ihry, R. J., Sapiro, A. L. and Bashirullah, A. (2012). Translational control by the DEAD Box RNA helicase belle regulates ecdysone-triggered transcriptional cascades. PLoS Genet 8: e1003085. PubMed ID: 23209440

  • Konrad Basler Institute of Molecular Life Sciences, University of Zurich
    Cannac, F., Qi, C., Falschlunger, J., Hausmann, G., Basler, K. and Korkhov, V. M. (2020). Cryo-EM structure of the Hedgehog release protein Dispatched. Sci Adv 6(16): eaay7928. PubMed ID: 32494603

    Bosch, P. S., Pepperl, J. and Basler, K. (2020). Anchor Away - A Fast, Reliable and Reversible Technique To Inhibit Proteins in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32217630

    Shokri, L., Inukai, S., Hafner, A., Weinand, K., Hens, K., Vedenko, A., Gisselbrecht, S. S., Dainese, R., Bischof, J., Furger, E., Feuz, J. D., Basler, K., Deplancke, B. and Bulyk, M. L. (2019). A comprehensive Drosophila melanogaster transcription factor interactome. Cell Rep 27(3): 955-970. PubMed ID: 30995488

    Germani, F., Hain, D., Sternlicht, D., Moreno, E. and Basler, K. (2018). The Toll pathway inhibits tissue growth and regulates cell fitness in an infection-dependent manner. Elife 7. PubMed ID: 30451683

    Baillon, L., Germani, F., Rockel, C., Hilchenbach, J. and Basler, K. (2018). Xrp1 is a transcription factor required for cell competition-driven elimination of loser cells. Sci Rep 8(1): 17712. PubMed ID: 30531963

    Bischof, J., Duffraisse, M., Furger, E., Ajuria, L., Giraud, G., Vanderperre, S., Paul, R., Bjorklund, M., Ahr, D., Ahmed, A. W., Spinelli, L., Brun, C., Basler, K. and Merabet, S. (2018). Generation of a versatile BiFC ORFeome library for analyzing protein-protein interactions in live Drosophila. Elife 7. PubMed ID: 30247122

    Sanchez Bosch, P., Ziukaite, R., Alexandre, C., Basler, K. and Vincent, J. B. (2017). Dpp controls growth and patterning in Drosophila wing precursors through distinct modes of action. Elife 6. PubMed ID: 28675374

    Franz, A., Shlyueva, D., Brunner, E., Stark, A. and Basler, K. (2017). Probing the canonicity of the Wnt/Wingless signaling pathway. PLoS Genet 13(4): e1006700. PubMed ID: 28369070

    Jenny, F. H. and Basler, K. (2016). Drosophila DDX3/Belle exerts its function outside of the Wnt/Wingless signaling pathway. PLoS One 11(12): e0166862. PubMed ID: 28030561

    Restrepo, S. and Basler, K. (2016). Drosophila wing imaginal discs respond to mechanical injury via slow InsP3R-mediated intercellular calcium waves. Nat Commun 7: 12450. PubMed ID: 27503836

    Heller, D., Hoppe, A., Restrepo, S., Gatti, L., Tournier, A. L., Tapon, N., Basler, K. and Mao, Y. (2016). EpiTools: An open-source image analysis toolkit for quantifying epithelial growth dynamics. Dev Cell 36: 103-116. PubMed ID: 26766446

    Marty, F., Rockel-Bauer, C., Simigdala, N., Brunner, E. and Basler, K. (2014). Large-scale imaginal disc sorting: A protocol for "omics"-approaches. Methods 68: 260-264. PubMed ID: 24736056

  • Renata Basto Institut Curie, Paris
    Goupil, A., Nano, M., Letort, G., Gemble, S., Edwards, F., Goundiam, O., Gogendeau, D., Pennetier, C. and Basto, R. (2020). Chromosomes function as a barrier to mitotic spindle bipolarity in polyploid cells. J Cell Biol 219(4). PubMed ID: 32328633

    Gambarotto, D., Pennetier, C., Ryniawec, J. M., Buster, D. W., Gogendeau, D., Goupil, A., Nano, M., Simon, A., Blanc, D., Racine, V., Kimata, Y., Rogers, G. C. and Basto, R. (2019). Plk4 regulates centriole asymmetry and spindle orientation in neural stem cells. Dev Cell. PubMed ID: 31130353

    Gogendeau, D., Siudeja, K., Gambarotto, D., Pennetier, C., Bardin, A. J. and Basto, R. (2015). Aneuploidy causes premature differentiation of neural and intestinal stem cells. Nat Commun 6: 8894. PubMed ID: 26573328

    Mendes Maia, T., Paul-Gilloteaux, P. and Basto, R. (2015). Quantitative analysis of flagellar proteins in Drosophila sperm tails. Methods Cell Biol 127: 263-278. PubMed ID: 25837396

    Sabino, D., Gogendeau, D., Gambarotto, D., Nano, M., Pennetier, C., Dingli, F., Arras, G., Loew, D. and Basto, R. (2015). Moesin is a major regulator of centrosome behavior in epithelial cells with extra centrosomes. Curr Biol 25: 879-889. PubMed ID: 25772448

    Mendes Maia, T., Gogendeau, D., Pennetier, C., Janke, C. and Basto, R. (2014). Bug22 influences cilium morphology and the post-translational modification of ciliary microtubules. Biol Open. PubMed ID: 24414207

    Rujano, M. A., Sanchez-Pulido, L., Pennetier, C., le Dez, G. and Basto, R. (2013). The microcephaly protein Asp regulates neuroepithelium morphogenesis by controlling the spatial distribution of myosin II. Nat Cell Biol. PubMed ID: 24142104

    Sabino, D., Brown, N. H. and Basto, R. (2011). Drosophila Ajuba is not an Aurora-A activator but is required to maintain Aurora-A at the centrosome. J Cell Sci 124: 1156-1166. PubMed ID: 21402878

  • Michael Bate Neuroscience, University of Cambridge
    Pulver, S. R., Bayley, T. G., Taylor, A. L., Berni, J., Bate, M. and Hedwig, B. (2015). Imaging fictive locomotor patterns in larval Drosophila. J Neurophysiol: jn 00731 02015. PubMed ID: 26311188

    Prieto-Godino, L. L., Diegelmann, S. and Bate, M. (2012). Embryonic origin of olfactory circuitry in Drosophila: contact and activity-mediated interactions pattern connectivity in the antennal lobe. PLoS Biol 10: e1001400. PubMed ID: 23055825

    Berni, J., Pulver, S. R., Griffith, L. C. and Bate, M. (2012). Autonomous circuitry for substrate exploration in freely moving Drosophila larvae. Curr Biol 22: 1861-1870. PubMed ID: 22940472

    Nair, A., Bate, M. and Pulver, S. R. (2010). Characterization of voltage-gated ionic currents in a peripheral sensory neuron in larval Drosophila. BMC Res Notes 3: 154. PubMed ID: 20525165

  • Jack Bateman Biology Department, Bowden College, Brunswick, Maine
    Vinsland, E., Baskaran, P., Mihaylov, S. R., Hobbs, C., Wood, H., Bouybayoune, I., Shah, K., Houart, C., Tee, A. R., Murn, J., Fernandes, C. and Bateman, J. M. (2021). The zinc finger/RING domain protein Unkempt regulates cognitive flexibility. Sci Rep 11(1): 16299. PubMed ID: 34381067

    Maierbrugger, K. T., Sousa-Nunes, R. and Bateman, J. M. (2020). The mTOR pathway component Unkempt regulates neural stem cell and neural progenitor cell cycle in the Drosophila central nervous system. Dev Biol. PubMed ID: 31978396

    Hunt, R. J., Granat, L., McElroy, G. S., Ranganathan, R., Chandel, N. S. and Bateman, J. M. (2019). Mitochondrial stress causes neuronal dysfunction via an ATF4-dependent increase in L-2-hydroxyglutarate. J Cell Biol. PubMed ID: 31645461

    King, T., Johnson, J. E. and Bateman, J. R. (2019). Position effects influence transvection in Drosophila melanogaster. Genetics. PubMed ID: 31611231

    Tian, K., Henderson, R. E., Parker, R., Brown, A., Johnson, J. E. and Bateman, J. R. (2019). Two modes of transvection at the eyes absent gene of Drosophila demonstrate plasticity in transcriptional regulatory interactions in cis and in trans. PLoS Genet 15(5): e1008152. PubMed ID: 31075100

    Blick, A. J., Mayer-Hirshfeld, I., Malibiran, B. R., Cooper, M. A., Martino, P. A., Johnson, J. E. and Bateman, J. R. (2016). The Capacity To Act in trans Varies Among Drosophila Enhancers. Genetics. PubMed ID: 26984057

    Bing, X., Rzezniczak, T. Z., Bateman, J. R. and Merritt, T. J. (2014). Transvection-based gene regulation in Drosophila is a complex and plastic trait. G3 (Bethesda) 4: 2175-2187. PubMed ID: 25213691

    Bateman, J. R., Palopoli, M. F., Dale, S. T., Stauffer, J. E., Shah, A. L., Johnson, J. E., Walsh, C. W., Flaten, H. and Parsons, C. M. (2013). Captured segment exchange: a strategy for custom engineering large genomic regions in Drosophila melanogaster. Genetics 193: 421-430. PubMed ID: 23150604

  • Joseph Bateman Wolfson Centre for Age-Related Diseases, King's College London
    Cagin, U., Duncan, O. F., Gatt, A. P., Dionne, M. S., Sweeney, S. T. and Bateman, J. M. (2015). Mitochondrial retrograde signaling regulates neuronal function. Proc Natl Acad Sci U S A 112: E6000-6009. PubMed ID: 26489648

    Avet-Rochex, A., Maierbrugger, K. T. and Bateman, J. M. (2014). Glial enriched gene expression profiling identifies novel factors regulating the proliferation of specific glial subtypes in the Drosophila brain. Gene Expr Patterns 16: 61-68. PubMed ID: 25217886

    Avet-Rochex, A., Carvajal, N., Christoforou, C. P., Yeung, K., Maierbrugger, K. T., Hobbs, C., Lalli, G., Cagin, U., Plachot, C., McNeill, H. and Bateman, J. M. (2014). Unkempt Is Negatively Regulated by mTOR and Uncouples Neuronal Differentiation from Growth Control. PLoS Genet 10: e1004624. PubMed ID: 25210733

    Avet-Rochex, A., Kaul, A. K., Gatt, A. P., McNeill, H. and Bateman, J. M. (2012). Concerted control of gliogenesis by InR/TOR and FGF signalling in the Drosophila post-embryonic brain. Development 139: 2763-2772. PubMed ID: 22745312

    Sun, F. F., Johnson, J. E., Zeidler, M. P. and Bateman, J. R. (2012). Simplified Insertion of Transgenes Onto Balancer Chromosomes via Recombinase-Mediated Cassette Exchange. G3 (Bethesda) 2: 551-553. PubMed ID: 22670225

    Bateman, J. R., Johnson, J. E. and Locke, M. N. (2012). Comparing enhancer action in cis and in trans. Genetics 191: 1143-1155. PubMed ID: 22649083

  • Philip Batterham Australian National University
    Perry, T., Chen W., Ghazali, R., Yang, Y. T., Christesen, D., Martelli, F., Lumb, C., Bao Luong, H. N., Mitchell, J., Holien, J. K., Parker, M. W., Sparks, T. C. and Batterham, P. (2021). Role of nicotinic acetylcholine receptor subunits in the mode of action of neonicotinoid, sulfoximine and spinosyn insecticides in Drosophila melanogaster. Insect Biochem Mol Biol 131: 103547. PubMed ID: 33548485

    Martelli, F., Zhongyuan, Z., Wang, J., Wong, C. O., Karagas, N. E., Roessner, U., Rupasinghe, T., Venkatachalam, K., Perry, T., Bellen, H. J. and Batterham, P. (2020). Low doses of the neonicotinoid insecticide imidacloprid induce ROS triggering neurological and metabolic impairments in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 32989137

    Denecke, S., Fusetto, R., Martelli, F., Giang, A., Battlay, P., Fournier-Level, A., RA, O. H. and Batterham, P. (2017). Multiple P450s and variation in neuronal genes underpins the response to the insecticide Imidacloprid in a population of Drosophila melanogaster. Sci Rep 7(1): 11338. PubMed ID: 28900129

    Remnant, E. J., Morton, C. J., Daborn, P. J., Lumb, C., Yang, Y. T., Ng, H. L., Parker, M. W. and Batterham, P. (2014). The role of Rdl in resistance to phenylpyrazoles in Drosophila melanogaster. Insect Biochem Mol Biol 54C: 11-21. PubMed ID: 25193377

    Harrop, T. W., Pearce, S. L., Daborn, P. J. and Batterham, P. (2014). Whole-Genome Expression Analysis in the Third Instar Larval Midgut of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 25193493

    Remnant, E. J., Good, R. T., Schmidt, J. M., Lumb, C., Robin, C., Daborn, P. J. and Batterham, P. (2013). Gene duplication in the major insecticide target site, Rdl, in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 23959864

    Van Hiel, M. B., Breugelmans, B., Pagel, C. N., Williams, A. K., Varan, A. K., Burke, R., Bowles, V. M. and Batterham, P. (2012). The ovicidal, larvacidal and adulticidal properties of 5,5'-dimethyl-2,2'-bipyridyl against Drosophila melanogaster. PLoS One 7: e49961. PubMed ID: 23185497

    Daborn, P. J., Lumb, C., Harrop, T. W., Blasetti, A., Pasricha, S., Morin, S., Mitchell, S. N., Donnelly, M. J., Muller, P. and Batterham, P. (2012). Using Drosophila melanogaster to validate metabolism-based insecticide resistance from insect pests. Insect Biochem Mol Biol 42: 918-924. PubMed ID: 23023059

    Sztal, T., Chung, H., Berger, S., Currie, P. D., Batterham, P. and Daborn, P. J. (2012). A cytochrome p450 conserved in insects is involved in cuticle formation. PLoS One 7: e36544. PubMed ID: 22574182

  • Buzz Baum MRC Laboratory for Molecular Cell Biology, University College London
    Cazzagon, G., Roubinet, C. and Baum, B. (2023). Polarized SCAR and the Arp2/3 complex regulate apical cortical remodeling in asymmetrically dividing neuroblasts. iScience 26(7): 107129. PubMed ID: 37434695 ]

    Roubinet, C., White, I. J. and Baum, B. (2021). Asymmetric nuclear division in neural stem cells generates sibling nuclei that differ in size, envelope composition, and chromatin organization. Curr Biol. PubMed ID: 34297912

    Curran, S., Strandkvist, C., Bathmann, J., de Gennes, M., Kabla, A., Salbreux, G. and Baum, B. (2017). Myosin II controls junction fluctuations to guide epithelial tissue ordering. Dev Cell 43(4): 480-492.e486. PubMed ID: 29107560

    Rodrigues, N. T., Lekomtsev, S., Jananji, S., Kriston-Vizi, J., Hickson, G. R. and Baum, B. (2015). Kinetochore-localized PP1-Sds22 couples chromosome segregation to polar relaxation. Nature [Epub ahead of print]. PubMed ID: 26168397

    Rosa, A., Vlassaks, E., Pichaud, F. and Baum, B. (2015). Ect2/Pbl acts via Rho and polarity proteins to direct the assembly of an isotropic actomyosin cortex upon mitotic entry. Dev Cell 32: 604-616. PubMed ID: 25703349

    Conte, V., Ulrich, F., Baum, B., Munoz, J., Veldhuis, J., Brodland, W. and Miodownik, M. (2012). A biomechanical analysis of ventral furrow formation in the Drosophila melanogaster embryo. PLoS One 7: e34473. PubMed ID: 22511944

    Brodland, G. W., Conte, V., Cranston, P. G., Veldhuis, J., Narasimhan, S., Hutson, M. S., Jacinto, A., Ulrich, F., Baum, B. and Miodownik, M. (2010). Video force microscopy reveals the mechanics of ventral furrow invagination in Drosophila. Proc Natl Acad Sci U S A 107: 22111-22116. PubMed ID: 21127270

  • Aaron Baumann Institute of Agriculture, University of Tennessee, Knoxville
    Blenau, W., Daniel, S., Balfanz, S., Thamm, M. and Baumann, A. (2017). Dm5-HT2B: Pharmacological characterization of the fifth serotonin receptor subtype of Drosophila melanogaster. Front Syst Neurosci 11: 28. PubMed ID: 28553207

    Hoff, M., Balfanz, S., Ehling, P., Gensch, T. and Baumann, A. (2011). A single amino acid residue controls Ca2+ signaling by an octopamine receptor from Drosophila melanogaster. FASEB J 25(7): 2484-2491. PubMed ID: 21478261

  • Stefan Baumgartner Developmental Genetics, Lund University
    Shlemov, A., Alexandrov, T., Golyandina, N., Holloway, D., Baumgartner, S. and Spirov, A. V. (2021). Quantification reveals early dynamics in Drosophila maternal gradients. PLoS One 16(8): e0244701. PubMed ID: 34411119

    Cai, X., Rondeel, I. and Baumgartner, S. (2021). Modulating the bicoid gradient in space and time. Hereditas 158(1): 29. PubMed ID: 34404481

    Cai, X., Fahmy, K. and Baumgartner, S. (2019). bicoid RNA localization requires the trans-Golgi network. Hereditas 156: 30. PubMed ID: 31528161

    Castillejo-Lopez, C., Cai, X., Fahmy, K. and Baumgartner, S. (2018). Drosophila exoribonuclease nibbler is a tumor suppressor, acts within the RNA(i) machinery and is not enriched in the nuage during early oogenesis. Hereditas 155: 12. PubMed ID: 28974923

    Cai, X., Akber, M., Spirov, A. and Baumgartner, S. (2017). Cortical movement of Bicoid in early Drosophila embryos is actin- and microtubule-dependent and disagrees with the SDD diffusion model. PLoS One 12(10): e0185443. PubMed ID: 28973031

    Suksuwan, W., Cai, X., Ngernsiri, L. and Baumgartner, S. (2017). Segmentation gene expression patterns in Bactrocera dorsalis and related insects: regulation and shape of blastoderm and larval cuticle. Int J Dev Biol 61(6-7): 439-450. PubMed ID: 28695964

    Hunding, A. and Baumgartner, S. (2017). Ancient role of ten-m/odz in segmentation and the transition from sequential to syncytial segmentation. Hereditas 154: 8. PubMed ID: 28461810

    Fahmy, K., Akber, M., Cai, X., Koul, A., Hayder, A. and Baumgartner, S. (2014). alphaTubulin 67C and Ncd are essential for establishing a cortical microtubular network and formation of the Bicoid mRNA gradient in drosophila. PLoS One 9: e112053. PubMed ID: 25390693

    Fahmy, K. and Baumgartner, S. (2013). Expression analysis of a family of developmentally-regulated cytosolic sulfotransferases (SULTs) in Drosophila. Hereditas 150: 44-48. PubMed ID: 23865965

    Broadie, K., Baumgartner, S. and Prokop, A. (2011). Extracellular matrix and its receptors in Drosophila neural development. Dev Neurobiol 71: 1102-1130. PubMed ID: 21688401

  • Mary Baylies Memorial Sloan-Kettering Cancer Center
    Zapater, I. M. C., Carman, P. J., Soffar, D. B., Windner, S. E., Dominguez, R. and Baylies, M. K. (2023). Drosophila Tropomodulin is required for multiple actin-dependent processes within developing myofibers. Development 150(6). PubMed ID: 36806912

    Deng, S., Silimon, R. L., Balakrishnan, M., Bothe, I., Juros, D., Soffar, D. B. and Baylies, M. K. (2020). The actin polymerization factor diaphanous and the actin severing protein flightless I collaborate to regulate sarcomere size. Dev Biol. PubMed ID: 32980309

    Balakrishnan, M., Yu, S. F., Chin, S. M., Soffar, D. B., Windner, S. E., Goode, B. L. and Baylies, M. K. (2020). Cofilin Loss in Drosophila Muscles Contributes to Muscle Weakness through Defective Sarcomerogenesis during Muscle Growth. Cell Rep 32(3): 107893. PubMed ID: 32697999

    Manhart, A., Azevedo, M., Baylies, M. and Mogilner, A. (2020). Reverse engineering forces responsible for dynamic clustering and spreading of multiple nuclei in developing muscle cells. Mol Biol Cell: mbcE19120711. PubMed ID: 32129712

    Windner, S. E., Manhart, A., Brown, A., Mogilner, A. and Baylies, M. K. (2019). Nuclear scaling is coordinated among individual nuclei in multinucleated muscle fibers. Dev Cell 49(1): 48-62.e43. PubMed ID: 30905770

    Rosen, J. N., Azevedo, M., Soffar, D. B., Boyko, V. P., Brendel, M. B., Schulman, V. K. and Baylies, M. K. (2019). The Drosophila Ninein homologue Bsg25D cooperates with Ensconsin in myonuclear positioning. J Cell Biol. PubMed ID:
    30626718

    Deng, S., Bothe, I. and Baylies, M. K. (2015). The formin Diaphanous regulates myoblast fusion through actin polymerization and Arp2/3 regulation. PLoS Genet 11: e1005381. PubMed ID: 26295716

    Dobi, K. C., Schulman, V. K. and Baylies, M. K. (2015). Specification of the somatic musculature in Drosophila. Wiley Interdiscip Rev Dev Biol [Epub ahead of print]. PubMed ID: 25728002

    Schulman, V. K., Dobi, K. C. and Baylies, M. K. (2015). Morphogenesis of the somatic musculature in Drosophila melanogaster. Wiley Interdiscip Rev Dev Biol. PubMed ID: 25758712

    Kumar, R.P., Dobi, K.C., Baylies, M.K. and Abmayr, S.M. (2015). Muscle cell fate choice requires the T-Box transcription factor Midline in Drosophila. Genetic [Epub ahead of print]. PubMed ID: 25614583

    Schulman, V. K., Folker, E. S., Rosen, J. N. and Baylies, M. K. (2014). Syd/JIP3 and JNK signaling are required for myonuclear positioning and muscle function. PLoS Genet 10: e1004880. PubMed ID: 25522254

    Wong, M. C., Dobi, K. C. and Baylies, M. K. (2014). Discrete Levels of Twist Activity Are Required to Direct Distinct Cell Functions during Gastrulation and Somatic Myogenesis. PLoS One 9: e99553. PubMed ID: 24915423

    Bothe, I., Deng, S. and Baylies, M. (2014). PI(4,5)P2 regulates myoblast fusion through Arp2/3 regulator localization at the fusion site. Development. PubMed ID: 24821989

    Folker, E. S., Schulman, V. K. and Baylies, M. K. (2013). Translocating myonuclei have distinct leading and lagging edges that require Kinesin and Dynein. Development. PubMed ID: 24335254

  • Isabelle Becam
    Goncalves Antunes, M., Sanial, M., Contremoulins, V., Carvalho, S., Plessis, A. and Becam, I. (2022). High hedgehog signaling is transduced by a multikinase-dependent switch controlling the apico-basal distribution of the GPCR smoothened. Elife 11. PubMed ID: 36083801

    Roberto, N., Becam, I., Plessis, A. and Holmgren, R. A. (2022). Engrailed, Suppressor of fused and Roadkill modulate the Drosophila GLI transcription factor Cubitus interruptus at multiple levels. Development 149(6). PubMed ID: 35290435

    Bruzzone, L., Arguelles, C., Sanial, M., Miled, S., Alvisi, G., Goncalves-Antunes, M., Qasrawi, F., Holmgren, R. A., Smibert, C. A., Lipshitz, H. D., Boccaccio, G. L., Plessis, A. and Becam, I. (2020). Regulation of the RNA-binding protein Smaug by the GPCR Smoothened via the kinase Fused. EMBO Rep 21(7): e48425. PubMed ID: 32383557

  • Peter Becker Adolf-Butenandt-Institut Molekularbiologie LMU München
    Eggers, N., Gkountromichos, F., Krause, S., Campos-Sparr, A. and Becker, P. B. (2023). Physical interaction between MSL2 and CLAMP assures direct cooperativity and prevents competition at composite binding sites. Nucleic Acids Res. PubMed ID: 37602401

    Villa, R., Jagtap, P. K. A., Thomae, A. W., Campos Sparr, A., Forne, I., Hennig, J., Straub, T. and Becker, P. B. (2021). Divergent evolution toward sex chromosome-specific gene regulation in Drosophila. Genes Dev 35(13-14): 1055-1070. PubMed ID: 34140353

    Eggers, N. and Becker, P. B. (2021). Cell-free genomics reveal intrinsic, cooperative and competitive determinants of chromatin interactions. Nucleic Acids Res 49(13): 7602-7617. PubMed ID: 34181732

    Scacchetti, A., Schauer, T., Reim, A., Apostolou, Z., Campos Sparr, A., Krause, S., Heun, P., Wierer, M. and Becker, P. B. (2020). Drosophila SWR1 and NuA4 complexes are defined by DOMINO isoforms. Elife 9. PubMed ID: 32432549

    Muller, M., Schauer, T., Krause, S., Villa, R., Thomae, A. W. and Becker, P. B. (2020). Two-step mechanism for selective incorporation of lncRNA into a chromatin modifier. Nucleic Acids Res. PubMed ID: 32510132

    Albig, C., Wang, C., Dann, G. P., Wojcik, F., Schauer, T., Krause, S., Maenner, S., Cai, W., Li, Y., Girton, J., Muir, T. W., Johansen, J., Johansen, K. M., Becker, P. B. and Regnard, C. (2019). JASPer controls interphase histone H3S10 phosphorylation by chromosomal kinase JIL-1 in Drosophila. Nat Commun 10(1): 5343. PubMed ID: 31767855

    Prayitno, K., Schauer, T., Regnard, C. and Becker, P. B. (2019). Progressive dosage compensation during Drosophila embryogenesis is reflected by gene arrangement. EMBO Rep: e48138. PubMed ID: 31286660

    Harpprecht, L., Baldi, S., Schauer, T., Schmidt, A., Bange, T., Robles, M. S., Kremmer, E., Imhof, A. and Becker, P. B. (2019). A Drosophila cell-free system that senses DNA breaks and triggers phosphorylation signalling. Nucleic Acids Res. PubMed ID: 31147711

    Ankush Jagtap, P. K., Muller, M., Masiewicz, P., von Bulow, S., Hollmann, N. M., Chen, P. C., Simon, B., Thomae, A. W., Becker, P. B. and Hennig, J. (2019). Structure, dynamics and roX2-lncRNA binding of tandem double-stranded RNA binding domains dsRBD1,2 of Drosophila helicase Maleless. Nucleic Acids Res. PubMed ID: 30805612

    Scacchetti, A., Brueckner, L., Jain, D., Schauer, T., Zhang, X., Schnorrer, F., van Steensel, B., Straub, T. and Becker, P. B. (2018). CHRAC/ACF contribute to the repressive ground state of chromatin. Life Sci Alliance 1(1): e201800024. PubMed ID: 30456345

    Albig, C., Tikhonova, E., Krause, S., Maksimenko, O., Regnard, C. and Becker, P. B. (2018). Factor cooperation for chromosome discrimination in Drosophila. Nucleic Acids Res. PubMed ID: 30541149

  • Kathleen Beckingham Department of Biochemistry & Cell Biology, Rice University, Houston
    Zhou, F., Green, S. R., Tsay, M., Hsu, S., Dibbs, R. and Beckingham, K. M. (2020). The roles of jim lovell and uninflatable in different endopolyploid larval tissues of Drosophila melanogaster. PLoS One 15(8): e0237662. PubMed ID: 32822370

    Qiang, K. M., Zhou, F. and Beckingham, K. M. (2018). A burrowing/tunneling assay for detection of hypoxia in Drosophila melanogaster larvae. J Vis Exp(133). PubMed ID: 29658928

    Zhou, F., Qiang, K. M. and Beckingham, K. M. (2016). Failure to burrow and tunnel reveals roles for jim lovell in the growth and endoreplication of the Drosophila larval tracheae. PLoS One 11: e0160233. PubMed ID: 27494251

    Taylor, K., Kleinhesselink, K., George, M. D., Morgan, R., Smallwood, T., Hammonds, A. S., Fuller, P. M., Saelao, P., Alley, J., Gibbs, A. G., Hoshizaki, D. K., von Kalm, L., Fuller, C. A., Beckingham, K. M. and Kimbrell, D. A. (2014). Toll mediated infection response is altered by gravity and spaceflight in Drosophila. PLoS One 9: e86485. PubMed ID: 24475130

    Bjorum, S. M., Simonette, R. A., Alanis, R., Jr., Wang, J. E., Lewis, B. M., Trejo, M. H., Hanson, K. A., Beckingham, K. M. (2013) The Drosophila BTB Domain Protein Jim Lovell Has Roles in Multiple Larval and Adult Behaviors. PLoS One 8: e61270. PubMed ID: 23620738

    Texada, M. J., Simonette, R. A., Deery, W. J. and Beckingham, K. M. (2011). Tropomyosin is an interaction partner of the Drosophila coiled coil protein yuri gagarin. Exp Cell Res 317: 474-487. PubMed ID: 21126519

  • David Begun Department Of Evolution and Ecology, U. C. Davis
    Lombardo, K. D., Sheehy, H. K., Cridland, J. M. and Begun, D. J. (2023). Identifying candidate de novo genes expressed in the somatic female reproductive tract of Drosophila melanogaster. G3 (Bethesda) 13(8). PubMed ID: 37259569

    Cridland, J. M., Contino, C. E. and Begun, D. J. (2023). Selection and Geography Shape Male Reproductive Tract Transcriptomes in Drosophila Melanogaster. Genetics. PubMed ID: 36869688

    Majane, A. C., Cridland, J. M. and Begun, D. J. (2021). Single-nucleus transcriptomes reveal evolutionary and functional properties of cell types in the Drosophila accessory gland. Genetics. PubMed ID: 34849871

    Cridland, J. M., Majane, A. C., Zhao, L. and Begun, D. J. (2021). Population biology of accessory gland-expressed de novo genes in Drosophila melanogaster. Genetics. PubMed ID: 34791207

    Sarikaya, D. P., Rickelton, K., Cridland, J. M., Hatmaker, R., Sheehy, H. K., Davis, S., Khan, N., Kochummen, A. and Begun, D. J. (2021). Sex and tissue-specific evolution of developmental plasticity in Drosophila melanogaster. Ecol Evol 11(3): 1334-1341. PubMed ID: 33598134

    Sarikaya, D. P., Cridland, J., Tarakji, A., Sheehy, H., Davis, S., Kochummen, A., Hatmaker, R., Khan, N., Chiu, J. and Begun, D. J. (2020). Phenotypic coupling of sleep and starvation resistance evolves in D. melanogaster. BMC Evol Biol 20(1): 126. PubMed ID: 32962630

    Cridland, J. M., Majane, A. C., Sheehy, H. K. and Begun, D. J. (2020). Polymorphism and Divergence of Novel Gene Expression Patterns in Drosophila melanogaster. Genetics. PubMed ID: 32737121

    Svetec, N., Saelao, P., Cridland, J. M., Hoffmann, A. A. and Begun, D. J. (2018). Functional Analysis of a Putative Target of Spatially Varying Selection in the Menin1 Gene of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 30404774

    Zhao, L. and Begun, D. J. (2017). Genomics of parallel adaptation at two timescales in Drosophila. PLoS Genet 13(10): e1007016. PubMed ID: 28968391

    Svetec, N., Cridland, J. M., Zhao, L. and Begun, D. J. (2016). The adaptive significance of natural genetic variation in the DNA damage response of Drosophila melanogaster. PLoS Genet 12: e1005869. PubMed ID: 26950216

    Schrider, D. R., Hahn, M. W. and Begun, D. J. (2016). Parallel evolution of copy-number variation across continents in Drosophila melanogaster. Mol Biol Evol [Epub ahead of print]. PubMed ID: 26809315

  • Greg Beitel Northwestern University
    Yang, R., Li, E., Kwon, Y. J., Mani, M. and Beitel, G. J. (2019). QuBiT: a quantitative tool for analyzing epithelial tubes reveals unexpected patterns of organization in the Drosophila trachea. Development. PubMed ID: 30967427

    McSharry, S. S. and Beitel, G. J. (2019). The Caspase-3 homolog DrICE regulates endocytic trafficking during Drosophila tracheal morphogenesis. Nat Commun 10(1): 1031. PubMed ID: 30833576

    Martin, R. L., Maiorano, J., Beitel, G. J., Marko, J. F., McVicker, G. and Fondufe-Mittendorf, Y. N. (2017). A comparison of nucleosome organization in Drosophila cell lines. PLoS One 12(6): e0178590. PubMed ID: 28570602

    Helenius, I. T., Haake, R. J., Kwon, Y. J., Hu, J. A., Krupinski, T., Casalino-Matsuda, S. M., Sporn, P. H., Sznajder, J. I. and Beitel, G. J. (2015). Identification of Drosophila Zfh2 as a Mediator of Hypercapnic Immune Regulation by a Genome-Wide RNA Interference Screen. J Immunol. PubMed ID: 26643480

    Robbins, R. M., Gbur, S. C. and Beitel, G. J. (2014). Non-Canonical Roles for Yorkie and Drosophila Inhibitor of Apoptosis 1 in Epithelial Tube Size Control. PLoS One 9: e101609. PubMed ID: 25036253

    Vadasz, I., Dada, L. A., Briva, A., Helenius, I. T., Sharabi, K., Welch, L. C., Kelly, A. M., Grzesik, B. A., Budinger, G. R., Liu, J., Seeger, W., Beitel, G. J., Gruenbaum, Y. and Sznajder, J. I. (2012). Evolutionary Conserved Role of c-Jun-N-Terminal Kinase in CO(2)-Induced Epithelial Dysfunction. PLoS One 7: e46696. PubMed ID: 23056407

    Nelson, K. S., Khan, Z., Molnar, I., Mihaly, J., Kaschube, M. and Beitel, G. J. (2012). Drosophila Src regulates anisotropic apical surface growth to control epithelial tube size. Nat Cell Biol 14: 518-525. PubMed ID: 22446737

    Choi, W., Jung, K. C., Nelson, K. S., Bhat, M. A., Beitel, G. J., Peifer, M. and Fanning, A. S. (2011). The single Drosophila ZO-1 protein Polychaetoid regulates embryonic morphogenesis in coordination with Canoe/afadin and Enabled. Mol Biol Cell 22: 2010-2030. PubMed ID: 21508316

  • Amy Bejsovec Biology Department, Duke University
    Rizzo, N. P. and Bejsovec, A. (2017). SoxNeuro and shavenbaby act cooperatively to shape denticles in the embryonic epidermis of Drosophila. Development. PubMed ID: 28506986

    Greer, E. R., Chao, A. T. and Bejsovec, A. (2013). Pebble/ECT2 RhoGEF negatively regulates the Wingless/Wnt signaling pathway. Development. PubMed ID: 24198276

    Bejsovec, A. and Chao, A. (2012). crinkled reveals a new role for Wingless signaling in Drosophila denticle formation, Development 139: 690-698. PubMed ID: 22219350

    Jones, W. M., Chao, A. T., Zavortink, M., Saint, R. and Bejsovec, A. (2010). The cytokinesis proteins Tum and Pav play a nuclear role in Wnt regulation. J. Cell Sci. 123: 2179-2189. PubMed ID: 20516152

  • Yohanns Bellaïche Institut Curie, Paris
    Villedieu, A., Alpar, L., Gaugue, I., Joudat, A., Graner, F., Bosveld, F. and Bellaiche, Y. (2023). Homeotic compartment curvature and tension control spatiotemporal folding dynamics. Nat Commun 14(1): 594. PubMed ID: 36737611

    di Pietro, F., Osswald, M., De Las Heras, J. M., Cristo, I., Lopez-Gay, J., Wang, Z., Pelletier, S., Gaugue, I., Leroy, A., Martin, C., Morais-de-Sa, E. and Bellaiche, Y. (2023). Systematic analysis of RhoGEF/GAP localizations uncovers regulators of mechanosensing and junction formation during epithelial cell division. Curr Biol 33(5): 858-874.e857. PubMed ID: 36917931

    Lopez-Gay, J. M., Nunley, H., Spencer, M., di Pietro, F., Guirao, B., Bosveld, F., Markova, O., Gaugue, I., Pelletier, S., Lubensky, D. K. and Bellaiche, Y. (2020). Apical stress fibers enable a scaling between cell mechanical response and area in epithelial tissue. Science 370(6514). PubMed ID: 33060329

    Wang, Z., Bosveld, F. and Bellaiche, Y. (2018). Tricellular junction proteins promote disentanglement of daughter and neighbour cells during epithelial cytokinesis. J Cell Sci. Pubmed ID: 29739875

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

    Pinheiro, D., Hannezo, E., Herszterg, S., Bosveld, F., Gaugue, I., Balakireva, M., Wang, Z., Cristo, I., Rigaud, S. U., Markova, O. and Bellaiche, Y. (2017). Transmission of cytokinesis forces via E-cadherin dilution and actomyosin flows. Nature [Epub ahead of print]. PubMed ID: 28296858

    Bosveld, F., Markova, O., Guirao, B., Martin, C., Wang, Z., Pierre, A., Balakireva, M., Gaugue, I., Ainslie, A., Christophorou, N., Lubensky, D. K., Minc, N. and Bellaiche, Y. (2016). Epithelial tricellular junctions act as interphase cell shape sensors to orient mitosis. Nature 530: 495-498. PubMed ID: 26886796

    Bosveld, F., Guirao, B., Wang, Z., Riviere, M., Bonnet, I., Graner, F. and Bellaiche, Y. (2016). Modulation of junction tension by tumor-suppressors and proto-oncogenes regulates cell-cell contacts. Development. PubMed ID: 26811379

    Degoutin, J. L., Milton, C. C., Yu, E., Tipping, M., Bosveld, F., Yang, L., Bellaiche, Y., Veraksa, A. and Harvey, K. F. (2013). Riquiqui and Minibrain are regulators of the Hippo pathway downstream of Dachsous. Nat Cell Biol. PubMed ID: 23955303

    Bardet, P. L., Guirao, B., Paoletti, C., Serman, F., Leopold, V., Bosveld, F., Goya, Y., Mirouse, V., Graner, F., Bellaiche, Y. (2013) PTEN Controls Junction Lengthening and Stability during Cell Rearrangement in Epithelial Tissue. Dev Cell. PubMed ID: 23707736

    Herszterg, S., Leibfried, A., Bosveld, F., Martin, C., Bellaiche, Y. (2013) Interplay between the dividing cell and its neighbors regulates adherens junction formation during cytokinesis in epithelial tissue. Dev Cell 24: 256-270. PubMed ID: 23410940

  • Hugo Bellen Baylor College of Medicine
    Ravenscroft, T. A., Jacobs, A., Gu, M., Eberl, D. F. and Bellen, H. J. (2023). The Voltage-Gated Sodium Channel in Drosophila, Para, Localizes to Dendrites As Well As Axons in Mechanosensitive Chordotonal Neurons. eNeuro 10(6). PubMed ID: 37328295

    Guichard, A., Lu, S., Kanca, O., Bressan, D., Huang, Y., Ma, M., Sanz Juste, S., Andrews, J. C., Jay, K. L., Sneider, M., Schwartz, R., Huang, M. C., Bei, D., Pan, H., Ma, L., Lin, W. W., Auradkar, A., Bhagwat, P., Park, S., Wan, K. H., Ohsako, T., Takano-Shimizu, T., Celniker, S. E., Wangler, M. F., Yamamoto, S., Bellen, H. J. and Bier, E. (2023). A comprehensive Drosophila resource to identify key functional interactions between SARS-CoV-2 factors and host proteins. Cell Rep 42(8): 112842. PubMed ID: 37480566

    Scott, H., Novikov, B., Ugur, B., Allen, B., Mertsalov, I., Monagas-Valentin, P., Koff, M., Baas Robinson, S., Aoki, K., Veizaj, R., Lefeber, D. J., Tiemeyer, M., Bellen, H. and Panin, V. (2023). Glia-neuron coupling via a bipartite sialylation pathway promotes neural transmission and stress tolerance in Drosophila. Elife 12. PubMed ID: 36946697

    Tepe, B., Macke, E. L., Niceta, M., Weisz Hubshman, M., Kanca, O., Schultz-Rogers, L., Zarate, Y. A., Schaefer, G. B., Granadillo De Luque, J. L., Wegner, D. J., Cogne, B., Gilbert-Dussardier, B., Le Guillou, X., Wagner, E. J., Pais, L. S., Neil, J. E., Mochida, G. H., Walsh, C. A., Magal, N., Drasinover, V., Shohat, M., Schwab, T., Schmitz, C., Clark, K., Fine, A., Lanpher, B., Gavrilova, R., Blanc, P., Burglen, L., Afenjar, A., Steel, D., Kurian, M. A., Prabhakar, P., Goswein, S., Di Donato, N., Bertini, E. S., Wangler, M. F., Yamamoto, S., Tartaglia, M., Klee, E. W. and Bellen, H. J. (2023). Bi-allelic variants in INTS11 are associated with a complex neurological disorder. Am J Hum Genet. PubMed ID: 37054711

    Lin, G., Tepe, B., McGrane, G., Tipon, R. C., Croft, G., Panwala, L., Hope, A., Liang, A. J. H., Zuo, Z., Byeon, S. K., Wang, L., Pandey, A. and Bellen, H. J. (2023). Exploring therapeutic strategies for infantile neuronal axonal dystrophy (INAD/PARK14). Elife 12. PubMed ID: 36645408

    Ma, M., Zhang, X., Zheng, Y., Lu, S., Pan, X., Mao, X., Pan, H., Chung, H. L., Wang, H., Guo, H. and Bellen, H. J. (2022). The fly homolog of SUPT16H, a gene associated with neurodevelopmental disorders, is required in a cell-autonomous fashion for cell survival. Hum Mol Genet. PubMed ID: 36255738

    Lu, S., Ma, M., Mao, X., Bacino, C. A., Jankovic, J., Sutton, V. R., Bartley, J. A., Wang, X., Rosenfeld, J. A., Beleza-Meireles, A., Chauhan, J., Pan, X., Li, M., Liu, P., Prescott, K., Amin, S., Davies, G., Wangler, M. F., Dai, Y. and Bellen, H. J. (2022). De novo variants in FRMD5 are associated with developmental delay, intellectual disability, ataxia, and abnormalities of eye movement. Am J Hum Genet 109(10): 1932-1943. PubMed ID: 36206744

    aKaragas, N. E., Gupta, R., Rastegari, E., Tan, K. L., Leung, H. H., Bellen, H. J., Venkatachalam, K. and Wong, C. O. (2022). Loss of Activity-Induced Mitochondrial ATP Production Underlies the Synaptic Defects in a Drosophila Model of ALS. J Neurosci 42(42): 8019-8037. PubMed ID: 36261266 Bosch, J. A., Ugur, B., Pichardo-Casas, I., Rabasco, J., Escobedo, F., Zuo, Z., Brown, B., Celniker, S., Sinclair, D. A., Bellen, H. J. and Perrimon, N. (2022). Two neuronal peptides encoded from a single transcript regulate mitochondrial complex III in Drosophila. Elife 11. PubMed ID: 36346220

    Karagas, N. E., Gupta, R., Rastegari, E., Tan, K. L., Leung, H. H., Bellen, H. J., Venkatachalam, K. and Wong, C. O. (2022). Loss of Activity-Induced Mitochondrial ATP Production Underlies the Synaptic Defects in a Drosophila Model of ALS. J Neurosci 42(42): 8019-8037. PubMed ID: 36261266

    Huang, Y., Lemire, G., Briere, L. C., Liu, F., Wessels, M. W., Wang, X., Osmond, M., Kanca, O., Lu, S., High, F. A., Walker, M. A., Rodan, L. H., Kernohan, K. D., Sweetser, D. A., Boycott, K. M. and Bellen, H. J. (2022). The recurrent de novo c.2011C>T missense variant in MTSS2 causes syndromic intellectual disability. Am J Hum Genet. PubMed ID: 36067766

    Lu, S., Hernan, R., Marcogliese, P. C., Huang, Y., Gertler, T. S., Akcaboy, M., Liu, S., Chung, H. L., Pan, X., Sun, X., Oguz, M. M., Oztoprak, U., de Baaij, J. H. F., Ivanisevic, J., McGinnis, E., Guillen Sacoto, M. J., Chung, W. K. and Bellen, H. J. (2022). Loss-of-function variants in TIAM1 are associated with developmental delay, intellectual disability, and seizures. Am J Hum Genet 109(4): 571-586. PubMed ID: 35240055

    Martelli, F., Hernandes, N. H., Zuo, Z., Wang, J., Wong, C. O., Karagas, N. E., Roessner, U., Rupasinghe, T., Robin, C., Venkatachalam, K., Perry, T., Batterham, P. and Bellen, H. J. (2022). Low doses of the organic insecticide spinosad trigger lysosomal defects, elevated ROS, lipid dysregulation, and neurodegeneration in flies. Elife 11. PubMed ID: 35191376

  • Xavier Belles Institut de Biologia Evolutiva, Barcelona
    Karagas, N. E., Gupta, R., Rastegari, E., Tan, K. L., Leung, H. H., Bellen, H. J., Venkatachalam, K. and Wong, C. O. (2022). Loss of Activity-Induced Mitochondrial ATP Production Underlies the Synaptic Defects in a Drosophila Model of ALS. J Neurosci 42(42): 8019-8037. PubMed ID: 36261266

    Santos, C. G., Fernandez-Nicolas, A. and Belles, X. (2016). Smads and insect hemimetabolan metamorphosis. Dev Biol [Epub ahead of print]. PubMed ID: 27452629

    Fernandez-Nicolas, A. and Belles, X. (2015). CREB-binding protein contributes to the regulation of endocrine and developmental pathways in insect hemimetabolan pre-metamorphosis. Biochim Biophys Acta. PubMed ID: 26706852

    Lozano, J., Kayukawa, T., Shinoda, T. and Belles, X. (2014). A role for Taiman in insect metamorphosis. PLoS Genet 10: e1004769. PubMed ID: 25356827

    Lozano, J., Montanez, R. and Belles, X. (2015). MiR-2 family regulates insect metamorphosis by controlling the juvenile hormone signaling pathway. Proc Natl Acad Sci U S A 112: 3740-3745. PubMed ID: 25775510

    Huang, J. H., Lozano, J. and Belles, X. (2013). Broad-complex functions in postembryonic development of the cockroach Blattella germanica shed new light on the evolution of insect metamorphosis. Biochim Biophys Acta 1830: 2178-2187. PubMed ID: 23041750

    Rubio, M., de Horna, A., Belles, X. (2012) MicroRNAs in metamorphic and non-metamorphic transitions in hemimetabolan insect metamorphosis. BMC Genomics 13: 386. PubMed ID: 22882747

    Boher, F., Trefault, N., Piulachs, M. D., Belles, X., Godoy-Herrera, R. and Bozinovic, F. (2012). Biogeographic origin and thermal acclimation interact to determine survival and hsp90 expression in Drosophila species submitted to thermal stress. Comp Biochem Physiol A Mol Integr Physiol 162: 391-396. PubMed ID: 22561660

    Lozano, J. and Belles, X. (2011). Conserved repressive function of Kruppel homolog 1 on insect metamorphosis in hemimetabolous and holometabolous species. Sci Rep 1: 163. PubMed ID: 22355678

  • Paola Bellosta Department Of Genetics And Development, Columbia University
    Destefanis, F., Manara, V., Santarelli, S., Zola, S., Brambilla, M., Viola, G., Maragno, P., Signoria, I., Viero, G., Pasini, M. E., Penzo, M. and Bellosta, P. (2022). Reduction of nucleolar NOC1 accumulates pre-rRNAs and induces Xrp1 affecting growth and resulting in cell competition. J Cell Sci. PubMed ID: 36314272

    Paiardi, C., Mirzoyan, Z., Zola, S., Parisi, F., Vingiani, A., Pasini, M. E. and Bellosta, P. (2017). The Stearoyl-CoA Desaturase-1 (Desat1) in Drosophila cooperated with Myc to induce autophagy and growth, a potential new link to tumor survival. Genes (Basel) 8(5). PubMed ID: 28452935

    Paiardi, C., Mirzoyan, Z., Zola, S., Parisi, F., Vingiani, A., Pasini, M.E. and Bellosta, P. (2017). The Stearoyl-CoA Desaturase-1 (Desat1) in Drosophila cooperated with Myc to Induce Autophagy and Growth, a Potential New Link to Tumor Survival. Genes (Basel) 8. PubMed ID: 28452935

    Grifoni, D. and Bellosta, P. (2014). Drosophila Myc: a master regulator of cellular performance. Biochim Biophys Acta. PubMed ID: 25010747

    Parisi, F., Riccardo, S., Zola, S., Lora, C., Grifoni, D., Brown, L. M., Bellosta, P. (2013) dMyc expression in the fat body affects DILP2 release and increases the expression of the fat desaturase Desat1 resulting in organismal growth. Dev Biol. PubMed ID: 23608455

    Parisi, F., et al. (2011). Drosophila insulin and target of rapamycin (TOR) pathways regulate GSK3 beta activity to control Myc stability and determine Myc expression in vivo. BMC Biol. 9: 65. PubMed ID: 21951762

    Ziosi, M, et al. (2010). dMyc functions downstream of Yorkie to promote the supercompetitive behavior of hippo pathway mutant cells. PLoS Genet. 6(9) e1001140. PubMed ID:
    20885789

  • Welcome Bender Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School
    Bowman, S. K., Deaton, A. M., Domingues, H., Wang, P. I., Sadreyev, R. I., Kingston, R. E. and Bender, W. (2014). H3K27 modifications define segmental regulatory domains in the Drosophila bithorax complex. Elife (Cambridge): e02833. PubMed ID: 25082344

    Pease, B., Borges, A. C. and Bender, W. (2013). Non-Coding RNAs of the Ultrabithorax Domain of the Drosophila Bithorax Complex. Genetics. PubMed ID: 24077301

    Bender, W. and Lucas, M. (2013). The Border between the Ultrabithorax and abdominal-A Domains in the Drosophila Bithorax Complex. Genetics. PubMed ID: 23288934

    Gummalla, M., Maeda, R. K., Castro Alvarez, J. J., Gyurkovics, H., Singari, S., Edwards, K. A., Karch, F. and Bender, W. (2012). abd-A regulation by the iab-8 noncoding RNA. PLoS Genet 8: e1002720. PubMed ID: 22654672

    Ho, M. C., Johnsen, H., Goetz, S. E., Schiller, B. J., Bae, E., Tran, D. A., Shur, A. S., Allen, J. M., Rau, C., Bender, W., Fisher, W. W., Celniker, S. E. and Drewell, R. A. (2009). Functional evolution of cis-regulatory modules at a homeotic gene in Drosophila. PLoS Genet 5: e1000709. PubMed ID: 19893611

  • Jonathan Benito-Sipos Department of Biology, Universidad Autónoma de Madrid
    Rubio-Ferrera, I., Clarembaux-Badell, L., Baladrón-de-Juan, P., Berrocal-Rubio, M., Thor, S., Cobeta, I. M. and Benito-Sipos, J. (2023). Specification of the Drosophila Orcokinin A neurons by combinatorial coding. Cell Tissue Res 391(2): 269-286. PubMed ID: 36512054

    Clarembaux-Badell, L., Baladron-de-Juan, P., Gabilondo, H., Rubio-Ferrera, I., Millan, I., Estella, C., Valverde-Ortega, F. S., Cobeta, I. M., Thor, S. and Benito-Sipos, J. (2022). Dachshund acts with Abdominal-B to trigger programmed cell death in the Drosophila central nervous system at the frontiers of Abd-B expression. Dev Neurobiol 82(6): 495-504. PubMed ID: 35796156

    Monedero Cobeta, I., Stadler, C. B., Li, J., Yu, P., Thor, S. and Benito-Sipos, J. (2018). Specification of Drosophila neuropeptidergic neurons by the splicing component brr2. PLoS Genet 14(8): e1007496. PubMed ID: 30133436

    Gabilondo, H., Rubio-Ferrera, I., Losada-Perez, M., Del Saz, D., Leon, Y., Molina, I., Torroja, L., D, W. A. and Benito-Sipos, J. (2018). Segmentally homologous neurons acquire two different terminal neuropeptidergic fates in the Drosophila nervous system. PLoS One 13(4): e0194281. PubMed ID: 29634720

    Stratmann, J., Gabilondo, H., Benito-Sipos, J. and Thor, S. (2016). Neuronal cell fate diversification controlled by sub-temporal action of Kruppel. Elife 5 [Epub ahead of print]. PubMed ID: 27740908
    Gabilondo, H., Stratmann, J., Rubio-Ferrera, I., Millán-Crespo, I., Contero-García, P., Bahrampour, S., Thor, S. and Benito-Sipos, J. (2016). Neuronal cell fate specification by the convergence of different spatiotemporal cues on a common terminal selector cascade. PLoS Biol 14: e1002450. PubMed ID: 27148744

    Gabilondo, H., Losada-Perez, M., Monedero, I., Torres-Herraez, A., Molina, I., Torroja, L. and Benito-Sipos, J. (2014). A new role of Klumpfuss in establishing cell fate during the GMC asymmetric cell division. Cell Tissue Res. PubMed ID: 25129108

    Gabilondo, H., Losada-Perez, M., del Saz, D., Molina, I., Leon, Y., Canal, I., Torroja, L. and Benito-Sipos, J. (2011). A targeted genetic screen identifies crucial players in the specification of the Drosophila abdominal Capaergic neurons. Mech Dev 128: 208-221. PubMed ID: 21236339

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

  • Daimark Bennett Institute of Integrative Biology, University of Liverpool
    Badmos, H., Cobbe, N., Campbell, A., Jackson, R. and Bennett, D. (2021). Drosophila USP22/nonstop polarizes the actin cytoskeleton during collective border cell migration. J Cell Biol 220(7). PubMed ID: 33988679

    Jonchere, V., Alqadri, N., Herbert, J., Dodgson, L., Mason, D., Messina, G., Falciani, F. and Bennett, D. (2017). Transcriptional responses to hyperplastic MRL signalling in Drosophila. Open Biol 7(2). PubMed ID: 28148822

    Ciurciu, A., Duncalf, L., Jonchere, V., Lansdale, N., Vasieva, O., Glenday, P., Rudenko, A., Vissi, E., Cobbe, N., Alphey, L. and Bennett, D. (2013). PNUTS/PP1 Regulates RNAPII-Mediated Gene Expression and Is Necessary for Developmental Growth. PLoS Genet 9: e1003885. PubMed ID: 24204300

    Jonchere, V. and Bennett, D. (2013). Validating RNAi Phenotypes in Drosophila Using a Synthetic RNAi-Resistant Transgene. PLoS One 8: e70489. PubMed ID: 23950943

    Hutchence, K. J., Pade, R., Swift, H. L., Bennett, D. and Hurst, G. D. (2012). Phenotype and transmission efficiency of artificial and natural male-killing Spiroplasma infections in Drosophila melanogaster. J Invertebr Pathol 109: 243-247. PubMed ID: 22085686

    Shulman, J. M., Chipendo, P., Chibnik, L. B., Aubin, C., Tran, D., Keenan, B. T., Kramer, P. L., Schneider, J. A., Bennett, D. A., Feany, M. B. and De Jager, P. L. (2011). Functional screening of Alzheimer pathology genome-wide association signals in Drosophila. Am J Hum Genet 88: 232-238. PubMed ID: 21295279

  • Yehuda Ben-Shahar Department of Biology, Washington University, St. Louis
    Vernier, C. L., Leitner, N., Zelle, K. M., Foltz, M., Dutton, S., Liang, X., Halloran, S., Millar, J. G. and Ben-Shahar, Y. (2023). A pleiotropic chemoreceptor facilitates the production and perception of mating pheromones. iScience 26(1): 105882. PubMed ID: 36691619

    Lucas, C. and Ben-Shahar, Y. (2021). The foraging gene as a modulator of division of labour in social insects. J Neurogenet: 1-11. PubMed ID: 34151702

    McKinney, R. M., Valdez, R. and Ben-Shahar, Y. (2021). The genetic architecture of larval aggregation behavior in Drosophila. J Neurogenet: 1-16. PubMed ID: 33629904

    Hill, A. S., Jain, P., Folan, N. E. and Ben-Shahar, Y. (2019). The Drosophila ERG channel seizure plays a role in the neuronal homeostatic stress response. PLoS Genet 15(8): e1008288. PubMed ID: 31393878

    Søvik, E., LaMora, A., Seehra, G., Barron, A.B., Duncan, J.G. and Ben-Shahar, Y. (2017). Drosophila divalent metal ion transporter Malvolio is required in dopaminergic neurons for feeding decisions. Genes Brain Behav [Epub ahead of print]. PubMed ID: 28220999

    Hill, A., Zheng, X., Li, X., McKinney, R., Dickman, D. and Ben-Shahar, Y. (2017). The Drosophila postsynaptic DEG/ENaC channel ppk29 contributes to excitatory neurotransmission. J Neurosci [Epub ahead of print]. PubMed ID: 28213447

    Zheng, X., Valakh, V., Diantonio, A. and Ben-Shahar, Y. (2014). Natural antisense transcripts regulate the neuronal stress response and excitability. Elife 3: e01849. PubMed ID: 24642409

    Lu, B., Zelle, K. M., Seltzer, R., Hefetz, A. and Ben-Shahar, Y. (2014). Feminization of pheromone-sensing neurons affects mating decisions in Drosophila males. Biol Open. PubMed ID: 24463366

    Zelle, K. M., Lu, B., Pyfrom, S. C., Ben-Shahar, Y. (2013) The genetic architecture of degenerin/epithelial sodium channels in Drosophila. G3 (Bethesda) 3: 441-450. Pubmed ID: 23449991

    Lu, B., LaMora, A., Sun, Y., Welsh, M. J. and Ben-Shahar, Y. (2012). ppk23-Dependent chemosensory functions contribute to courtship behavior in Drosophila melanogaster. PLoS Genet 8: e1002587. PubMed ID: 22438833

  • Matthew Benton Department of Zoology, University of Cambridge, Cambridge, United Kingdom.
    Clark, E., Battistara, M. and Benton, M. A. (2022). A timer gene network is spatially regulated by the terminal system in the Drosophila embryo. Elife 11. PubMed ID: 36524728

    Richards, L., Lord, C. L., Benton, M. L., Capra, J. A. and Nordman, J. T. (2022). Nucleoporins facilitate ORC loading onto chromatin. Cell Rep 41(6): 111590. PubMed ID: 36351393

    Munden, A., Benton, M. L., Capra, J. A. and Nordman, J. T. (2022). R-loop Mapping and Characterization During Drosophila Embryogenesis Reveals Developmental Plasticity in R-loop Signatures. J Mol Biol 434(13): 167645. PubMed ID: 35609632

    Tidswell, O. R. A., Benton, M. A. and Akam, M. (2021). The neuroblast timer gene nubbin exhibits functional redundancy with gap genes to regulate segment identity in Tribolium. Development 148(16). PubMed ID: 34351412

  • Richard Benton Faculté de biologie et de médecine, Center for Integrative Genomics, Lausanne, Switzerland
    Prieto-Godino, L. L., Schmidt, H. R. and Benton, R. (2021). Molecular reconstruction of recurrent evolutionary switching in olfactory receptor specificity. Elife 10. PubMed ID: 34677122

    Mika, K., Cruchet, S., Chai, P. C., Prieto-Godino, L. L., Auer, T. O., Pradervand, S. and Benton, R. (2021). Olfactory receptor-dependent receptor repression in Drosophila. Sci Adv 7(32). PubMed ID: 34362730

    Arguello, J. R., Abuin, L., Armida, J., Mika, K., Chai, P. C. and Benton, R. (2021). Targeted molecular profiling of rare olfactory sensory neurons identifies fate, wiring and functional determinants. Elife 10. PubMed ID: 33666172

    Prieto-Godino, L. L., Silbering, A. F., Khallaf, M. A., Cruchet, S., Bojkowska, K., Pradervand, S., Hansson, B. S., Knaden, M. and Benton, R. (2020). Functional integration of "undead" neurons in the olfactory system. Sci Adv 6(11): eaaz7238. PubMed ID: 32195354

    Auer, T. O., Khallaf, M. A., Silbering, A. F., Zappia, G., Ellis, K., Alvarez-Ocana, R., Arguello, J. R., Hansson, B. S., Jefferis, G., Caron, S. J. C., Knaden, M. and Benton, R. (2020). Olfactory receptor and circuit evolution promote host specialization. Nature 579(7799): 402-408. PubMed ID: 32132713

    Abuin, L., Prieto-Godino, L. L., Pan, H., Gutierrez, C., Huang, L., Jin, R. and Benton, R. (2019). In vivo assembly and trafficking of olfactory Ionotropic Receptors. BMC Biol 17(1): 34. PubMed ID: 30995910

    Chai, P. C., Cruchet, S., Wigger, L. and Benton, R. (2019). Sensory neuron lineage mapping and manipulation in the Drosophila olfactory system. Nat Commun 10(1): 643. PubMed ID: 30733440

    Sanchez-Alcaniz, J. A., Silbering, A. F., Croset, V., Zappia, G., Sivasubramaniam, A. K., Abuin, L., Sahai, S. Y., Munch, D., Steck, K., Auer, T. O., Cruchet, S., Neagu-Maier, G. L., Sprecher, S. G., Ribeiro, C., Yapici, N. and Benton, R. (2018). An expression atlas of variant ionotropic glutamate receptors identifies a molecular basis of carbonation sensing. Nat Commun 9(1): 4252. PubMed ID: 30315166

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

    Sanchez-Alcaniz, J. A., Zappia, G., Marion-Poll, F. and Benton, R. (2017). A mechanosensory receptor required for food texture detection in Drosophila. Nat Commun 8: 14192. PubMed ID: 28128210

    Prieto-Godino, L. L., Rytz, R., Cruchet, S., Bargeton, B., Abuin, L., Silbering, A.

  • Celeste Berg Department of Genome Sciences, University of Washington, Seattle
    Sustar, A. E., Strand, L. G., Zimmerman, S. G. and Berg, C. A. (2022). Imaginal disc growth factors are Drosophila Chitinase-like Proteins with roles in morphogenesis and CO2 response. Genetics. PubMed ID: 36576887

    Espinoza, C. Y. and Berg, C. A. (2020). Detecting New Allies: Modifier Screen Identifies a Genetic Interaction Between Imaginal disc growth factor 3 and combover, a Rho-kinase Substrate, During Dorsal Appendage Tube Formation in Drosophila. G3 (Bethesda). PubMed ID: 32855169

    O'Hanlon, K. N., Dam, R. A., Archambeault, S. L. and Berg, C. A. (2017). Two Drosophilids exhibit distinct EGF pathway patterns in oogenesis. Dev Genes Evol. PubMed ID: 29264645

    Zimmerman, S. G., Merrihew, G. E., MacCoss, M. J. and Berg, C. A. (2017). Proteomics analysis identifies orthologs of human chitinase-like proteins as inducers of tube-morphogenesis defects in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28404605

    Peters, N. C. and Berg, C. A. (2015). Dynamin-mediated endocytosis is required for tube closure, cell intercalation, and biased apical expansion during epithelial tubulogenesis in the Drosophila ovary. Dev Biol. PubMed ID: 26542010

    Zimmerman, S. G., Peters, N. C., Altaras, A. E. and Berg, C. A. (2013). Optimized RNA ISH, RNA FISH and protein-RNA double labeling (IF/FISH) in Drosophila ovaries. Nat Protoc 8: 2158-2179. PubMed ID: 24113787

    Peters, N. C., Thayer, N. H., Kerr, S. A., Tompa, M., Berg, C. A. (2013) Following the 'tracks': Tramtrack69 regulates epithelial tube expansion in the Drosophila ovary through Paxillin, Dynamin, and the homeobox protein Mirror. Dev Biol. PubMed ID: 23545328

    Kim, Y., Andreu, M. J., Lim, B., Chung, K., Terayama, M., Jimenez, G., Berg, C. A., Lu, H. and Shvartsman, S. Y. (2011). Gene regulation by MAPK substrate competition. Dev Cell 20: 880-887. PubMed ID: 21664584

    Boyle, M. J., French, R. L., Cosand, K. A., Dorman, J. B., Kiehart, D. P. and Berg, C. A. (2010). Division of labor: subsets of dorsal-appendage-forming cells control the shape of the entire tube. Dev Biol 346: 68-79. PubMed ID: 20659448

  • Christian Berger Institute of Molecular Biology, Mainz, Germany
    Zhang, W., et al. (2016). The nucleosome remodeling and deacetylase complex NuRD is built from preformed catalytically active sub-modules. J Mol Biol. PubMed ID: 27117189

    Becker, H., Renner, S., Technau, G.M. and Berger, C. (2016). Cell-autonomous and non-cell-autonomous function of Hox genes specify segmental neuroblast identity in the gnathal region of the embryonic CNS in Drosophila. PLoS Genet 12: e1005961. PubMed ID: 27015425

    Ding, R., Weynans, K., Bossing, T., Barros, C. S. and Berger, C. (2016). The Hippo signalling pathway maintains quiescence in Drosophila neural stem cells. Nat Commun 7: 10510. PubMed ID: 26821647

    Technau, G. M., Rogulja-Ortmann, A., Berger, C., Birkholz, O. and Rickert, C. (2014). Composition of a neuromere and its segmental diversification under the control of Hox genes in the embryonic CNS of Drosophila. J Neurogenet 28: 171-180. PubMed ID: 24913688

    Homem, C. C., Reichardt, I., Berger, C., Lendl, T. and Knoblich, J. A. (2013). Long-term live cell imaging and automated 4D analysis of drosophila neuroblast lineages. PLoS One 8: e79588. PubMed ID: 24260257

    Birkholz, O., Vef, O., Rogulja-Ortmann, A., Berger, C. and Technau, G. M. (2013). Abdominal-B and caudal inhibit the formation of specific neuroblasts in the Drosophila tail region. Development 140: 3552-3564. PubMed ID: 23903193

  • Alan Bergland Department of Biology, University of Virginia, Charlottesville
    Yu, Y. and Bergland, A. O. (2022). Distinct signals of clinal and seasonal allele frequency change at eQTLs in Drosophila melanogaster. Evolution. PubMed ID: 36097359

    Erickson, P. A., Weller, C. A., Song, D. Y., Bangerter, A. S., Schmidt, P. and Bergland, A. O. (2020). Unique genetic signatures of local adaptation over space and time for diapause, an ecologically relevant complex trait, in Drosophila melanogaster. PLoS Genet 16(11): e1009110. PubMed ID: 33216740

    Kapun, M., Barron, M. G., Staubach, F., Obbard, D. J., Wiberg, R. A. W., Vieira, J., Goubert, C., Rota-Stabelli, O., Kankare, M., Bogaerts-Marquez, M., Haudry, A., Waidele, L., Kozeretska, I., Pasyukova, E. G., Loeschcke, V., Pascual, M., Vieira, C. P., Serga, S., Montchamp-Moreau, C., Abbott, J., Gibert, P., Porcelli, D., Posnien, N., Sanchez-Gracia, A., Grath, S., Sucena, E., Bergland, A. O., Guerreiro, M. P. G., Onder, B. S., Argyridou, E., Guio, L., Schou, M. F., Deplancke, B., Vieira, C., Ritchie, M. G., Zwaan, B. J., Tauber, E., Orengo, D. J., Puerma, E., Aguade, M., Schmidt, P., Parsch, J., Betancourt, A. J., Flatt, T. and Gonzalez, J. (2020). Genomic Analysis of European Drosophila melanogaster Populations Reveals Longitudinal Structure, Continent-Wide Selection, and Previously Unknown DNA Viruses. Mol Biol Evol 37(9): 2661-2678. PubMed ID: 32413142

    Stone, H. M., Erickson, P. A. and Bergland, A. O. (2020). Phenotypic plasticity, but not adaptive tracking, underlies seasonal variation in post-cold hardening freeze tolerance of Drosophila melanogaster. Ecol Evol 10(1): 217-231. PubMed ID: 31988724

  • Aviv Bergman Albert Einstein College of Medicine, Bronx, NY
    Lambros, M., Sella, Y. and Bergman, A. (2023). Phenotypic pliancy and the breakdown of epigenetic polycomb mechanisms. PLoS Comput Biol 19(2): e1010889. PubMed ID: 36809239

    Gombar, S., MacCarthy, T. and Bergman, A. (2014). Epigenetics decouples mutational from environmental robustness. Did it also facilitate multicellularity? PLoS Comput Biol 10(3): e1003450. PubMed ID: 24604070

    Chang, A. L. S., Atzmon, G., Bergman, A., Brugmann, S., Atwood, S. X., Chang, H. Y. and Barzilai, N. (2014). Identification of genes promoting skin youthfulness by genome-wide association study. J Invest Dermatol 134(3): 651-657. PubMed ID: 24037343

  • Casey Bergman Department of Genetics, University of Georgia
    Han, S., Basting, P. J., Dias, G. B., Luhur, A., Zelhof, A. C. and Bergman, C. M. (2021). Transposable element profiles reveal cell line identity and loss of heterozygosity in Drosophila cell culture. Genetics 219(2). PubMed ID: 34849875

    Manee, M. M., Jackson, J. and Bergman, C. M. (2018). Conserved noncoding elements influence the transposable element landscape in Drosophila. Genome Biol Evol. PubMed ID: 29850787

    Miller, D. E., Cook, K. R., Yeganeh Kazemi, N., Smith, C. B., Cockrell, A. J., Hawley, R. S. and Bergman, C. M. (2016). Rare recombination events generate sequence diversity among balancer chromosomes in Drosophila melanogaster. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26903656

    Rahman, R., Chirn, G. W., Kanodia, A., Sytnikova, Y. A., Brembs, B., Bergman, C. M. and Lau, N. C. (2015). Unique transposon landscapes are pervasive across Drosophila melanogaster genomes. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 26578579

    Gutzwiller, F., Carmo, C. R., Miller, D. E., Rice, D. W., Newton, I. L., Hawley, R. S., Teixeira, L. and Bergman, C. M. (2015). Dynamics of Wolbachia pipientis gene expression across the Drosophila melanogaster life cycle. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26497146

    Blumenstiel, J. P., Chen, X., He, M. and Bergman, C. M. (2013). An Age-of-Allele Test of Neutrality for Transposable Element Insertions. Genetics. PubMed ID: 24336751

    Richardson, M. F., Weinert, L. A., Welch, J. J., Linheiro, R. S., Magwire, M. M., Jiggins, F. M. and Bergman, C. M. (2012). Population Genomics of the Wolbachia Endosymbiont in Drosophila melanogaster. PLoS Genet 8: e1003129. PubMed ID: 23284297

    Linheiro, R. S. and Bergman, C. M. (2012). Whole genome resequencing reveals natural target site preferences of transposable elements in Drosophila melanogaster. PLoS One 7: e30008. PubMed ID: 22347367

    Mendez-Lago, M., Bergman, C. M., de Pablos, B., Tracey, A., Whitehead, S. L. and Villasante, A. (2011). A large palindrome with interchromosomal gene duplications in the pericentromeric region of the D. melanogaster Y chromosome. Mol Biol Evol 28: 1967-1971. PubMed ID: 21297157

  • Andreas Bergmann University of Massachusetts Medical School, Worcester
    Farrell, L., Puig-Barbe, A., Haque, M. I., Amcheslavsky, A., Yu, M., Bergmann, A. and Fan, Y. (2022). Actin remodeling mediates ROS production and JNK activation to drive apoptosis-induced proliferation. PLoS Genet 18(12): e1010533. PubMed ID: 36469525

    Shields, A., Amcheslavsky, A., Brown, E., Lee, T. V., Nie, Y., Tanji, T., Ip, Y. T. and Bergmann, A. (2022). Toll-9 interacts with Toll-1 to mediate a feedback loop during apoptosis-induced proliferation in Drosophila. Cell Rep 39(7): 110817. PubMed ID: 35584678

    Lindblad, J. L., Tare, M., Amcheslavsky, A., Shields, A. and Bergmann, A. (2021). Non-apoptotic enteroblast-specific role of the initiator caspase Dronc for development and homeostasis of the Drosophila intestine. Sci Rep 11(1): 2645. PubMed ID: 33514791

    Amcheslavsky, A., Lindblad, J. L. and Bergmann, A. (2020). Transiently "Undead" Enterocytes Mediate Homeostatic Tissue Turnover in the Adult Drosophila Midgut. Cell Rep 33(8): 108408. PubMed ID: 33238125

    Amcheslavsky, A., Wang, S., Fogarty, C. E., Lindblad, J. L., Fan, Y. and Bergmann, A. (2018). Plasma membrane localization of apoptotic caspases for non-apoptotic functions. Dev Cell 45(4): 450-464.e453. PubMed ID: 29787709

    Kamber Kaya, H.E., Ditzel, M., Meier, P. and Bergmann, A. (2017). An inhibitory mono-ubiquitylation of the Drosophila initiator caspase Dronc functions in both apoptotic and non-apoptotic pathways. PLoS Genet 13: e1006438. PubMed ID: 28207763

    Lee, T. V., Kamber Kaya, H. E., Simin, R., Baehrecke, E. H. and Bergmann, A. (2016). The initiator caspase Dronc is subject of enhanced autophagy upon proteasome impairment in Drosophila. Cell Death Differ. PubMed ID: 27104928

    Fogarty, C. E., Diwanji, N., Lindblad, J. L., Tare, M., Amcheslavsky, A., Makhijani, K., Bruckner, K., Fan, Y. and Bergmann, A. (2016). Extracellular Reactive Oxygen Species Drive Apoptosis-Induced Proliferation via Drosophila Macrophages. Curr Biol [Epub ahead of print]. PubMed ID: 26898463

    Wu, Y., Lindblad, J. L., Garnett, J., Kamber Kaya, H. E., Xu, D., Zhao, Y., Flores, E. R., Hardy, J. and Bergmann, A. (2015). Genetic characterization of two gain-of-function alleles of the effector caspase DrICE in Drosophila. Cell Death Differ. PubMed ID: 26542461

    Perez, E., Das, G., Bergmann, A. and Baehrecke, E. H. (2014). Autophagy regulates tissue overgrowth in a context-dependent manner. Oncogene 0. PubMed ID: 25174403

    Fan, Y. and Bergmann, A. (2014). Multiple Mechanisms Modulate Distinct Cellular Susceptibilities toward Apoptosis in the Developing Drosophila Eye. Dev Cell. PubMed ID: 24981611

    Fogarty, C. E. and Bergmann, A. (2014). Detecting caspase activity in Drosophila larval imaginal discs. Methods Mol Biol 1133: 109-117. PubMed ID: 24567098

  • Dan Bergstralh Department of Biology, University of Rochester, Rochester, NY
    Neville, K. E., Finegan, T. M., Lowe, N., Bellomio, P. M., Na, D. and Bergstralh, D. T. (2023). The Drosophila mitotic spindle orientation machinery requires activation, not just localization. EMBO Rep: e56074. PubMed ID: 36629398

    Cammarota, C., Finegan, T. M., Wilson, T. J., Yang, S. and Bergstralh, D. T. (2020). An Axon-Pathfinding Mechanism Preserves Epithelial Tissue Integrity. Curr Biol. PubMed ID: 33065006

    Finegan, T. M., Na, D., Cammarota, C., Skeeters, A. V., Nadasi, T. J., Dawney, N. S., Fletcher, A. G., Oakes, P. W. and Bergstralh, D. T. (2018). Tissue tension and not interphase cell shape determines cell division orientation in the Drosophila follicular epithelium. Embo j. PubMed ID: 30478193

    Wilson, T. J. and Bergstralh, D. T. (2017). Cell reintegration: Stray epithelial cells make their way home. Bioessays 39(6). PubMed ID: 28440545

    Bergstralh, D. T., Lovegrove, H. E., Kujawiak, I., Dawney, N. S., Zhu, J., Cooper, S., Zhang, R. and St Johnston, D. (2016). Pins is not required for spindle orientation in the Drosophila wing disc. Development 143(14): 2573-2581. PubMed ID: 27287805

  • Andre Bernards Massachusetts General Hospital, Boston
    Walker, J. A. and Bernards, A. (2014). A Drosophila screen identifies neurofibromatosis-1 genetic modifiers involved in systemic and synaptic growth. Rare Dis 2: e28341. PubMed ID: 25054093

    Walker, J. A., Gouzi, J. Y., Long, J. B., Huang, S., Maher, R. C., Xia, H., Khalil, K., Ray, A., Van Vactor, D., Bernards, R. and Bernards, A. (2013). Genetic and Functional Studies Implicate Synaptic Overgrowth and Ring Gland cAMP/PKA Signaling Defects in the Drosophila melanogaster Neurofibromatosis-1 Growth Deficiency. PLoS Genet 9: e1003958. PubMed ID: 24278035

    Gouzi, J. Y., Moressis, A., Walker, J. A., Apostolopoulou, A. A., Palmer, R. H., Bernards, A. and Skoulakis, E. M. (2011). The receptor tyrosine kinase Alk controls neurofibromin functions in Drosophila growth and learning. PLoS Genet 7: e1002281. PubMed ID: 21949657

  • Jimena Berni Department of Zoology, University of Cambridge
    Wosniack, M. E., Festa, D., Hu, N., Gjorgjieva, J. and Berni, J. (2022). Adaptation of Drosophila larva foraging in response to changes in food resources. Elife 11. PubMed ID: 36458693

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

    Berni, J. (2015). Genetic dissection of a regionally differentiated network for exploratory behavior in Drosophila larvae. Curr Biol 25: 1319-1326. PubMed ID: 25959962

    Gjorgjieva, J., Berni, J., Evers, J. F. and Eglen, S. J. (2013). Neural circuits for peristaltic wave propagation in crawling Drosophila larvae: analysis and modeling. Front Comput Neurosci 7: 24. PubMed ID: 23576980

    Pulver, S. R. and Berni, J. (2012). The fundamentals of flying: simple and inexpensive strategies for employing Drosophila genetics in neuroscience teaching laboratories. J Undergrad Neurosci Educ 11: A139-148. PubMed ID: 23493248

    Berni, J., Pulver, S. R., Griffith, L. C. and Bate, M. (2012). Autonomous circuitry for substrate exploration in freely moving Drosophila larvae. Curr Biol 22: 1861-1870. PubMed ID: 22940472

  • Sanford Bernstein San Diego State University
    Guo, Y., Kronert, W. A., Hsu, K. H., Huang, A., Sarsoza, F., Bell, K. M., Suggs, J. A., Swank, D. M. and Bernstein, S. I. (2020). Drosophila myosin mutants model the disparate severity of type 1 and type 2B distal arthrogryposis and indicate an enhanced actin affinity mechanism. Skelet Muscle 10(1): 24. PubMed ID: 32799913

    Bloemink, M. J., Hsu, K. H., Geeves, M. A. and Bernstein, S. I. (2020). Alternative N-terminal regions of Drosophila myosin heavy chain II regulate communication of the purine binding loop with the essential light chain. J Biol Chem. PubMed ID: 32817166

    Rao, D. S., Kronert, W. A., Guo, Y., Hsu, K. H., Sarsoza, F. and Bernstein, S. I. (2018). Reductions in ATPase activity, actin sliding velocity and myofibril stability yield muscle dysfunction in Drosophila models of myosin-based Freeman Sheldon syndrome. Mol Biol Cell: mbcE18080526. PubMed ID: 30379605

    Suggs, J. A., Melkani, G. C., Glasheen, B. M., Detor, M. M., Melkani, A., Marsan, N. P., Swank, D. M. and Bernstein, S. I. (2017). A Drosophila model of dominant inclusion body myopathy 3 shows diminished myosin kinetics that reduce muscle power and yield myofibrillar defects. Dis Model Mech. PubMed ID: 28258125

    Achal, M., Trujillo, A. S., Melkani, G. C., Farman, G. P., Ocorr, K., Viswanathan, M. C., Kaushik, G., Newhard, C. S., Glasheen, B. M., Melkani, A., Suggs, J. A., Moore, J. R., Swank, D. M., Bodmer, R., Cammarato, A. and Bernstein, S. I. (2016). A restrictive cardiomyopathy mutation in an invariant proline at the myosin head/rod junction enhances head flexibility and function, yielding muscle defects in Drosophila. J Mol Biol. PubMed ID: 27107639

    Trujillo, A. S., Ramos, R., Bodmer, R., Bernstein, S. I., Ocorr, K. and Melkani, G. C. (2014). Drosophila as a potential model to ameliorate mutant Huntington-mediated cardiac amyloidosis. Rare Dis 2: e968003. PubMed ID: 26942103

    Melkani, G. C., Trujillo, A. S., Ramos, R., Bodmer, R., Bernstein, S. I. and Ocorr, K. (2013). Huntington's disease induced cardiac amyloidosis is reversed by modulating protein folding and oxidative stress pathways in the Drosophila heart. PLoS Genet 9: e1004024. PubMed ID: 24367279

  • Florence Besse Institute of Biology Valrose, France
    Heim, M., Blot, L. and Besse, F. (2022). An RNA-immunoprecipitation protocol to identify RNAs associated with RNA-binding proteins in cytoplasmic and nuclear Drosophila head fractions. STAR Protoc 3(2): 101415. PubMed ID: 35634357

    Pushpalatha, K. V., Solyga, M., Nakamura, A. and Besse, F. (2022). RNP components condense into repressive RNP granules in the aging brain. Nat Commun 13(1): 2782. PubMed ID: 35589695

    De Graeve, F., Formicola, N., Pushpalatha, K. V., Nakamura, A., Debreuve, E., Descombes, X. and Besse, F. (2022). Detecting Stress Granules in Drosophila Neurons. Methods Mol Biol 2428: 229-242. PubMed ID: 35171483

    Formicola, N., Heim, M., Dufourt, J., Lancelot, A. S., Nakamura, A., Lagha, M. and Besse, F. (2021). Tyramine induces dynamic RNP granule remodeling and translation activation in the Drosophila brain. Elife 10. PubMed ID: 33890854

    Vijayakumar, J., Perrois, C., Heim, M., Bousset, L., Alberti, S. and Besse, F. (2019). The prion-like domain of Drosophila Imp promotes axonal transport of RNP granules in vivo. Nat Commun 10(1): 2593. PubMed ID: 31197139

    Bruckert, H., Marchetti, G., Ramialison, M. and Besse, F. (2015). Drosophila Hrp48 Is Required for Mushroom Body Axon Growth, Branching and Guidance. PLoS One 10: e0136610. PubMed ID: 26313745

    Medioni, C., Ephrussi, A. and Besse, F. (2015). Live imaging of axonal transport in Drosophila pupal brain explants. Nat Protoc 10: 574-584. PubMed ID: 25763834

  • Mónica Bettencourt-Dias Instituto Gulbenkian de Ciência, Oeiras, Portugal
    Nabais, C., Pessoa, D., de-Carvalho, J., van Zanten, T., Duarte, P., Mayor, S., Carneiro, J., Telley, I. A. and Bettencourt-Dias, M. (2021). Plk4 triggers autonomous de novo centriole biogenesis and maturation. J Cell Biol 220(5). PubMed ID: 33760919

    Pimenta-Marques, A., Bento, I., Lopes, C.A., Duarte, P., Jana, S.C. and Bettencourt-Dias, M. (2016). A mechanism for the elimination of the female gamete centrosome in Drosophilanmelanogaster Science [Epub ahead of print]. PubMed ID: 27229142

    Lopes, C. A., Jana, S. C., Cunha-Ferreira, I., Zitouni, S., Bento, I., Duarte, P., Gilberto, S., Freixo, F., Guerrero, A., Francia, M., Lince-Faria, M., Carneiro, J. and Bettencourt-Dias, M. (2015). PLK4 trans-autoactivation controls centriole biogenesis in space. Dev Cell 35: 222-235. PubMed ID: 26481051

    Chen, J. V., Kao, L. R., Jana, S. C., Sivan-Loukianova, E., Mendonca, S., Cabrera, O. A., Singh, P., Cabernard, C., Eberl, D. F., Bettencourt-Dias, M. and Megraw, T. L. (2015). Rootletin organizes the ciliary rootlet to achieve neuron sensory function in Drosophila. J Cell Biol 211: 435-453. PubMed ID: 26483560

    Cunha-Ferreira, I., Bento, I., Pimenta-Marques, A., Jana, S. C., Lince-Faria, M., Duarte, P., Borrego-Pinto, J., Gilberto, S., Amado, T., Brito, D., Rodrigues-Martins, A., Debski, J., Dzhindzhev, N. and Bettencourt-Dias, M. (2013). Regulation of autophosphorylation controls PLK4 self-destruction and centriole number. Curr Biol 23: 2245-2254. PubMed ID: 24184099

    Carvalho-Santos, Z., Machado, P., Alvarez-Martins, I., Gouveia, S. M., Jana, S. C., Duarte, P., Amado, T., Branco, P., Freitas, M. C., Silva, S. T., Antony, C., Bandeiras, T. M. and Bettencourt-Dias, M. (2012). BLD10/CEP135 is a microtubule-associated protein that controls the formation of the flagellum central microtubule pair. Dev Cell 23: 412-424. PubMed ID: 22898782

    Holland, A. J., Fachinetti, D., Da Cruz, S., Zhu, Q., Vitre, B., Lince-Faria, M., Chen, D., Parish, N., Verma, I. M., Bettencourt-Dias, M. and Cleveland, D. W. (2012). Polo-like kinase 4 controls centriole duplication but does not directly regulate cytokinesis. Mol Biol Cell 23: 1838-1845. PubMed ID: 22456511

    Martins, A. R., Machado, P., Callaini, G. and Bettencourt-Dias, M. (2010). Microscopy methods for the study of centriole biogenesis and function in Drosophila. Methods Cell Biol 97: 223-242. PubMed ID: 20719274

  • Esther Betran University of Texas at Arlington
    Mirsalehi, A., Markova, D. N., Eslamieh, M. and Betran, E. (2021). Nuclear transport genes recurrently duplicate by means of RNA intermediates in Drosophila but not in other insects. BMC Genomics 22(1): 876. PubMed ID: 34863092

    Sorourian, M., Kunte, M. M., Domingues, S., Gallach, M., Ozdil, F., Rio, J. and Betran, E. (2014). Relocation Facilitates the Acquisition of Short Cis-Regulatory Regions that Drive the Expression of Retrogenes during Spermatogenesis in Drosophila. Mol Biol Evol. PubMed ID: 24855141

    Calvete, O., Gonzalez, J., Betran, E. and Ruiz, A. (2012). Segmental duplication, microinversion, and gene loss associated with a complex inversion breakpoint region in Drosophila. Mol Biol Evol 29: 1875-1889. PubMed ID: 22328714

    Gallach, M., Chandrasekaran, C. and Betran, E. (2010). Analyses of nuclearly encoded mitochondrial genes suggest gene duplication as a mechanism for resolving intralocus sexually antagonistic conflict in Drosophila. Genome Biol Evol 2: 835-850. PubMed ID: 21037198

  • Eric Betzig Janelia Research Campus
    Wong, K. K. L., Li, T., Fu, T. M., Liu, G., Lyu, C., Kohani, S., Xie, Q., Luginbuhl, D. J., Upadhyayula, S., Betzig, E. and Luo, L. (2023). Origin of wiring specificity in an olfactory map revealed by neuron type-specific, time-lapse imaging of dendrite targeting. Elife 12. PubMed ID: 36975203

    Gao, R., Asano, S. M., Upadhyayula, S., Pisarev, I., Milkie, D. E., Liu, T. L., Singh, V., Graves, A., Huynh, G. H., Zhao, Y., Bogovic, J., Colonell, J., Ott, C. M., Zugates, C., Tappan, S., Rodriguez, A., Mosaliganti, K. R., Sheu, S. H., Pasolli, H. A., Pang, S., Xu, C. S., Megason, S. G., Hess, H., Lippincott-Schwartz, J., Hantman, A., Rubin, G. M., Kirchhausen, T., Saalfeld, S., Aso, Y., Boyden, E. S. and Betzig, E. (2019). Cortical column and whole-brain imaging with molecular contrast and nanoscale resolution. Science 363(6424). PubMed ID: 30655415

    Xie, J., Wooten, M., Tran, V., Chen, B. C., Pozmanter, C., Simbolon, C., Betzig, E. and Chen, X. (2015). Histone H3 Threonine Phosphorylation Regulates Asymmetric Histone Inheritance in the Drosophila Male Germline. Cell 163(4): 920-933. PubMed ID: 26522592

    Billington, N., Beach, J. R., Heissler, S. M., Remmert, K., Guzik-Lendrum, S., Nagy, A., Takagi, Y., Shao, L., Li, D., Yang, Y., Zhang, Y., Barzik, M., Betzig, E., Hammer, J. A., 3rd and Sellers, J. R. (2015). Myosin 18A coassembles with nonmuscle myosin 2 to form mixed bipolar filaments. Curr Biol 25(7): 942-948. PubMed ID: 25754640

    Chen, B. C., Legant, W. R., Wang, K., Shao, L., Milkie, D. E., Davidson, M. W., Janetopoulos, C., Wu, X. S., Hammer, J. A., 3rd, Liu, Z., English, B. P., Mimori-Kiyosue, Y., Romero, D. P., Ritter, A. T., Lippincott-Schwartz, J., Fritz-Laylin, L., Mullins, R. D., Mitchell, D. M., Bembenek, J. N., Reymann, A. C., Bohme, R., Grill, S. W., Wang, J. T., Seydoux, G., Tulu, U. S., Kiehart, D. P. and Betzig, E. (2014). Lattice light-sheet microscopy: imaging molecules to embryos at high spatiotemporal resolution. Science 346(6208): 1257998. PubMed ID: 25342811

  • Utpal Bhadra CCMB Hyderabad
    Das, P. and Bhadra, M. P. (2020). Histone deacetylase (Rpd3) regulates Drosophila early brain development via regulation of Tailless. Open Biol 10(9): 200029. PubMed ID: 32873153

    Mondal, T., Bag, I., Sncvl, P., Garikapati, K. R., Bhadra, U. and Pal Bhadra, M. (2018). Two way controls of apoptotic regulators consign DmArgonaute-1 a better clasp on it. PLoS One 13(1): e0190548. PubMed ID: 29385168

    Mondal, T., Lavanya, A. V., Mallick, A., Dadmala, T. L., Kumbhare, R. M., Bhadra, U. and Bhadra, M. P. (2017). Novel Triazole linked 2-phenyl benzoxazole derivatives induce apoptosis by inhibiting miR-2, miR-13 and miR-14 function in Drosophila melanogaster. Apoptosis [Epub ahead of print]. PubMed ID: 28401354

    Bhadra, U., Gandhi, S. G., Palaparthi, R., Balyan, M. K. and Pal-Bhadra, M. (2016). The maleless gene mitigates global aneuploid effect and evolutionary shift from X to autosomes. FEBS J. [Epub ahead of print] PubMed ID: 27456781

    Pushpavalli, S. N., Sarkar, A., Ramaiah, M. J., Koteswara Rao, G., Bag, I., Bhadra, U. and Pal-Bhadra, M. (2015). Drosophila MOF regulates DIAP1 and induces apoptosis in a JNK dependent pathway. Apoptosis. PubMed ID: 26711898

    Bhadra, U., Mondal, T., Bag, I., Mukhopadhyay, D., Das, P., Parida, B. B., Mainkar, P. S., Reddy, C. R. and Bhadra, M. P. (2015). HDAC inhibitor misprocesses bantam oncomiRNA, but stimulates hid induced apoptotic pathway. Sci Rep 5: 14747. PubMed ID: 26442596

    Gandhi, S. G., Bag, I., Sengupta, S., Pal-Bhadra, M. and Bhadra, U. (2014). Drosophila oncogene Gas41 is RNAi modulator that intersects heterochromatin and siRNA pathway. FEBS J. PubMed ID: 25323651

    Pushpavalli, S. N., Sarkar, A., Bag, I., Hunt, C. R., Ramaiah, M. J., Pandita, T. K., Bhadra, U. and Pal-Bhadra, M. (2013). Argonaute-1 functions as a mitotic regulator by controlling Cyclin B during Drosophila early embryogenesis. FASEB J. PubMed ID: 24165481

    Pushpavalli, S. N., Sarkar, A., Ramaiah, M. J., Chowdhury, D. R., Bhadra, U., Pal-Bhadra, M. (2013) Drosophila MOF controls Checkpoint protein2 and regulates genomic stability during early embryogenesis. BMC Mol Biol 14: 1. PubMed ID: 23347679

    Pushpavalli, S. N., Bag, I., Pal-Bhadra, M. and Bhadra, U. (2012). Drosophila Argonaute-1 is critical for transcriptional cosuppression and heterochromatin formation. Chromosome Res 20: 333-351. PubMed ID: 22476395

  • Krishna Bhat Neuroscience and Cell Biology, UTMB
    Mar, J., Makhijani, K., Flaherty, D. and Bhat, K. M. (2022). Nuclear Prospero allows one-division potential to neural precursors and post-mitotic status to neurons via opposite regulation of Cyclin E. PLoS Genet 18(8): e1010339. PubMed ID: 35939521

    Gaziova, I., Gazi, M., Mar, J. and Bhat, K. M. (2020). Restriction on self-renewing asymmetric division is coupled to terminal asymmetric division in the Drosophila CNS. PLoS Genet 16(9): e1009011. PubMed ID: 32986715

    Manavalan, M. A., Jayasinghe, V. R., Grewal, R. and Bhat, K. M. (2017). The glycosylation pathway is required for the secretion of Slit and for the maintenance of the Slit receptor Robo on axons. Sci Signal 10(484). PubMed ID: 28634210

    Manavalan, M. A., Gaziova, I. and Bhat, K. M. (2013). The Midline Protein Regulates Axon Guidance by Blocking the Reiteration of Neuroblast Rows within the Drosophila Ventral Nerve Cord. PLoS Genet 9: e1004050. PubMed ID: 24385932

    Poddighe, S., Bhat, K. M., Setzu, M. D., Solla, P., Angioy, A. M., Marotta, R., Ruffilli, R., Marrosu, F. and Liscia, A. (2013). Impaired sense of smell in a Drosophila Parkinson's model. PLoS One 8: e73156. PubMed ID: 24009736

    Zhu, Z. and Bhat, K. M. (2011). The Drosophila Hem/Kette/Nap1 protein regulates asymmetric division of neural precursor cells by regulating localization of Inscuteable and Numb. Mech Dev 128: 483-495. PubMed ID: 21996673

    Hafer, N., Xu, S., Bhat, K. M. and Schedl, P. (2011). The Drosophila CPEB protein Orb2 has a novel expression pattern and is important for asymmetric cell division and nervous system function. Genetics 189: 907-921. PubMed ID: 21900268

  • Manzoor Bhat School of Medicine, The University of Texas Health Science Center at San Antonio University web policie
    Chen, S., Venkatesan, A., Lin, Y. Q., Xie, J., Neely, G., Banerjee, S. and Bhat, M. A. (2022). Drosophila Homolog of the Human Carpenter Syndrome Linked Gene, MEGF8, Is Required for Synapse Development and Function. J Neurosci 42(37): 7016-7030. PubMed ID: 35944997

    Banerjee, S., Mino, R. E., Fisher, E. S. and Bhat, M. A. (2017). A versatile genetic tool to study midline glia function in the Drosophila CNS. Dev Biol [Epub ahead of print]. PubMed ID: 28602954

    Banerjee, S., Venkatesan, A. and Bhat, M. A. (2016). Neurexin, neuroligin and wishful thinking coordinate synaptic cytoarchitecture and growth at neuromuscular junctions. Mol Cell Neurosci [Epub ahead of print]. PubMed ID: 27838296

    Mino, R. E., Rogers, S. L., Risinger, A. L., Rohena, C., Banerjee, S. and Bhat, M. A. (2016). Drosophila Ringmaker regulates microtubule stabilization and axonal extension during embryonic development. J Cell Sci. PubMed ID: 27422099

    Chen, Y. C., Lin, Y. Q., Banerjee, S., Venken, K., Li, J., Ismat, A., Chen, K., Duraine, L., Bellen, H. J. and Bhat, M. A. (2012). Drosophila neuroligin 2 is required presynaptically and postsynaptically for proper synaptic differentiation and synaptic transmission. J Neurosci 32: 16018-16030. PubMed ID: 23136438

    Choi, W., Jung, K. C., Nelson, K. S., Bhat, M. A., Beitel, G. J., Peifer, M. and Fanning, A. S. (2011). The single Drosophila ZO-1 protein Polychaetoid regulates embryonic morphogenesis in coordination with Canoe/afadin and Enabled. Mol Biol Cell 22: 2010-2030. PubMed ID: 21508316

  • Sharmila Bhattacharya Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Biological and Physical Sciences Division, NASA Headquarters, Washington DC 20024, USA.
    Mhatre, S. D., Iyer, J., Petereit, J., Dolling-Boreham, R. M., Tyryshkina, A., Paul, A. M., Gilbert, R., Jensen, M., Woolsey, R. J., Anand, S., Sowa, M. B., Quilici, D. R., Costes, S. V., Girirajan, S. and Bhattacharya, S. (2022). Artificial gravity partially protects space-induced neurological deficits in Drosophila melanogaster. Cell Rep 40(10): 111279. PubMed ID: 36070701

    psaro, J. J., O'Brien, P. A., Bhattacharya, S., Palmer, A. G., 3rd and Joshua-Tor, L. (2021). Asterix/Gtsf1 links tRNAs and piRNA silencing of retrotransposons. Cell Rep 34(13): 108914. PubMed ID: 33789107

    Walls, S., Diop, S., Birse, R., Elmen, L., Gan, Z., Kalvakuri, S., Pineda, S., Reddy, C., Taylor, E., Trinh, B., Vogler, G., Zarndt, R., McCulloch, A., Lee, P., Bhattacharya, S., Bodmer, R. and Ocorr, K. (2020). Prolonged Exposure to Microgravity Reduces Cardiac Contractility and Initiates Remodeling in Drosophila. Cell Rep 33(10): 108445. PubMed ID: 33242407

  • Xiaolin Bi Nantong University - School of Medicine
    Xu, Q., Liu, J., Du, X., Xue, D., Li, D. and Bi, X. (2023). Long noncoding RNA CR46040 is essential for injury-stimulated regeneration of intestinal stem cells in Drosophila. Genetics 224(1). PubMed ID: 36930573

    Liu, J., Jin, T., Ran, L., Zhao, Z., Zhu, R., Xie, G. and Bi, X. (2022). Profiling ATM regulated genes in Drosophila at physiological condition and after ionizing radiation. Hereditas 159(1): 41. PubMed ID: 36271387

    Li, D., Ge, Y., Zhao, Z., Zhu, R., Wang, X. and Bi, X. (2021). Distinct and Coordinated Regulation of Small Non-coding RNAs by E2f1 and p53 During Drosophila Development and in Response to DNA Damage. Front Cell Dev Biol 9: 695311. PubMed ID: 33057338

  • Sharon Bickel Department of Biological Sciences, Dartmouth University, Hanover, NH
    Perkins, A. T., Greig, M. M., Sontakke, A. A., Peloquin, A. S., McPeek, M. A. and Bickel, S. E. (2019). Increased levels of superoxide dismutase suppress meiotic segregation errors in aging oocytes. Chromosoma. PubMed ID: 31037468

    Perkins, A. T., Das, T. M., Panzera, L. C. and Bickel, S. E. (2016). Oxidative stress in oocytes during midprophase induces premature loss of cohesion and chromosome segregation errors. Proc Natl Acad Sci U S A 113: E6823-E6830. PubMed ID: 27791141

    Giauque, C. C. and Bickel, S. E. (2016). Heterochromatin-Associated Proteins HP1a and Piwi Collaborate to Maintain the Association of Achiasmate Homologs in Drosophila Oocytes. Genetics 203: 173-189. PubMed ID: 26984058

    Weng, K. A., Jeffreys, C. A. and Bickel, S. E. (2014). Rejuvenation of meiotic cohesion in oocytes during prophase I is required for chiasma maintenance and accurate chromosome segregation. PLoS Genet 10: e1004607. PubMed ID: 25211017

    Fisher, W. W., Li, J. J., Hammonds, A. S., Brown, J. B., Pfeiffer, B. D., Weiszmann, R., MacArthur, S., Thomas, S., Stamatoyannopoulos, J. A., Eisen, M. B., Bickel, P. J., Biggin, M. D. and Celniker, S. E. (2012). DNA regions bound at low occupancy by transcription factors do not drive patterned reporter gene expression in Drosophila. Proc Natl Acad Sci U S A 109: 21330-21335. PubMed ID: 23236164

  • Ashok Bidwai West Virginia University, Morgantown
    Jozwick, L. M. and Bidwai, A. P. (2022). Protein kinase CK2 phosphorylates a conserved motif in the Notch effector E(spl)-Mgamma. Mol Cell Biochem. PubMed ID: 36087252

    Majot, A. T. and Bidwai, A. P. (2017). Analysis of transient hypermorphic activity of E(spl)D during R8 specification. PLoS One 12(10): e0186439. PubMed ID: 29036187

    Bose, A., Majot, A. T. and Bidwai, A. P. (2014). The Ser/Thr Phosphatase PP2A Regulatory Subunit Widerborst Inhibits Notch Signaling. PLoS One 9: e101884. PubMed ID: 25006677

    Kahali, B., Kim, J. E., Karandikar, U., Bishop, C. P. and Bidwai, A. P. (2010). Evidence that the C-terminal domain (CtD) autoinhibits neural repression by Drosophila E(spl)M8. Genesis 48: 44-55. PubMed ID: 20014344

    Kunttas-Tatli, E., Bose, A., Kahali, B., Bishop, C. P. and Bidwai, A. P. (2009). Functional dissection of Timekeeper (Tik) implicates opposite roles for CK2 and PP2A during Drosophila neurogenesis. Genesis 47: 647-658. PubMed ID: 19536808

  • Ethan Bier Division of Biological Sciences, U. C. San Diego
    Guichard, A., Lu, S., Kanca, O., Bressan, D., Huang, Y., Ma, M., Sanz Juste, S., Andrews, J. C., Jay, K. L., Sneider, M., Schwartz, R., Huang, M. C., Bei, D., Pan, H., Ma, L., Lin, W. W., Auradkar, A., Bhagwat, P., Park, S., Wan, K. H., Ohsako, T., Takano-Shimizu, T., Celniker, S. E., Wangler, M. F., Yamamoto, S., Bellen, H. J. and Bier, E. (2023). A comprehensive Drosophila resource to identify key functional interactions between SARS-CoV-2 factors and host proteins. Cell Rep 42(8): 112842. PubMed ID: 37480566

    Kaduskar, B., Kushwah, R. B. S., Auradkar, A., Guichard, A., Li, M., Bennett, J. B., Julio, A. H. F., Marshall, J. M., Montell, C. and Bier, E. (2022). Reversing insecticide resistance with allelic-drive in Drosophila melanogaster. Nat Commun 13(1): 291. PubMed ID: 35022402

    Terradas, G., Buchman, A. B., Bennett, J. B., Shriner, I., Marshall, J. M., Akbari, O. S. and Bier, E. (2021). Inherently confinable split-drive systems in Drosophila. Nat Commun 12(1): 1480. PubMed ID: 33674604

    Xu, X. S., Bulger, E. A., Gantz, V. M., Klanseck, C., Heimler, S. R., Auradkar, A., Bennett, J. B., Miller, L. A., Leahy, S., Juste, S. S., Buchman, A., Akbari, O. S., Marshall, J. M. and Bier, E. (2020). Active Genetic Neutralizing Elements for Halting or Deleting Gene Drives. Mol Cell. PubMed ID: 32949493

    Schwartz, R., Guichard, A., Franc, N. C., Roy, S. and Bier, E. (2020). A Drosophila Model for Clostridium difficile Toxin CDT Reveals Interactions with Multiple Effector Pathways. iScience 23(2): 100865. PubMed ID: 32058973

    Guichard, A., Haque, T., Bobik, M., Xu, X. S., Klanseck, C., Kushwah, R. B. S., Berni, M., Kaduskar, B., Gantz, V. M. and Bier, E. (2019). Efficient allelic-drive in Drosophila. Nat Commun 10(1): 1640. PubMed ID: 30967548

    Xu, X. S., Gantz, V. M., Siomava, N. and Bier, E. (2017). CRISPR/Cas9 and active genetics-based trans-species replacement of the endogenous Drosophila kni-L2 CRM reveals unexpected complexity. Elife 6. PubMed ID: 29274230

    Gantz, V. M. and Bier, E. (2015). Genome editing. The mutagenic chain reaction: a method for converting heterozygous to homozygous mutations. Science 348: 442-444. PubMed ID: 25908821

    Esteves, F. F., Springhorn, A., Kague, E., Taylor, E., Pyrowolakis, G., Fisher, S. and Bier, E. (2014). BMPs Regulate msx Gene Expression in the Dorsal Neuroectoderm of Drosophila and Vertebrates by Distinct Mechanisms. PLoS Genet 10: e1004625. PubMed ID: 25210771

    Grossman, T. R., Gamliel, A., Wessells, R. J., Taghli-Lamallem, O., Jepsen, K., Ocorr, K., Korenberg, J. R., Peterson, K. L., Rosenfeld, M. G., Bodmer, R. and Bier, E. (2011). Over-expression of DSCAM and COL6A2 cooperatively generates congenital heart defects. PLoS Genet 7: e1002344. PubMed ID: 22072978

  • Mariann Bienz Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge
    Mieszczanek, J., Strutt, H., Rutherford, T. J., Strutt, D., Bienz, M. and Gammons, M. V. (2022). Selective function of the PDZ domain of Dishevelled in noncanonical Wnt signalling. J Cell Sci 135(11). PubMed ID: 35542970

    Flack, J. E., Mieszczanek, J., Novcic, N. and Bienz, M. (2017). Wnt-Dependent Inactivation of the Groucho/TLE Co-repressor by the HECT E3 Ubiquitin Ligase Hyd/UBR5. Mol Cell 67(2): 181-193.e185. PubMed ID: 28689657

    van Tienen, L. M., Mieszczanek, J., Fiedler, M., Rutherford, T. J. and Bienz, M. (2017). Constitutive scaffolding of multiple Wnt enhanceosome components by Legless/BCL9. Elife 6. PubMed ID: 28296634

    Fiedler, M., Graeb, M., Mieszczanek, J., Rutherford, T. J., Johnson, C. M. and Bienz, M. (2015). An ancient Pygo-dependent Wnt enhanceosome integrated by Chip/LDB-SSDP. Elife 4. PubMed ID: 26312500

    Miller, T. C., Mieszczanek, J., Sanchez-Barrena, M. J., Rutherford, T. J., Fiedler, M. and Bienz, M. (2013). Evolutionary Adaptation of the Fly Pygo PHD Finger toward Recognizing Histone H3 Tail Methylated at Arginine 2. Structure. PubMed ID: 24183574

    Mendoza-Topaz, C., Mieszczanek, J. and Bienz, M. (2011). The Adenomatous polyposis coli tumour suppressor is essential for Axin complex assembly and function and opposes Axin's interaction with Dishevelled. Open Biol 1: 110013. PubMed ID: 22645652

    Fiedler, M., Mendoza-Topaz, C., Rutherford, T. J., Mieszczanek, J. and Bienz, M. (2011). Dishevelled interacts with the DIX domain polymerization interface of Axin to interfere with its function in down-regulating beta-catenin. Proc Natl Acad Sci U S A 108: 1937-1942. PubMed ID: 21245303

  • Mark Biggin Lawrence Berkeley Lab
    Arbel, H., Basu, S., Fisher, W. W., Hammonds, A. S., Wan, K. H., Park, S., Weiszmann, R., Booth, B. W., Keranen, S. V., Henriquez, C., Shams Solari, O., Bickel, P. J., Biggin, M. D., Celniker, S. E. and Brown, J. B. (2019). Exploiting regulatory heterogeneity to systematically identify enhancers with high accuracy. Proc Natl Acad Sci U S A 116(3): 900-908. PubMed ID: 30598455

    Knowles, D. W. and Biggin, M. D. (2013). Building quantitative, three-dimensional atlases of gene expression and morphology at cellular resolution. Wiley Interdiscip Rev Dev Biol 2: 767-779. PubMed ID: 24123936

    Fisher, W. W., Li, J. J., Hammonds, A. S., Brown, J. B., Pfeiffer, B. D., Weiszmann, R., Macarthur, S., Thomas, S., Stamatoyannopoulos, J. A., Eisen, M. B., Bickel, P. J., Biggin, M. D. and Celniker, S. E. (2012). DNA regions bound at low occupancy by transcription factors do not drive patterned reporter gene expression in Drosophila. Proc. Natl. Acad. Sci. [Epub ahead of print]. PubMed ID: 23236164

  • David Bilder Department of Molecular and Cell Biology, University of California, Berkeley
    Hsi, T. C., Ong, K. L., Sepers, J. J., Kim, J. and Bilder, D. (2023). Systemic coagulopathy promotes host lethality in a new Drosophila tumor model. Curr Biol 33(14): 3002-3010.e3006. PubMed ID: 37354901

    Ku, H. Y., Harris, L. K. and Bilder, D. (2023). Specialized cells that sense tissue mechanics to regulate Drosophila morphogenesis. Dev Cell. PubMed ID: 36708706

    Gerlach, S. U., de Vreede, G. and Bilder, D. (2022). PTP10D-mediated cell competition is not obligately required for elimination of polarity-deficient clones. Biol Open 11(11). PubMed ID: 36355597

    de Vreede, G., Gerlach, S. U. and Bilder, D. (2022). Epithelial monitoring through ligand-receptor segregation ensures malignant cell elimination. Science 376(6590): 297-301. PubMed ID: 35420935

    Kim, J., Chuang, H. C., Wolf, N. K., Nicolai, C. J., Raulet, D. H., Saijo, K. and Bilder, D. (2021). Tumor-induced disruption of the blood-brain barrier promotes host death. Dev Cell 56(19): 2712-2721. PubMed ID: 34496290

    Sharp, K. A., Khoury, M. J., Wirtz-Peitz, F. and Bilder, D. (2021). Evidence for a nuclear role for Drosophila Dlg as a regulator of the NURF complex. Mol Biol Cell 32(21): ar23. PubMed ID: 34495684

    Khoury, M. J. and Bilder, D. (2020). Distinct activities of Scrib module proteins organize epithelial polarity. Proc Natl Acad Sci U S A 117(21): 11531-11540. PubMed ID: 32414916

    Chen, D. Y., Crest, J., Streichan, S. J. and Bilder, D. (2019). Extracellular matrix stiffness cues junctional remodeling for 3D tissue elongation. Nat Commun 10(1): 3339. PubMed ID: 31350387

    de Vreede, G., Morrison, H. A., Houser, A. M., Boileau, R. M., Andersen, D., Colombani, J. and Bilder, D. (2018). A Drosophila tumor suppressor gene prevents tonic TNF signaling through receptor N-glycosylation. Dev Cell 45(5): 595-605.e594. PubMed ID: 29870719

    Kim, J., Bilder, D. and Neufeld, T. P. (2018). Mechanical stress regulates insulin sensitivity through integrin-dependent control of insulin receptor localization. Genes Dev 32(2): 156-164. PubMed ID: 29440263

    Chen, D. Y., Crest, J. and Bilder, D. (2017). A cell migration tracking tool supports coupling of tissue rotation to elongation. Cell Rep 21(3): 559-569. PubMed ID: 29045826

    Crest, J., Diz-Munoz, A., Chen, D. Y., Fletcher, D. A. and Bilder, D. (2017). Organ sculpting by patterned extracellular matrix stiffness. Elife 6. PubMed ID: 28653906

    Katheder, N. S., Khezri, R., O'Farrell, F., Schultz, S. W., Jain, A., Rahman, M. M., Schink, K. O., Theodossiou, T. A., Johansen, T., Juhasz, G., Bilder, D., Brech, A., Stenmark, H. and Rusten, T. E. (2017). Microenvironmental autophagy promotes tumour growth. Nature. PubMed ID: 28077876

  • Jean-Christophe Billeter Centre for Behaviour and Neurosciences, University of Groningen
    Verschut, T. A., Ng, R., Doubovetzky, N. P., Le Calvez, G., Sneep, J. L., Minnaard, A. J., Su, C. Y., Carlsson, M. A., Wertheim, B. and Billeter, J. C. (2023). Aggregation pheromones have a non-linear effect on oviposition behavior in Drosophila melanogaster. Nat Commun 14(1): 1544. PubMed ID: 36941252

    Bailly, T. P. M., Kohlmeier, P., Etienne, R. S., Wertheim, B. and Billeter, J. C. (2023). Social modulation of oogenesis and egg laying in Drosophila melanogaster. Curr Biol 33(14): 2865-2877.e2864. PubMed ID: 37369209

    Kohlmeier, P., Zhang, Y., Gorter, J. A., Su, C. Y. and Billeter, J. C. (2021). Mating increases Drosophila melanogaster females' choosiness by reducing olfactory sensitivity to a male pheromone. Nat Ecol Evol. PubMed ID: 34155384

    Soto-Padilla, A., Ruijsink, R., Span, M., van Rijn, H. and Billeter, J. C. (2018). An Automated Method to Determine the Performance of Drosophila in Response to Temperature Changes in Space and Time. J Vis Exp(140). PubMed ID: 30371661

    Laturney, M., van Eijk, R. and Billeter, J. C. (2018). Last male sperm precedence is modulated by female remating rate in Drosophila melanogaster. Evol Lett 2(3): 180-189. PubMed ID: 30283675

    Soto-Padilla, A., Ruijsink, R., Sibon, O. C. M., van Rijn, H. and Billeter, J. C. (2018). Thermosensory perception regulates speed of movement in response to temperature changes in Drosophila melanogaster. J Exp Biol. PubMed ID: 29650755

    Laturney, M. and Billeter, J.C. (2016). Drosophila females restore their attractiveness after mating by removing male anti-aphrodisiac pheromones. Nat Commun 7: 12322. PubMed ID: 27484362

    Dumenil, C., Woud, D., Pinto, F., Alkema, J. T., Jansen, I., Van Der Geest, A. M., Roessingh, S. and Billeter, J. C. (2016). Pheromonal Cues Deposited by Mated Females Convey Social Information about Egg-Laying Sites in Drosophila Melanogaster. J Chem Ecol [Epub ahead of print]. PubMed ID: 26994611

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

    Laturney, M. and Billeter, J. C. (2014). Neurogenetics of Female Reproductive Behaviors in Drosophila melanogaster. Adv Genet 85: 1-108. PubMed ID: 24880733

  • James Birchler Division of Biological Sciences, University of Missouri, Columbia
    Birchler, J. A. (2014). Does ectopic cell death cause somatic mutations in the neighboring cells by activating transposons? Mob Genet Elements 4: e28040. PubMed ID: 24567848

    Sun, L., Johnson, A. F., Li, J., Lambdin, A. S., Cheng, J. and Birchler, J. A. (2013). Differential effect of aneuploidy on the X chromosome and genes with sex-biased expression in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24062456

    Xie, W., Donohue, R. C. and Birchler, J. A. (2013). Quantitatively Increased Somatic Transposition of Transposable Elements in Drosophila Strains Compromised for RNAi. PLoS One 8: e72163. PubMed ID: 23940807

    Sun, L., Johnson, A. F., Donohue, R. C., Li, J., Cheng, J., Birchler, J. A. (2013) Dosage compensation and inverse effects in triple X metafemales of Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 23589863

    Sun, L., Fernandez, H. R., Donohue, R. C., Li, J., Cheng, J., Birchler, J. A. (2013) Male-specific lethal complex in Drosophila counteracts histone acetylation and does not mediate dosage compensation. Proc Natl Acad Sci U S A. PubMed ID: 23382189

    Xie, W. and Birchler, J. A. (2012). Identification of Inverse Regulator-a (Inr-a) as Synonymous with Pre-mRNA Cleavage Complex II Protein (Pcf11) in Drosophila. G3 (Bethesda) 2: 701-706. PubMed ID: 22690379

    Birchler, J. A. and Xie, W. (2011). Reflections on the inhibition of RNAi by cell death signaling. Fly (Austin) 5. PubMed ID: 21795852

  • Serge Birman Laboratoire de Neurobiologie, ESPCI ParisTech
    Dulac, A., Issa, A. R., Sun, J., Matassi, G., Jonas, C., Cherif-Zahar, B., Cattaert, D. and Birman, S. (2021). A Novel Neuron-Specific Regulator of the V-ATPase in Drosophila. eNeuro 8(5). PubMed ID: 34620624

    Dulac, A., Issa, A. R., Sun, J., Matassi, G., Jonas, C., Cherif-Zahar, B., Cattaert, D. and Birman, S. (2021). A Novel Neuron-Specific Regulator of the V-ATPase in Drosophila. eNeuro 8(5). PubMed ID: 34620624

    Yon, M., Decoville, M., Sarou-Kanian, V., Fayon, F. and Birman, S. (2020). Spatially-resolved metabolic profiling of living Drosophila in neurodegenerative conditions using (1)H magic angle spinning NMR. Sci Rep 10(1): 9516. PubMed ID: 32528106

    Lecompte, M., Cattaert, D., Vincent, A., Birman, S. and Cherif-Zahar, B. (2019). The Drosophila ammonium transporter Rh50 is required for integrity of larval muscles and neuromuscular system. J Comp Neurol. PubMed ID: 31273786

    Hajji, K., Mteyrek, A., Sun, J., Cassar, M., Mezghani, S., Leprince, J., Vaudry, D., Masmoudi-Kouki, O. and Birman, S. (2019). Neuroprotective effects of PACAP against paraquat-induced oxidative stress in the Drosophila central nervous system. Hum Mol Genet. PubMed ID: 30715303

    Issa, A. R., Sun, J., Petitgas, C., Mesquita, A., Dulac, A., Robin, M., Mollereau, B., Jenny, A., Cherif-Zahar, B. and Birman, S. (2018). The lysosomal membrane protein LAMP2A promotes autophagic flux and prevents SNCA-induced Parkinson disease-like symptoms in the Drosophila brain. Autophagy. PubMed ID: 29989488

    Sun, J., Xu, A. Q., Giraud, J., Poppinga, H., Riemensperger, T., Fiala, A. and Birman, S. (2018). Neural control of startle-induced locomotion by the mushroom bodies and associated neurons in Drosophila. Front Syst Neurosci 12: 6. PubMed ID: 29643770

    Arthaut, L. D., Jourdan, N., Mteyrek, A., Procopio, M., El-Esawi, M., d'Harlingue, A., Bouchet, P. E., Witczak, J., Ritz, T., Klarsfeld, A., Birman, S., Usselman, R. J., Hoecker, U., Martino, C. F. and Ahmad, M. (2017). Blue-light induced accumulation of reactive oxygen species is a consequence of the Drosophila cryptochrome photocycle. PLoS One 12(3): e0171836. PubMed ID: 28296892

    Vaccaro, A., Issa, A. R., Seugnet, L., Birman, S. and Klarsfeld, A. (2017). Drosophila Clock Is Required in Brain Pacemaker Neurons to Prevent Premature Locomotor Aging Independently of Its Circadian Function. PLoS Genet 13(1): e1006507. PubMed ID: 28072817

    Cichewicz, K., Garren, E. J., Adiele, C., Aso, Y., Wang, Z., Wu, M., Birman, S., Rubin, G. M. and Hirsh, J. (2016). A new brain dopamine deficient Drosophila and its pharmacological and genetic Rescue. Genes Brain Behav [Epub ahead of print]. PubMed ID: 27762066

  • Marcus Bischoff University of St Andrews, England
    Pulido Companys, P., Norris, A. and Bischoff, M. (2020). Coordination of cytoskeletal dynamics and cell behaviour during Drosophila abdominal morphogenesis. J Cell Sci 133(6). PubMed ID: 32229579

    Topfer, U., Bischoff, M. C., Bartkuhn, M. and Holz, A. (2019). Serpent/dGATAb regulates Laminin B1 and Laminin B2 expression during Drosophila embryogenesis. Sci Rep 9(1): 15910. PubMed ID: 31685844

    Rothenbusch-Fender, S., Fritzen, K., Bischoff, M. C., Buttgereit, D., Oenel, S. F. and Renkawitz-Pohl, R. (2017). Myotube migration to cover and shape the testis of Drosophila depends on Heartless, Cadherin/Catenin, and myosin II. Biol Open 6(12): 1876-1888. PubMed ID: 29122742

  • Benoit Biteau Department of Biomedical Genetics, University of Rochester, Rochester, NY
    Sreejith, P., Malik, S., Kim, C. and Biteau, B. (2022). Imp interacts with Lin28 to regulate adult stem cell proliferation in the Drosophila intestine. PLoS Genet 18(9): e1010385. PubMed ID: 36070313

    Meng, F. W., Rojas Villa, S. E. and Biteau, B. (2020). Sox100B Regulates Progenitor-Specific Gene Expression and Cell Differentiation in the Adult Drosophila Intestine. Stem Cell Reports 14(2): 226-240. PubMed ID: 32032550

    Rojas Villa, S. E., Meng, F. W. and Biteau, B. (2019). zfh2 controls progenitor cell activation and differentiation in the adult Drosophila intestinal absorptive lineage. PLoS Genet 15(12): e1008553. PubMed ID: 31841513

    Allbee, A. W., Rincon-Limas, D. E. and Biteau, B. (2018). Lmx1a is required for the development of the ovarian stem cell niche in Drosophila. Development 145(8). PubMed ID: 29615466

    McClelland, L., Jasper, H. and Biteau, B. (2017). Tis11 mediated mRNA decay promotes the reacquisition of Drosophila intestinal stem cell quiescence. Dev Biol 426(1): 8-16. PubMed ID: 28445691

    Meng, F. W. and Biteau, B. (2015). A Sox Transcription Factor Is a Critical Regulator of Adult Stem Cell Proliferation in the Drosophila Intestine. Cell Rep 13: 906-914. PubMed ID: 26565904

    Biteau, B. and Jasper, H. (2014). Slit/Robo signaling regulates cell fate decisions in the intestinal stem cell lineage of Drosophila. Cell Rep 7: 1867-1875. PubMed ID: 24931602

    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: 1250-1261. PubMed ID: 24035390

  • Seth Blair Department of Zoology, University of Wisconsin
    Wang, X., Zhang, Y. and Blair, S. S. (2019). Fat-regulated adaptor protein Dlish binds the growth suppressor Expanded and controls its stability and ubiquitination. Proc Natl Acad Sci U S A. PubMed ID: 30606799

    Zhang, Y., Wang, X., Matakatsu, H., Fehon, R. and Blair, S.S. (2016). The novel SH3 domain protein Dlish/CG10933 mediates fat signaling in Drosophila by binding and regulating Dachs. Elife 5. PubMed ID: 27692068

    Avanesov, A. and Blair, S. S. (2013). The Drosophila WIF1 homolog Shifted maintains glypican-independent Hedgehog signaling and interacts with the Hedgehog co-receptors Ihog and Boi. Development 140: 107-116. PubMed ID: 23154411

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

  • Todd Blankenship Department of Biological Sciences, University of Denver
    Miao, H., Millage, M., Rollins, K. R. and Blankenship, J. T. (2023). A Rab39-Klp98A-Rab35 endocytic recycling pathway is essential for rapid Golgi-dependent furrow ingression. Development 150(16). PubMed ID: 37590130

    Henry, S. M., Xie, Y., Rollins, K. R. and Blankenship, J. T. (2022). Sponge/DOCK-dependent regulation of F-actin networks directing cortical cap behaviors and syncytial furrow ingression. Dev Biol 491: 82-93. PubMed ID: 36067836

    Vanderleest, T. E., Xie, Y., Smits, C., Blankenship, J. T. and Loerke, D. (2022). Interface extension is a continuum property suggesting a linkage between AP contractile and DV lengthening processes. Mol Biol Cell: mbcE21070352. PubMed ID: 36129772

    Miao, H., Vanderleest, T. E., Budhathoki, R., Loerke, D. and Blankenship, J. T. (2021). A PtdIns(3,4,5)P(3) dispersal switch engages cell ratcheting at specific cell surfaces. Dev Cell 56(18): 2579-2591.e2574. PubMed ID: 34525342

    Vanderleest, T. E., Smits, C. M., Xie, Y., Jewett, C. E., Blankenship, J. T. and Loerke, D. (2018). Vertex sliding drives intercalation by radial coupling of adhesion and actomyosin networks during Drosophila germband extension. Elife 7. PubMed ID: 29985789

    Xie, Y. and Blankenship, J. T. (2018). Differentially-dimensioned furrow formation by zygotic gene expression and the MBT. PLoS Genet 14(1): e1007174. PubMed ID: 29337989

    Jewett, C. E., Vanderleest, T. E., Miao, H., Xie, Y., Madhu, R., Loerke, D. and Blankenship, J. T. (2017). Planar polarized Rab35 functions as an oscillatory ratchet during cell intercalation in the Drosophila epithelium. Nat Commun 8(1): 476. PubMed ID: 28883443

    Patel, P. H., Barbee, S. A. and Blankenship, J. T. (2016). GW-Bodies and P-Bodies Constitute Two Separate Pools of Sequestered Non-Translating RNAs. PLoS One 11: e0150291. PubMed ID: 26930655

    Mavor, L. M., Miao, H., Zuo, Z., Holly, R. M., Xie, Y., Loerke, D. and Blankenship, J. T. (2016). Rab8 directs furrow ingression and membrane addition during epithelial formation in Drosophila melanogaster. Development [Epub ahead of print]. PubMed ID: 26839362

    Giansanti, M. G., Vanderleest, T. E., Jewett, C. E., Sechi, S., Frappaolo, A., Fabian, L., Robinett, C. C., Brill, J. A., Loerke, D., Fuller, M. T. and Blankenship, J. T. (2015). Exocyst-dependent membrane addition is required for anaphase cell elongation and cytokinesis in Drosophila. PLoS Genet 11: e1005632. PubMed ID: 26528720

  • Justin Blau Department of Biology, NYU
    Cavey, M., Collins, B., Bertet, C. and Blau, J. (2016). Circadian rhythms in neuronal activity propagate through output circuits. Nat Neurosci [Epub ahead of print]. PubMed ID: 26928065

    Petsakou, A., Sapsis, T.P. and Blau, J. (2015). Circadian rhythms in Rho1 activity regulate neuronal plasticity and network hierarchy. Cell [Epub ahead of print]. PubMed ID: 26234154

    Collins, B., Kaplan, H. S., Cavey, M., Lelito, K. R., Bahle, A. H., Zhu, Z., Macara, A. M., Roman, G., Shafer, O. T. and Blau, J. (2014). Differentially timed extracellular signals synchronize pacemaker neuron clocks. PLoS Biol 12: e1001959. PubMed ID: 25268747

    Keene, A. C., Mazzoni, E. O., Zhen, J., Younger, M. A., Yamaguchi, S., Blau, J., Desplan, C. and Sprecher, S. G. (2011). Distinct visual pathways mediate Drosophila larval light avoidance and circadian clock entrainment. J Neurosci 31: 6527-6534. PubMed ID: 21525293

    Dahdal, D., Reeves, D. C., Ruben, M., Akabas, M. H. and Blau, J. (2010). Drosophila pacemaker neurons require g protein signaling and GABAergic inputs to generate twenty-four hour behavioral rhythms. Neuron 68: 964-977. PubMed ID: 21145008

  • Franziska Bleichert, Yale School of Medicine
    Schmidt, J. M. and Bleichert, F. (2020). Structural mechanism for replication origin binding and remodeling by a metazoan origin recognition complex and its co-loader Cdc6. Nat Commun 11(1): 4263. PubMed ID: 32848132

    Bleichert, F., Leitner, A., Aebersold, R., Botchan, M. R. and Berger, J. M. (2018). Conformational control and DNA-binding mechanism of the metazoan origin recognition complex. Proc Natl Acad Sci U S A 115(26): E5906-E5915. PubMed ID: 29899147

    Bleichert, F., Botchan, M. R. and Berger, J. M. (2015). Crystal structure of the eukaryotic origin recognition complex. Nature 519(7543): 321-326. PubMed ID: 25762138

  • Mark Blows School of Biological Sciences, University of Queensland, Australia
    Collet, J. M., McGuigan, K., Allen, S. L., Chenoweth, S. F. and Blows, M. W. (2018). Mutational Pleiotropy and the Strength of Stabilizing Selection Within and Between Functional Modules of Gene Expression. Genetics. PubMed ID: 29437825

    McGuigan, K., Collet, J. M., Allen, S. L., Chenoweth, S. F. and Blows, M. W. (2014). Pleiotropic Mutations Are Subject to Strong Stabilizing Selection. Genetics. PubMed ID: 24793289

    McGuigan, K., Blows, M. W. (2013) Joint allelic effects on fitness and metric traits. Evolution 67: 1131-1142. PubMed ID: 23550761

    Aguirre, J. D., Hine, E., McGuigan, K. and Blows, M. W. (2013). Comparing G: multivariate analysis of genetic variation in multiple populations. Heredity (Edinb). PubMed ID: 23486079

    Delcourt, M., Blows, M. W., Aguirre, J. D. and Rundle, H. D. (2012). Evolutionary optimum for male sexual traits characterized using the multivariate Robertson-Price Identity. Proc Natl Acad Sci U S A 109: 10414-10419. PubMed ID: 22615415

    McGraw, E. A., Ye, Y. H., Foley, B., Chenoweth, S. F., Higgie, M., Hine, E. and Blows, M. W. (2011). High-dimensional variance partitioning reveals the modular genetic basis of adaptive divergence in gene expression during reproductive character displacement. Evolution 65: 3126-3137. PubMed ID: 22023580

  • Justin Blumenstiel University of Kansas, Lawrence
    Miller, D. E., Dorador, A. P., Van Vaerenberghe, K., Li, A., Grantham, E. K., Cerbin, S., Cummings, C., Barragan, M., Egidy, R. R., Scott, A. R., Hall, K. E., Perera, A., Gilliland, W. D., Hawley, R. S. and Blumenstiel, J. P. (2023). Off-target piRNA gene silencing in Drosophila melanogaster rescued by a transposable element insertion. PLoS Genet 19(2): e1010598. PubMed ID: 36809339

    Hemmer, L. W., Dias, G. B., Smith, B., Van Vaerenberghe, K., Howard, A., Bergman, C. M. and Blumenstiel, J. P. (2020). Hybrid dysgenesis in Drosophila virilis results in clusters of mitotic recombination and loss-of-heterozygosity but leaves meiotic recombination unaltered. Mob DNA 11: 10. PubMed ID: 32082426

    Erwin, A. A. and Blumenstiel, J. P. (2019). Aging in the Drosophila ovary: contrasting changes in the expression of the piRNA machinery and mitochondria but no global release of transposable elements. BMC Genomics 20(1): 305. PubMed ID: 31014230

    Hemmer, L. W. and Blumenstiel, J. P. (2016). Holding it together: rapid evolution and positive selection in the synaptonemal complex of Drosophila. BMC Evol Biol 16: 91. PubMed ID: 27150275

    Miller, D. E., Smith, C. B., Yeganeh Kazemi, N., Cockrell, A. J., Arvanitakis, A. V., Blumenstiel, J. P., Jaspersen, S. L. and Hawley, R. S. (2016). Whole-Genome Analysis of Individual Meiotic Events in Drosophila melanogaster Reveals that Noncrossover Gene Conversions are Insensitive to Interference and the Centromere Effect. Genetics. PubMed ID: 26944917

    Erwin, A. A., Galdos, M. A., Wickersheim, M. L., Harrison, C. C., Marr, K. D., Colicchio, J. M. and Blumenstiel, J. P. (2015). piRNAs are associated with diverse transgenerational effects on gene and transposon expression in a hybrid dysgenic syndrome of D. virilis. PLoS Genet 11: e1005332. PubMed ID: 26241928

    Collins, K. A., Unruh, J. R., Slaughter, B. D., Yu, Z., Lake, C. M., Nielsen, R. J., Box, K. S., Miller, D. E., Blumenstiel, J. P., Perera, A. G., Malanowski, K. E. and Hawley, R. S. (2014). Corolla is a novel protein that contributes to the architecture of the synaptonemal complex of Drosophila. Genetics 198: 219-228. PubMed ID: 24913682

    Wickersheim, M. L. and Blumenstiel, J. P. (2013). Terminator oligo blocking efficiently eliminates rRNA from Drosophila small RNA sequencing libraries. Biotechniques 55: 269-272. PubMed ID: 24215643

    Miller, D. E., Takeo, S., Nandanan, K., Paulson, A., Gogol, M. M., Noll, A. C., Perera, A. G., Walton, K. N., Gilliland, W. D., Li, H., Staehling, K. K., Blumenstiel, J. P. and Hawley, R. S. (2012). A Whole-Chromosome Analysis of Meiotic Recombination in Drosophila melanogaster. G3 (Bethesda) 2: 249-260. PubMed ID: 22384403

    Castillo, D. M., Mell, J. C., Box, K. S. and Blumenstiel, J. P. (2011). Molecular evolution under increasing transposable element burden in Drosophila: a speed limit on the evolutionary arms race. BMC Evol Biol 11: 258. PubMed ID: 21917173

  • Edward Blumenthal Biological Sciences Department, Marquette University, Milwaukee
    Conway, S., Sansone, C. L., Benske, A., Kentala, K., Billen, J., Broeck, J. V. and Blumenthal, E. M. (2018). Pleiotropic and novel phenotypes in the Drosophila gut caused by mutation of drop-dead. J Insect Physiol. PubMed ID: 29371099

    Zhang, H. and Blumenthal, E. M. (2017). Identification of multiple functional receptors for tyramine on an insect secretory epithelium. Sci Rep 7(1): 168. PubMed ID: 28279025

    Ruka, K. A., Miller, A. P., Blumenthal, E. M. (2013) Inhibition of diuretic stimulation of an insect secretory epithelium by a cGMP-dependent protein kinase. Am J Physiol Renal Physiol. PubMed ID: 23445619

    Blumenthal, E. M. (2009). Isoform- and cell-specific function of tyrosine decarboxylase in the Drosophila Malpighian tubule. J Exp Biol 212: 3802-3809. PubMed ID: 19915121

    Peller, C. R., Bacon, E. M., Bucheger, J. A. and Blumenthal, E. M. (2009). Defective gut function in drop-dead mutant Drosophila. J Insect Physiol 55: 834-839. PubMed ID: 19500585

  • Laszlo Bodai Department of Biochemistry and Molecular Biology, University of Szeged, Hungary
    Farago, A., Zsindely, N., Farkas, A., Neller, A., Siagi, F., Szabo, M. R., Csont, T. and Bodai, L. (2022). Acetylation State of Lysine 14 of Histone H3.3 Affects Mutant Huntingtin Induced Pathogenesis. Int J Mol Sci 23(23). PubMed ID: 36499499

    Farago, A., Urmosi, A., Farkas, A. and Bodai, L. (2021). The histone replacement gene His4r is involved in heat stress induced chromatin rearrangement. Sci Rep 11(1): 4878. PubMed ID: 33649489

    Varga, J., Korbai, S., Neller, A., Zsindely, N. and Bodai, L. (2019). Hat1 acetylates histone H4 and modulates the transcriptional program in Drosophila embryogenesis. Sci Rep 9(1): 17973. PubMed ID: 31784689

    Farago, A., Zsindely, N. and Bodai, L. (2019). Mutant huntingtin disturbs circadian clock gene expression and sleep patterns in Drosophila. Sci Rep 9(1): 7174. PubMed ID: 31073199

    Jankovics, F., Bence, M., Sinka, R., Farago, A., Bodai, L., Pettko-Szandtner, A., Ibrahim, K., Takacs, Z., Szarka-Kovacs, A. B. and Erdelyi, M. (2018). Drosophila small ovary gene is required for transposon silencing and heterochromatin organization, and ensures germline stem cell maintenance and differentiation. Development 145(23). PubMed ID: 30389853

    Vedelek, V., Bodai, L., Grezal, G., Kovacs, B., Boros, I. M., Laurinyecz, B. and Sinka, R. (2018). Analysis of Drosophila melanogaster testis transcriptome. BMC Genomics 19(1): 697. PubMed ID: 30249207

  • Rolf Bodmer Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
    Ren, J., Zeng, Q., Wu, H., Liu, X., Guida, M. C., Huang, W., Zhai, Y., Li, J., Ocorr, K., Bodmer, R. and Tang, M. (2023). Deacetylase-dependent and -independent role of HDAC3 in cardiomyopathy. J Cell Physiol 238(3): 647-658. PubMed ID: 36745702

    Blumrich, A., Vogler, G., Dresen, S., Diop, S. B., Jaeger, C., Leberer, S., Grune, J., Wirth, E. K., Hoeft, B., Renko, K., Foryst-Ludwig, A., Spranger, J., Sigrist, S., Bodmer, R. and Kintscher, U. (2021). Fat-body brummer lipase determines survival and cardiac function during starvation in Drosophila melanogaster. iScience 24(4): 102288. PubMed ID: 33889813

    Walls, S., Diop, S., Birse, R., Elmen, L., Gan, Z., Kalvakuri, S., Pineda, S., Reddy, C., Taylor, E., Trinh, B., Vogler, G., Zarndt, R., McCulloch, A., Lee, P., Bhattacharya, S., Bodmer, R. and Ocorr, K. (2020). Prolonged Exposure to Microgravity Reduces Cardiac Contractility and Initiates Remodeling in Drosophila. Cell Rep 33(10): 108445. PubMed ID: 33242407

    Vissers, L., Kalvakuri, S., ..., Muenke, M., Kleefstra, T., Bodmer, R. and de Brouwer, A. P. M. (2020). De Novo Variants in CNOT1, a Central Component of the CCR4-NOT Complex Involved in Gene Expression and RNA and Protein Stability, Cause Neurodevelopmental Delay. Am J Hum Genet 107(1): 164-172. PubMed ID: 32553196

    Elmen, L., Volpato, C. B., Kervadec, A., Pineda, S., Kalvakuri, S., Alayari, N. N., Foco, L., Pramstaller, P. P., Ocorr, K., Rossini, A., Cammarato, A., Colas, A. R., Hicks, A. A. and Bodmer, R. (2020). Silencing of CCR4-NOT complex subunits affect heart structure and function. Dis Model Mech. PubMed ID: 32471864

    Akasaka, T., Ocorr, K., Lin, L., Vogler, G., Bodmer, R. and Grossfeld, P. (2020). Overexpression of Kif1A in the Developing Drosophila Heart Causes Valvar and Contractility Defects: Implications for Human Congenital Heart Disease. J Cardiovasc Dev Dis 7(2). PubMed ID: 32498427

    Chang, K., Kang, P., Liu, Y., Huang, K., Miao, T., Sagona, A. P., Nezis, I. P., Bodmer, R., Ocorr, K. and Bai, H. (2019). TGFB-INHB/activin signaling regulates age-dependent autophagy and cardiac health through inhibition of MTORC2. Autophagy: 1-16. PubMed ID: 31884871

    Schroeder, A. M., Allahyari, M., Vogler, G., Missinato, M. A., Nielsen, T., Yu, M. S., Theis, J. L., Larsen, L. A., Goyal, P., Rosenfeld, J., Nelson, T. J., Olson, T. M., Colas, A. R., Grossfeld, P. and Bodmer, R. (2019). Model system identification of novel congenital heart disease gene candidates: focus on RPL13. Hum Mol Genet. PubMed ID: 31625562

  • Grace Boekhoff-Falk Laboratory of Genetics, University of Wisconsin, Madison
    Crocker, K. L., Ahern-Djamali, S. and Boekhoff-Falk, G. (2021). Stimulating and Analyzing Adult Neurogenesis in the Drosophila Central Brain. J Vis Exp(176). PubMed ID: 34694294

    Crocker, K. L., Marischuk, K., Rimkus, S. A., Zhou, H., Yin, J. C. P. and Boekhoff-Falk, G. (2021). Neurogenesis in the adult Drosophila brain. Genetics. PubMed ID: 34117750

    Plavicki, J. S., Squirrell, J. M., Eliceiri, K. W. and Boekhoff-Falk, G. (2015). Expression of the Drosophila homeobox gene, Distal-less supports an ancestral role in neural development. Dev Dyn. PubMed ID: 26472170

    Boekhoff-Falk, G. and Eberl, D. F. (2014). The Drosophila auditory system. Wiley Interdiscip Rev Dev Biol 3: 179-191. PubMed ID: 24719289

    Plavicki, J., Mader, S., Pueschel, E., Peebles, P. and Boekhoff-Falk, G. (2012). Homeobox gene distal-less is required for neuronal differentiation and neurite outgrowth in the Drosophila olfactory system. Proc Natl Acad Sci U S A 109: 1578-1583. PubMed ID: 22307614

  • Sven Bogdan Institut für Neuro- und Verhaltensbiologie, Westfälische Wilhelms-Universität Münster
    Lehne, F. and Bogdan, S. (2023). EFhD2/Swip-1 promotes exocytosis of glue granules in the exocrine Drosophila salivary gland. J Cell Sci. PubMed ID: 36727484

    Lehne, F., Pokrant, T., Parbin, S., Salinas, G., Grosshans, J., Rust, K., Faix, J. and Bogdan, S. (2022). Calcium bursts allow rapid reorganization of EFhD2/Swip-1 cross-linked actin networks in epithelial wound closure. Nat Commun 13(1): 2492. PubMed ID: 35524157

    Hirschhauser, A., van Cann, M. and Bogdan, S. (2021). CK1alpha protects WAVE from degradation to regulate cell shape and motility in immune response. J Cell Sci. PubMed ID: 34730182

    Bischoff, M. C., Lieb, S., Renkawitz-Pohl, R. and Bogdan, S. (2021). Filopodia-based contact stimulation of cell migration drives tissue morphogenesis. Nat Commun 12(1): 791. PubMed ID: 33542237

    Nagel, B. M., Bechtold, M., Rodriguez, L. G. and Bogdan, S. (2016). Drosophila WASH is required for integrin-mediated cell adhesion, cell motility and lysosomal neutralization. J Cell Sci. PubMed ID: 27884932

    Squarr, A. J., Brinkmann, K., Chen, B., Steinbacher, T., Ebnet, K., Rosen, M. K. and Bogdan, S. (2016). Fat2 acts through the WAVE regulatory complex to drive collective cell migration during tissue rotation. J Cell Biol 212: 591-603. PubMed ID: 26903538

    Brinkmann, K., Winterhoff, M., Onel, S. F., Schultz, J., Faix, J. and Bogdan, S. (2015). WHAMY is a novel actin polymerase promoting myoblast fusion, macrophage cell motility and sensory organ development. J Cell Sci. PubMed ID: 26675239

    Zobel, T., Brinkmann, K., Koch, N., Schneider, K., Seemann, E., Fleige, A., Qualmann, B., Kessels, M. M. and Bogdan, S. (2014). Cooperative functions of the two F-BAR proteins Cip4 and Nostrin in regulating E-cadherin in epithelial morphogenesis. J Cell Sci. PubMed ID: 25413347

    Chen, X. J., Squarr, A. J., Stephan, R., Chen, B., Higgins, T. E., Barry, D. J., Martin, M. C., Rosen, M. K., Bogdan, S. and Way, M. (2014). Ena/VASP Proteins Cooperate with the WAVE Complex to Regulate the Actin Cytoskeleton. Dev Cell 30: 569-584. PubMed ID: 25203209

    Lammel, U., Bechtold, M., Risse, B., Berh, D., Fleige, A., Bunse, I., Jiang, X., Klambt, C. and Bogdan, S. (2014). The Drosophila FHOD1-like formin Knittrig acts through Rok to promote stress fiber formation and directed macrophage migration during the cellular immune response. Development. PubMed ID: 24553290

    Sander, M., Squarr, A. J., Risse, B., Jiang, X. and Bogdan, S. (2013). Drosophila pupal macrophages - A versatile tool for combined ex vivo and in vivo imaging of actin dynamics at high resolution. Eur J Cell Biol. PubMed ID: 24183239

  • Dirk Bohmann Department of Biomedical Genetics, University of Rochester Medical Center
    Cheng, Y., Pitoniak, A., Wang, J. and Bohmann, D. (2021). Preserving transcriptional stress responses as an anti-aging strategy. Aging Cell 20(2): e13297. PubMed ID: 33474790

    Chatterjee, N., Tian, M., Spirohn, K., Boutros, M. and Bohmann, D. (2016). Keap1-Independent Regulation of Nrf2 Activity by Protein Acetylation and a BET Bromodomain Protein. PLoS Genet 12: e1006072. PubMed ID: 27233051

    Li, X., Chatterjee, N., Spirohn, K., Boutros, M. and Bohmann, D. (2016). Cdk12 is a gene-selective RNA polymerase II kinase that regulates a subset of the transcriptome, including Nrf2 target genes. Sci Rep 6: 21455. PubMed ID: 26911346

    Pitoniak, A. and Bohmann, D. (2015). Mechanisms and Functions of Nrf2 Signaling in Drosophila. Free Radic Biol Med [Epub ahead of print]. PubMed ID: 26117322

    Barone, M. C. and Bohmann, D. (2013). Assessing neurodegenerative phenotypes in Drosophila dopaminergic neurons by climbing assays and whole brain immunostaining. J Vis Exp. PubMed ID: 23644755

    Mahidur Rahman, M., Sykiotis, G. P., Nishimura, M., Bodmer, R., Bohmann, D. (2013) Declining signal-dependence of Nrf2-MafS regulated gene expressioncorrelates with aging phenotypes. Aging Cell. PubMed ID: 23521918

    Chatterjee, N. and Bohmann, D. (2012). A versatile PhiC31 based reporter system for measuring AP-1 and Nrf2 signaling in Drosophila and in tissue culture. PLoS One 7: e34063. PubMed ID: 22509270

    Barone, M. C., Sykiotis, G. P. and Bohmann, D. (2011). Genetic activation of Nrf2 signaling is sufficient to ameliorate neurodegenerative phenotypes in a Drosophila model of Parkinson's disease. Dis Model Mech 4: 701-707. PubMed ID: 21719443

    Hochmuth, C. E., Biteau, B., Bohmann, D. and Jasper, H. (2011). Redox regulation by Keap1 and Nrf2 controls intestinal stem cell proliferation in Drosophila. Cell Stem Cell 8: 188-199. PubMed ID: 21295275

  • Johannes Bohrmann Unit of Zoology and Human Biology, RWTH Aachen University, Institut für Biologie II, Abt. Zoologie und Humanbiologie, Aachen, Germany
    Schotthofer, S. K. and Bohrmann, J. (2020). Analysing bioelectrical phenomena in the Drosophila ovary with genetic tools: tissue-specific expression of sensors for membrane potential and intracellular pH, and RNAi-knockdown of mechanisms involved in ion exchange. BMC Dev Biol 20(1): 15. PubMed ID: 32635900

    Schotthofer, S. K. and Bohrmann, J. (2020). Bioelectrical and cytoskeletal patterns correlate with altered axial polarity in the follicular epithelium of the Drosophila mutant gurken. BMC Dev Biol 20(1): 5. PubMed ID: 32169045

    Weiss, I. and Bohrmann, J. (2019). Electrochemical gradients are involved in regulating cytoskeletal patterns during epithelial morphogenesis in the Drosophila ovary. BMC Dev Biol 19(1): 22. PubMed ID: 31718540

    Weiss, I. and Bohrmann, J. (2019). Electrochemical patterns during Drosophila oogenesis: ion-transport mechanisms generate stage-specific gradients of pH and membrane potential in the follicle-cell epithelium. BMC Dev Biol 19(1): 12. PubMed ID: 31226923

    Lautemann, J. and Bohrmann, J. (2016). Relating proton pumps with gap junctions: colocalization of ductin, the channel-forming subunit c of V-ATPase, with subunit a and with innexins 2 and 3 during Drosophila oogenesis. BMC Dev Biol 16(1): 24. PubMed ID: 27412523

    Kruger, J. and Bohrmann, J. (2015). Bioelectric patterning during oogenesis: stage-specific distribution of membrane potentials, intracellular pH and ion-transport mechanisms in Drosophila ovarian follicles. BMC Dev Biol 15: 1. PubMed ID: 25591552

  • Antoine Boivin Developmental Biology Laboratory, Sorbonne Universite, Paris
    Casier, K., Autaa, J., Gueguen, N., Delmarre, V., Marie, P. P., Ronsseray, S., Carre, C., Brasset, E., Teysset, L. and Boivin, A. (2023). The histone demethylase Kdm3 prevents auto-immune piRNAs production in Drosophila. Sci Adv 9(14): eade3872. PubMed ID: 37027460

    Marie, P. P., Ronsseray, S. and Boivin, A. (2017). From Embryo to Adult: piRNA-Mediated Silencing throughout Germline Development in Drosophila. G3 (Bethesda) 7(2): 505-516. PubMed ID: 27932388

    Saint-Charles, A., Michard-Vanhee, C., Alejevski, F., Chelot, E., Boivin, A. and Rouyer, F. (2016). Four of the six Drosophila rhodopsin-expressing photoreceptors can mediate circadian entrainment in low light. J Comp Neurol 524(14): 2828-2844. PubMed ID: 26972685

  • Christian Bökel Center for Regenerative Therapies, Dresden
    Albert, E. A., Puretskaia, O. A., Terekhanova, N. V., Labudina, A. and Bokel, C. (2018). Direct control of somatic stem cell proliferation factors by the Drosophila testis stem cell niche. Development. PubMed ID: 30002131

    Albert, E. A. and Bokel, C. (2017). A cell based, high throughput assay for quantitative analysis of Hedgehog pathway activation using a Smoothened activation sensor. Sci Rep 7(1): 14341. PubMed ID: 29085027

    Kupinski, A. P., Raabe, I., Michel, M., Ail, D., Brusch, L., Weidemann, T. and Bokel, C. (2013). Phosphorylation of the Smo tail is controlled by membrane localisation and is dispensable for clustering. J Cell Sci 126: 4684-4697. PubMed ID: 23943866

    Michel, M., Kupinski, A. P., Raabe, I. and Bokel, C. (2012). Hh signalling is essential for somatic stem cell maintenance in the Drosophila testis niche. Development 139: 2663-2669. PubMed ID: 22745310

    Michel, M., Raabe, I., Kupinski, A. P., Perez-Palencia, R. and Bokel, C. (2011). Local BMP receptor activation at adherens junctions in the Drosophila germline stem cell niche. Nat Commun 2: 415. PubMed ID: 21811244

  • Francois Bolduc University of Alberta
    Androschuk, A., He, R. X., Weber, S., Rosenfelt, C. and Bolduc, F. V. (2018). Stress Odorant Sensory Response Dysfunction in Drosophila Fragile X Syndrome Mutants. Front Mol Neurosci 11: 242. PubMed ID: 30135642

    Monyak, R. E., Emerson, D., Schoenfeld, B. P., Zheng, X., Chambers, D. B., Rosenfelt, C., Langer, S., Hinchey, P., Choi, C. H., McDonald, T. V., Bolduc, F. V., Sehgal, A., McBride, S. M. J. and Jongens, T. A. (2017). Insulin signaling misregulation underlies circadian and cognitive deficits in a Drosophila fragile X model. Mol Psychiatry 22(8): 1140-1148. PubMed ID: 27090306

    Julien, C., Lissouba, A., Madabattula, S., Fardghassemi, Y., Rosenfelt, C., Androschuk, A., Strautman, J., Wong, C., Bysice, A., O'Sullivan, J., Rouleau, G. A., Drapeau, P., Parker, J. A. and Bolduc, F. V. (2016). Conserved pharmacological rescue of hereditary spastic paraplegia-related phenotypes across model organisms. Hum Mol Genet 25(6): 1088-1099. PubMed ID: 26744324

  • Silvia Bonaccorsi Genetics and Molecular Biology, University of Rome
    Palumbo, V., Tariq, A., Borgal, L., Metz, J., Brancaccio, M., Gatti, M., Wakefield, J. G. and Bonaccorsi, S. (2020). Drosophila Morgana is an Hsp90-interacting protein with a direct role in microtubule polymerisation. J Cell Sci 133(2). PubMed ID: 31907206

    Palumbo, V., Pellacani, C., Heesom, K. J., Rogala, K. B., Deane, C. M., Mottier-Pavie, V., Gatti, M., Bonaccorsi, S. and Wakefield, J. G. (2015). Misato Controls Mitotic Microtubule Generation by Stabilizing the Tubulin Chaperone Protein-1 Complex. Curr Biol 25: 1777-1783. PubMed ID: 26096973

    Mengoli, V., Bucciarelli, E., Lattao, R., Piergentili, R., Gatti, M. and Bonaccorsi, S. (2014). The analysis of mutant alleles of different strength reveals multiple functions of topoisomerase 2 in regulation of Drosophila chromosome structure. PLoS Genet 10: e1004739. PubMed ID: 25340516

    Bonaccorsi, S., Giansanti, M. G., Cenci, G. and Gatti, M. (2012). Chromatin staining of Drosophila testes. Cold Spring Harb Protoc 2012. PubMed ID: 22854562

    Gatti, M., Bucciarelli, E., Lattao, R., Pellacani, C., Mottier-Pavie, V., Giansanti, M. G., Somma, M. P. and Bonaccorsi, S. (2012). The relative roles of centrosomal and kinetochore-driven microtubules in Drosophila spindle formation. Exp Cell Res 318: 1375-1380. PubMed ID: 22580224

    Lattao, R., Bonaccorsi, S. and Gatti, M. (2012). Giant meiotic spindles in males from Drosophila species with giant sperm tails. J Cell Sci 125: 584-588. PubMed ID: 22389398

  • Artem Bonchuk Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
    Fedotova, A. A., Georgiev, P. G. and Bonchuk, A. N. (2023). Study of the in Vivo Functional Role of Mutations in the BTB Domain of the CP190 Protein of Drosophila melanogaster. Dokl Biochem Biophys 509(1): 47-50. PubMed ID: 37340291

    Tikhonova, E., Mariasina, S., Efimov, S., Polshakov, V., Maksimenko, O., Georgiev, P. and Bonchuk, A. (2022). Structural basis for interaction between CLAMP and MSL2 proteins involved in the specific recruitment of the dosage compensation complex in Drosophila. Nucleic Acids Res 50(11): 6521-6531. PubMed ID: 35648444

    Bonchuk, A. N., Boyko, K. M., Nikolaeva, A. Y., Burtseva, A. D., Popov, V. O. and Georgiev, P. G. (2022). Structural insights into highly similar spatial organization of zinc-finger associated domains with a very low sequence similarity. Structure 30(7): 1004-1015 e1004. PubMed ID: 35580610

    Tikhonova, E., Mariasina, S., Arkova, O., Maksimenko, O., Georgiev, P. and Bonchuk, A. (2022). Dimerization Activity of a Disordered N-Terminal Domain from Drosophila CLAMP Protein. Int J Mol Sci 23(7). PubMed ID: 35409222

    >Sabirov, M., Popovich, A., Boyko, K., Nikolaeva, A., Kyrchanova, O., Maksimenko, O., Popov, V., Georgiev, P. and Bonchuk, A. (2021). Mechanisms of CP190 Interaction with Architectural Proteins in Drosophila Melanogaster. Int J Mol Sci 22(22). PubMed ID: 34830280

  • Sarah Bondos Department of Molecular and Cellular Medicine, Texas A&M, College Station
    Liu, Y., Huang, A., Booth, R. M., Mendes, G. G., Merchant, Z., Matthews, K. S. and Bondos, S. E. (2018). Evolution of the activation domain in a Hox transcription factor. Int J Dev Biol 62(11-12): 745-753. PubMed ID: 30604844

    Xie, X. J., Hsu, F. N., Gao, X., Xu, W., Ni, J. Q., Xing, Y., Huang, L., Hsiao, H. C., Zheng, H., Wang, C., Zheng, Y., Xiaoli, A. M., Yang, F., Bondos, S. E. and Ji, J. Y. (2015). CDK8-Cyclin C Mediates Nutritional Regulation of Developmental Transitions through the Ecdysone Receptor in Drosophila. PLoS Biol 13: e1002207. PubMed ID: 26222308

    Hsiao, H. C., Gonzalez, K. L., Catanese, D. J., Jordy, K. E., Matthews, K. S. and Bondos, S. E. (2014). The intrinsically disordered regions of the Drosophila melanogaster hox protein Ultrabithorax select interacting proteins based on partner topology. PLoS One 9: e108217. PubMed ID: 25286318

    Patterson, J. L., Arenas-Gamboa, A. M., Wang, T. Y., Hsiao, H. C., Howell, D. W., Pellois, J. P., Rice-Ficht, A. and Bondos, S. E. (2014). Materials composed of the Drosophila Hox protein Ultrabithorax are biocompatible and nonimmunogenic. J Biomed Mater Res A. PubMed ID: 25087647

    Patterson, J. L., Abbey, C. A., Bayless, K. J. and Bondos, S. E. (2013). Materials composed of the Drosophila melanogaster protein ultrabithorax are cytocompatible. J Biomed Mater Res A. PubMed ID: 23596050

  • Nancy Bonini Department of Biology, University of Pennsylvania
    Sheng, L., Shields, E. J., Gospocic, J., Sorida, M., Ju, L., Byrns, C. N., Carranza, F., Berger, S. L., Bonini, N. and Bonasio, R. (2023). Ensheathing glia promote increased lifespan and healthy brain aging. Aging Cell: e13803. PubMed ID: 36840361

    Perlegos, A. E., Shields, E. J., Shen, H., Liu, K. F. and Bonini, N. M. (2022). Mettl3-dependent m(6)A modification attenuates the brain stress response in Drosophila. Nat Commun 13(1): 5387. PubMed ID: 36104353

    Saikumar, J., Kim, J., Byrns, C. N., Hemphill, M., Meaney, D. F. and Bonini, N. M. (2021). Inducing different severities of traumatic brain injury in Drosophila using a piezoelectric actuator. Nat Protoc 16(1): 263-282. PubMed ID: 33277631

    Saikumar, J., Byrns, C. N., Hemphill, M., Meaney, D. F. and Bonini, N. M. (2020). Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 117(29): 17269-17277. PubMed ID: 32611818

    Goodman, L. D., Prudencio, M., Srinivasan, A. R., Rifai, O. M., Lee, V. M., Petrucelli, L. and Bonini, N. M. (2019). eIF4B and eIF4H mediate GR production from expanded G4C2 in a Drosophila model for C9orf72-associated ALS. Acta Neuropathol Commun 7(1): 62. PubMed ID: 31023341

    Kennerdell, J. R., Liu, N. and Bonini, N. M. (2018). MiR-34 inhibits polycomb repressive complex 2 to modulate chaperone expression and promote healthy brain aging. Nat Commun 9(1): 4188. PubMed ID: 30305625

    McGurk, L., Gomes, E., Guo, L., Mojsilovic-Petrovic, J., Tran, V., Kalb, R. G., Shorter, J. and Bonini, N. M. (2018). Poly(ADP-Ribose) Prevents Pathological Phase Separation of TDP-43 by Promoting Liquid Demixing and Stress Granule Localization. Mol Cell 71(5): 703-717.e709. PubMed ID: 30100264

    Berson, A., Sartoris, A., Nativio, R., Van Deerlin, V., Toledo, J. B., Porta, S., Liu, S., Chung, C. Y., Garcia, B. A., Lee, V. M., Trojanowski, J. Q., Johnson, F. B., Berger, S. L. and Bonini, N. M. (2017). TDP-43 promotes neurodegeneration by impairing chromatin remodeling. Curr Biol[Epub ahead of print]. PubMed ID: 29153328

    Burguete, A. S., Almeida, S., Gao, F. B., Kalb, R., Akins, M. R. and Bonini, N. M. (2015). GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. Elife 4. PubMed ID: 26650351

    Yu, Z., Goodman, L. D., Shieh, S. Y., Min, M., Teng, X., Zhu, Y. and Bonini, N. M. (2015). A fly model for the CCUG-repeat expansion of myotonic dystrophy type 2 reveals a novel interaction with MBNL1. Hum Mol Genet 24: 954-962. PubMed ID: 25305073

  • Seth Bordenstein Biological Sciences, Vanderbilt University, Nashville, Tenn
    Kaur, R., Leigh, B. A., Ritchie, I. T. and Bordenstein, S. R. (2022). The Cif proteins from Wolbachia prophage WO modify sperm genome integrity to establish cytoplasmic incompatibility. PLoS Biol 20(5): e3001584. PubMed ID: 35609042

    Perlmutter, J. I., Meyers, J. E. and Bordenstein, S. R. (2021). A single synonymous nucleotide change impacts the male-killing phenotype of prophage WO gene wmk. Elife 10. PubMed ID: 34677126

    Shropshire, J. D., Rosenberg, R. and Bordenstein, S. R. (2021). The impacts of cytoplasmic incompatibility factor (cifA and cifB) genetic variation on phenotypes. Genetics 217(1): 1-13. PubMed ID: 33683351

  • Erich Bornberg-Bauer Molecular Evolution And Bioinformatics at Univerity of Muenster, Germany
    Grandchamp, A., Kuhl, L., Lebherz, M., Bruggemann, K., Parsch, J. and Bornberg-Bauer, E. (2023). Population genomics reveals mechanisms and dynamics of de novo expressed open reading frame emergence in Drosophila melanogaster. Genome Res 33(6): 872-890. PubMed ID: 37442576

    Bitard-Feildel, T., M. Heberlein, E. Bornberg-Bauer and I. Callebaut (2015). Detection of orphan domains in Drosophila using "hydrophobic cluster analysis". Biochimie 119: 244-253. PubMed ID: 25736992

    Grandchamp, A., L. Kuhl, M. Lebherz, K. Bruggemann, J. Parsch and E. Bornberg-Bauer (2023). Population genomics reveals mechanisms and dynamics of de novo expressed open reading frame emergence in Drosophila melanogaster. Genome Res 33(6). PubMed ID: 37442576

  • Imre Boros Institute of Biochemistry, Eukaryotic Molecular Biology Unit, Biological Research Centre, Szeged
    Abraham, A., Villanyi, Z., Zsindely, N., Nagy, G., Szabo, A., Bodai, L., Henn, L. and Boros, I. M. (2022). Despite its sequence identity with canonical H4, Drosophila H4r product is enriched at specific chromatin regions. Sci Rep 12(1): 5007. PubMed ID: 35322122

    Vedelek, B., Kovacs, Á. and Boros, I. M. (2021). Evolutionary mode for the functional preservation of fast-evolving Drosophila telomere capping proteins. Open Biol 11(11): 210261. PubMed ID: 34784790

    Henn, L., et al. (2020). Alternative linker histone permits fast paced nuclear divisions in early Drosophila embryo. Nucleic Acids Res. PubMed ID: 32710625

    Vedelek, B., Blastyak, A. and Boros, I. M. (2015). Cross-species interaction between rapidly evolving telomere-specific Drosophila proteins. PLoS One 10: e0142771. PubMed ID: 26566042

    Pahi, Z., Kiss, Z., Komonyi, O., Borsos, B. N., Tora, L., Boros, I. M. and Pankotai, T. (2015). dTAF10- and dTAF10b-containing complexes are required for ecdysone-driven larval-pupal morphogenesis in Drosophila melanogaster. PLoS One 10: e0142226. PubMed ID: 26556600

    Borsos, B. N., Pankotai, T., Kovacs, D., Popescu, C., Pahi, Z. and Boros, I. M. (2015). Acetylations of Ftz-F1 and histone H4K5 are required for the fine-tuning of ecdysone biosynthesis during Drosophila metamorphosis. Dev Biol. PubMed ID: 25959239

    Pankotai, T., Zsindely, N., Vamos, E. E., Komonyi, O., Bodai, L. and Boros, I. M. (2013). Functional characterization and gene expression profiling of Drosophila melanogaster short dADA2b isoform-containing dSAGA complexes. BMC Genomics 14: 44. PubMed ID: 23336284

    uhasz, I., Villanyi, Z., Tombacz, I. and Boros, I. M. (2012). High Fcp1 phosphatase activity contributes to setting an intense transcription rate required in Drosophila nurse and follicular cells for egg production. Gene 509: 60-67. PubMed ID: 22903034

    Bodai, L., Zsindely, N., Gaspar, R., Kristo, I., Komonyi, O. and Boros, I. M. (2012). Ecdysone induced gene expression is associated with acetylation of histone H3 lysine 23 in Drosophila melanogaster. PLoS One 7: e40565. PubMed ID: 22808194

  • Alexander Borst Max Planck Institute of Neurobiology, Martinsried
    Pirogova, N. and Borst, A. (2023). Contrast normalization affects response time-course of visual interneurons. PLoS One 18(6): e0285686. PubMed ID: 37294743

    Mishra, A., Serbe-Kamp, E., Borst, A. and Haag, J. (2023). Voltage to Calcium Transformation Enhances Direction Selectivity in Drosophila T4 Neurons. J Neurosci 43(14): 2497-2514. PubMed ID: 36849417

    Ammer, G., Vieira, R. M., Fendl, S. and Borst, A. (2022). Anatomical distribution and functional roles of electrical synapses in Drosophila. Curr Biol 32(9): 2022-2036.e2024. PubMed ID: 35385694

    Groschner, L. N., Malis, J. G., Zuidinga, B. and Borst, A. (2022). A biophysical account of multiplication by a single neuron. Nature 603(7899): 119-123. PubMed ID: 35197635

    Leonte, M. B., Leonhardt, A., Borst, A. and Mauss, A. S. (2021). Aerial course stabilization is impaired in motion-blind flies. J Exp Biol. PubMed ID: 34180515

    Fendl, S., Vieira, R. M. and Borst, A. (2020). Conditional protein tagging methods reveal highly specific subcellular distribution of ion channels in motion-sensing neurons. Elife 9. PubMed ID: 33079061

    Schuetzenberger, A. and Borst, A. (2020). Seeing Natural Images through the Eye of a Fly with Remote Focusing Two-Photon Microscopy. iScience 23(6): 101170. PubMed ID: 32502966

    Hoermann, N., Schilling, T., Haji Ali, A., Serbe, E., Mayer, C., Borst, A. and Pujol-Marti, J. (2020). A combinatorial code of transcription factors specifies subtypes of visual motion-sensing neurons in Drosophila. Development. PubMed ID: 32238425

    Drews, M. S., Leonhardt, A., Pirogova, N., Richter, F. G., Schuetzenberger, A., Braun, L., Serbe, E. and Borst, A. (2020). Dynamic signal compression for robust motion vision in flies. Curr Biol 30(2): 209-221.e208. PubMed ID: 31928873

    Meier, M. and Borst, A. (2019). Extreme compartmentalization in a Drosophila amacrine cell. Curr Biol 29(9): 1545-1550.e1542. PubMed ID: 31031119

    Schilling, T., Ali, A. H., Leonhardt, A., Borst, A. and Pujol-Marti, J. (2019). Transcriptional control of morphological properties of direction-selective T4/T5 neurons in Drosophila. Development. PubMed ID: 30642835

    Busch, C., Borst, A. and Mauss, A. S. (2018). Bi-directional control of walking behavior by horizontal optic flow sensors. Curr Biol. PubMed ID: 30528583

  • Giovanni Bosco Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Hanover, NH
    Sadanandappa, M. K., Sathyanarayana, S. H., Kondo, S. and Bosco, G. (2021). Neuropeptide F signaling regulates parasitoid-specific germline development and egg-laying in Drosophila. PLoS Genet 17(3): e1009456. PubMed ID: 33770070

    Edwards, K. A., Hoppa, M. B. and Bosco, G. (2020). The Photoconvertible Fluorescent Probe, CaMPARI, Labels Active Neurons in Freely-Moving Intact Adult Fruit Flies. Front Neural Circuits 14: 22. PubMed ID: 32457580

    Bozler, J., Kacsoh, B. Z. and Bosco, G. (2019). Maternal priming of offspring immune system in Drosophila. G3 (Bethesda). PubMed ID: 31685524

    Kacsoh, B. Z., Bozler, J., Hodge, S. and Bosco, G. (2019). Neural circuitry of social learning in Drosophila requires multiple inputs to facilitate inter-species communication. Commun Biol 2: 309. PubMed ID: 31428697

    Bozler, J., Kacsoh, B. Z. and Bosco, G. (2019). Transgeneratonal inheritance of ethanol preference is caused by maternal NPF repression. Elife 8. PubMed ID: 31287057

    Kacsoh, B. Z., Barton, S., Jiang, Y., Zhou, N., Mooney, S. D., Friedberg, I., Radivojac, P., Greene, C. S. and Bosco, G. (2018). New Drosophila long-term memory genes revealed by assessing computational function prediction methods. G3 (Bethesda). PubMed ID: 30463884

    Kacsoh, B. Z., Bozler, J. and Bosco, G. (2018). Drosophila species learn dialects through communal living. PLoS Genet 14(7): e1007430. PubMed ID: 30024883

    Bozler, J., Kacsoh, B. Z., Chen, H., Theurkauf, W. E., Weng, Z. and Bosco, G. (2017). A systems level approach to temporal expression dynamics in Drosophila reveals clusters of long term memory genes. PLoS Genet 13(10): e1007054. PubMed ID: 29084214

    Kacsoh, B. Z., Greene, C. S. and Bosco, G. (2017). Machine learning analysis identifies DrosophilaGrunge/Atrophin as an important learning and memory gene required for memory retention and social learning. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28889104

    Kacsoh, B.Z., Bozler, J., Ramaswami, M. and Bosco, G. (2015). Social communication of predator-induced changes in Drosophila behavior and germline physiology. Elife [Epub ahead of print]. PubMed ID: 25970035

    Nguyen, H. Q., Nye, J., Buster, D. W., Klebba, J. E., Rogers, G. C. and Bosco, G. (2015). Drosophila Casein Kinase I alpha regulates homolog pairing and genome organization by modulating Condensin II subunit Cap-H2 levels. PLoS Genet 11: e1005014. PubMed ID: 25723539

  • Torsten Bossing School of Biological Sciences, Bangor University, Gwynedd, UK
    Barros, C. S. and Bossing, T. (2021). Microtubule disruption upon CNS damage triggers mitotic entry via TNF signaling activation. Cell Rep 36(1): 109325. PubMed ID: 34233183

    Gil-Ranedo, J., Gonzaga, E., Jaworek, K. J., Berger, C., Bossing, T. and Barros, C. S. (2019). STRIPAK Members Orchestrate Hippo and Insulin Receptor Signaling to Promote Neural Stem Cell Reactivation. Cell Rep 27(10): 2921-2933.e2925. PubMed ID: 31167138

    Ding, R., Weynans, K., Bossing, T., Barros, C. S. and Berger, C. (2016). The Hippo signalling pathway maintains quiescence in Drosophila neural stem cells. Nat Commun 7: 10510. PubMed ID: 26821647

    Liu, B., Campo, E. M. and Bossing, T. (2014). Drosophila Embryos as Model to Assess Cellular and Developmental Toxicity of Multi-Walled Carbon Nanotubes (MWCNT) in Living Organisms. PLoS One 9: e88681. PubMed ID: 24558411

    Cowan, C. M., Bossing, T., Page, A., Shepherd, D. and Mudher, A. (2010). Soluble hyper-phosphorylated tau causes microtubule breakdown and functionally compromises normal tau in vivo. Acta Neuropathol 120: 593-604. PubMed ID: 20617325

  • Juan Botas Molecular and Human Genetics, Baylor College of Medicine, Houston
    Li, J., Amoh, B. K., McCormick, E., Tarkunde, A., Zhu, K. F., Perez, A., Mair, M., Moore, J., Shulman, J. M., Al-Ramahi, I. and Botas, J. (2023). Integration of transcriptome-wide association study with neuronal dysfunction assays provides functional genomics evidence for Parkinson's disease genes. Hum Mol Genet 32(4): 685-695. PubMed ID: 36173927

    Hernandez, S. J., Lim, R. G., Onur, T., Dane, M. A., Smith, R., Wang, K., Jean, G. E., Devlin, K., Miramontes, R., Wu, J., Casale, M., Kilburn, D., Heiser, L. M., Korkola, J. E., Van Vactor, D., Botas, J., Thompson-Peer, K. L. and Thompson, L. M. (2022). An altered extracellular matrix-integrin interface contributes to Huntington's disease-associated CNS dysfunction in glial and vascular cells. Hum Mol Genet. PubMed ID: 36547263

    Kim, J., de Haro, M., Al-Ramahi, I., Garaicoechea, L. L., Jeong, H. H., Sonn, J. Y., Tadros, B., Liu, Z., Botas, J. and Zoghbi, H. Y. (2022). Evolutionarily conserved regulators of tau identify targets for new therapies. Neuron. PubMed ID: 36610398

    Suart, C. E., Perez, A. M., Al-Ramahi, I., Maiuri, T., Botas, J. and Truant, R. (2021). Spinocerebellar Ataxia Type 1 protein Ataxin-1 is signalled to DNA damage by Ataxia Telangiectasia Mutated kinase. Hum Mol Genet. PubMed ID: 33772540

    Onur, T. S., Laitman, A., Zhao, H., Keyho, R., Kim, H., Wang, J., Mair, M., Wang, H., Li, L., Perez, A., de Haro, M., Wan, Y. W., Allen, G., Lu, B., Al-Ramahi, I., Liu, Z. and Botas, J. (2021). Downregulation of glial genes involved in synaptic function mitigates Huntington's Disease pathogenesis. Elife 10. PubMed ID: 33871358

    Kim, Y. W., Al-Ramahi, I., Koire, A., Wilson, S. J., Konecki, D. M., Mota, S., Soleimani, S., Botas, J. and Lichtarge, O. (2020). Harnessing the paradoxical phenotypes of APOE epsilon2 and APOE epsilon4 to identify genetic modifiers in Alzheimer's disease. Alzheimers Dement. PubMed ID: 33576571

    Al-Ramahi, I., Lu, B., Di Paola, S., Pang, K., de Haro, M., Peluso, I., Gallego-Flores, T., Malik, N. T., Erikson, K., Bleiberg, B. A., Avalos, M., Fan, G., Rivers, L. E., Laitman, A. M., Diaz-Garcia, J. R., Hild, M., Palacino, J., Liu, Z., Medina, D. L. and Botas, J. (2018). High-Throughput Functional Analysis Distinguishes Pathogenic, Nonpathogenic, and Compensatory Transcriptional Changes in Neurodegeneration. Cell Syst. PubMed ID: 29936182

    Bouche, V., Perez Espinosa, A., Leone, L., Sardiello, M., Ballabio, A. and Botas, J. (2016). Drosophila Mitf regulates the V-ATPase and the lysosomal-autophagic pathway. Autophagy: 0. PubMed ID: 26761346

    El-Daher, M. T., Hangen, E., Bruyere, J., Poizat, G., Al-Ramahi, I., Pardo, R., Bourg, N., Souquere, S., Mayet, C., Pierron, G., Leveque-Fort, S., Botas, J., Humbert, S. and Saudou, F. (2015). Huntingtin proteolysis releases non-polyQ fragments that cause toxicity through dynamin 1 dysregulation. EMBO J. PubMed ID: 26165689

    Calap-Quintana, P., Soriano, S., Llorens, J. V., Al-Ramahi, I., Botas, J., Molto, M. D. and Martinez-Sebastian, M. J. (2015). TORC1 inhibition by rapamycin promotes antioxidant defences in a Drosophila model of Friedreich's ataxia. PLoS One 10: e0132376. PubMed ID: 26158631

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

  • Michael Botchan Biochemistry & Molecular Biology, U. C. Berkeley
    Parker, M. W., Kao, J. A., Huang, A., Berger, J. M. and Botchan, M. R. (2021). Molecular determinants of phase separation for Drosophila DNA replication licensing factors. Elife 10. PubMed ID: 34951585

    Bleichert, F., Leitner, A., Aebersold, R., Botchan, M. R. and Berger, J. M. (2018). Conformational control and DNA-binding mechanism of the metazoan origin recognition complex. Proc Natl Acad Sci U S A 115(26): E5906-e5915. PubMed ID: 29899147

    Bleichert, F., Botchan, M. R. and Berger, J. M. (2015). Crystal structure of the eukaryotic origin recognition complex. Nature. PubMed ID: 25762138

    Blanchard, D. P., Georlette, D., Antoszewski, L. and Botchan, M. R. (2014). Chromatin reader L(3)mbt requires the Myb-MuvB/DREAM transcriptional regulatory complex for chromosomal recruitment. Proc Natl Acad Sci U S A. PubMed ID: 25249635

    Bell, S. D. and Botchan, M. R. (2013). The minichromosome maintenance replicative helicase. Cold Spring Harb Perspect Biol 5: a012807. PubMed ID: 23881943

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

    Harrison, M. M., Li, X. Y., Kaplan, T., Botchan, M. R. and Eisen, M. B. (2011). Zelda binding in the early Drosophila melanogaster embryo marks regions subsequently activated at the maternal-to-zygotic transition. PLoS Genet 7: e1002266. PubMed ID: 22028662

    Harrison, M. M., Botchan, M. R. and Cline, T. W. (2010). Grainyhead and Zelda compete for binding to the promoters of the earliest-expressed Drosophila genes. Dev Biol 345: 248-255. PubMed ID: 20599892

  • Tamara Boto Trinity College Dublin
    Marquand, K., Roselli, C., Cervantes-Sandoval, I. and Boto, T. (2023). Sleep benefits different stages of memory in Drosophila. Front Physiol 14: 1087025. PubMed ID: 36744027

    Botero, V., Stanhope, B. A., Brown, E. B., Grenci, E. C., Boto, T., Park, S. J., King, L. B., Murphy, K. R., Colodner, K. J., Walker, J. A., Keene, A. C., Ja, W. W. and Tomchik, S. M. (2021). Neurofibromin regulates metabolic rate via neuronal mechanisms in Drosophila. Nat Commun 12(1): 4285. PubMed ID: 34257279

    King, L. B., Boto, T., Botero, V., Aviles, A. M., Jomsky, B. M., Joseph, C., Walker, J. A. and Tomchik, S. M. (2020). Developmental loss of neurofibromin across distributed neuronal circuits drives excessive grooming in Drosophila. PLoS Genet 16(7): e1008920. PubMed ID: 32697780

    Boto, T., Stahl, A. and Tomchik, S. M. (2020). Cellular and circuit mechanisms of olfactory associative learning in Drosophila. J Neurogenet 34(1): 36-46. PubMed ID: 32043414

  • Gabrielle Boulianne University of Toronto
    Lee, S. H., Gomes, S. M., Ghalayini, J., Iliadi, K. G. and Boulianne, G. L. (2020). Angiotensin Converting Enzyme Inhibitors and Angiotensin Receptor Blockers rescue memory defects in Drosophila expressing Alzheimer's disease-related transgenes independently of the canonical Renin Angiotensin System. eNeuro. PubMed ID: 33060184

    Wasserman, S. S., Shteiman-Kotler, A., Harris, K., Iliadi, K. G., Persaud, A., Zhong, Y., Zhang, Y., Fang, X., Boulianne, G. L., Stewart, B. and Rotin, D. (2018). Regulation of SH3PX1 by dNedd4-long at the Drosophila Neuromuscular Junction. J Biol Chem. PubMed ID: 30518551

    Trinh, I., Gluscencova, O. B. and Boulianne, G. L. (2018). An in vivo screen for neuronal genes involved in obesity identifies Diacylglycerol kinase as a regulator of insulin secretion. Mol Metab. PubMed ID: 30389349

    Iliadi, K. G., Gluscencova, O. B., Iliadi, N. and Boulianne, G. L. (2018). Mutations in the Drosophila homolog of human PLA2G6 give rise to age-dependent loss of psychomotor activity and neurodegeneration. Sci Rep 8(1): 2939. PubMed ID: 29440694

    Iliadi, K. G., Iliadi, N. and Boulianne, G. L. (2017). Drosophila mutants lacking octopamine exhibit impairment in aversive olfactory associative learning. Eur J Neurosci [Epub ahead of print]. PubMed ID: 28715094

    Safi, F., Shteiman-Kotler, A., Zhong, Y., Iliadi, K. G., Boulianne, G. L. and Rotin, D. (2016). Drosophila Nedd4-long reduces Amphiphysin levels in muscles and leads to impaired T-tubule formation. Mol Biol Cell

    Knight, D., Iliadi, K. G., Iliadi, N., Wilk, R., Hu, J., Krause, H. M., Taylor, P., Moran, M. F. and Boulianne, G. L. (2015). Distinct regulation of transmitter release at the Drosophila NMJ by different isoforms of nemy. PLoS One 10: e0132548. PubMed ID: 26237434

    Xu, H., Mohtashami, M., Stewart, B., Boulianne, G. and Trimble, W. S. (2014). Drosophila SNAP-29 Is an Essential SNARE That Binds Multiple Proteins Involved in Membrane Traffic. PLoS One 9: e91471. PubMed ID: 24626111

    Trimble, W. S.Su, J., Chow, B., Boulianne, G. L., Wilde, A. (2013) The BAR domain of Amphiphysin is required for pseudocleavage furrow tip (PFT)-tubule formation during cellularization in Drosophila embryos. Mol Biol Cell. PubMed ID: 23447705

    van de Hoef, D. L., Bonner, J. M., Boulianne, G. L. (2013) FKBP14 is an essential gene that regulates Presenilin protein levels and Notch signaling in Drosophila. Development. PubMed ID: 23318643

  • Henri-Marc Bourbon Centre de Biologie du Développement, Université Toulouse III - Paul Sabatier, Toulouse
    Immarigeon, C., Bernat-Fabre, S., Guillou, E., Verger, A., Prince, E., Benmedjahed, M. A., Payet, A., Couralet, M., Monte, D., Villeret, V., Bourbon, H. M. and Boube, M. (2020). Mediator complex subunit Med19 binds directly GATA transcription factors and is required with Med1 for GATA-driven gene regulation in vivo. J Biol Chem. PubMed ID: 32737196

    Immarigeon, C., Bernat-Fabre, S., Auge, B., Faucher, C., Gobert, V., Haenlin, M., Waltzer, L., Payet, A., Cribbs, D. L., Bourbon, H. G. and Boube, M. (2019). Drosophila mediator aubunit Med1 is required for GATA-dependent developmental processes: Divergent binding interfaces for conserved coactivator functions. Mol Cell Biol 39(7). PubMed ID: 30670567

    Immarigeon, C., Bernat-Fabre, S., Auge, B., Faucher, C., Gobert, V., Haenlin, M., Waltzer, L., Payet, A., Cribbs, D. L., Bourbon, H. G. and Boube, M. (2019). Drosophila Mediator subunit Med1 is required for GATA-dependent developmental processes: divergent binding interfaces for conserved coactivator functions. Mol Cell Biol. PubMed ID: 30670567

    Mojica-Vazquez, L. H., Benetah, M. H., Baanannou, A., Bernat-Fabre, S., Deplancke, B., Cribbs, D. L., Bourbon, H. M. and Boube, M. (2017). Tissue-specific enhancer repression through molecular integration of cell signaling inputs. PLoS Genet 13(4): e1006718. PubMed ID: 28394894

    Baanannou, A., Mojica-Vazquez, L. H., Darras, G., Couderc, J. L., Cribbs, D. L., Boube, M. and Bourbon, H. M. (2013). Distal-less and Rotund Bind a Single Enhancer Ensuring Reliable and Robust Expression in Distinct Limb Morphogenetic Fields. PLoS Genet 9: e1003581. PubMed ID: 23825964

    Makki, R., Meister, M., Pennetier, D., Ubeda, J. M., Braun, A., Daburon, V., Krzemien, J., Bourbon, H. M., Zhou, R., Vincent, A. and Crozatier, M. (2010). A short receptor downregulates JAK/STAT signalling to control the Drosophila cellular immune response. PLoS Biol 8: e1000441. PubMed ID: 20689801

    Gobert, V., Osman, D., Bras, S., Auge, B., Boube, M., Bourbon, H. M., Horn, T., Boutros, M., Haenlin, M. and Waltzer, L. (2010). A genome-wide RNA interference screen identifies a differential role of the mediator CDK8 module subunits for GATA/ RUNX-activated transcription in Drosophila. Mol Cell Biol 30: 2837-2848. PubMed ID: 20368357

  • Michael Boutros Deutsches Krebsforschungszentrum, Heidelberg
    Zhou, J., Valentini, E. and Boutros, M. (2021). Microenvironmental innate immune signaling and cell mechanical responses promote tumor growth. Dev Cell 56(13): 1884-1899.e1885. PubMed ID: 34197724

    Port, F., Starostecka, M. and Boutros, M. (2020). Multiplexed conditional genome editing with Cas12a in Drosophila. Proc Natl Acad Sci U S A 117(37): 22890-22899. PubMed ID: 32843348

    Zhou, J. and Boutros, M. (2020). JNK-dependent intestinal barrier failure disrupts host-microbe homeostasis during tumorigenesis. Proc Natl Acad Sci U S A 117(17): 9401-9412. PubMed ID: 32277031

    Chaudhary, V. and Boutros, M. (2019). Exocyst-mediated apical Wg secretion activates signaling in the Drosophila wing epithelium. PLoS Genet 15(9): e1008351. PubMed ID: 31527874

    Chaudhary, V., Hingole, S., Frei, J., Port, F., Strutt, D. and Boutros, M. (2019). Robust Wnt signaling is maintained by a Wg protein gradient and Fz2 receptor activity in the developing Drosophila wing. Development 146(15). PubMed ID: 31399474

    Bageritz, J., Willnow, P., Valentini, E., Leible, S., Boutros, M. and Teleman, A. A. (2019). Gene expression atlas of a developing tissue by single cell expression correlation analysis. Nat Methods 16(8): 750-756. PubMed ID: 31363221

    Heigwer, F., Scheeder, C., Miersch, T., Schmitt, B., Blass, C., Pour Jamnani, M. V. and Boutros, M. (2018). Time-resolved mapping of genetic interactions to model rewiring of signaling pathways. Elife 7. PubMed ID: 30592458

    Mattila, J., Kokki, K., Hietakangas, V. and Boutros, M. (2018). Stem cell intrinsic hexosamine metabolism regulates intestinal adaptation to nutrient content. Dev Cell 47(1): 112-121.e113. PubMed ID: 30220570

    Billmann, M., Chaudhary, V., ElMaghraby, M. F., Fischer, B. and Boutros, M. (2017). Widespread rewiring of genetic networks upon cancer signaling pathway activation. Cell Syst. PubMed ID: 29199019

    Merk, K., Breinig, M., Bottcher, R., Krebs, S., Blum, H., Boutros, M. and Forstemann, K. (2017). Splicing stimulates siRNA formation at Drosophila DNA double-strand breaks. PLoS Genet 13(6): e1006861. PubMed ID: 28628606

    Strassburger, K., Lorbeer, F. K., Lutz, M., Graf, F., Boutros, M. and Teleman, A. A. (2017). Oxygenation and adenosine deaminase support growth and proliferation of ex vivo cultured Drosophila wing imaginal discs. Development. PubMed ID: 28526754

  • Vani Brahmachari Ambedkar Center for Biomedical Research, Delhi, India
    Jain, S., Maini, J., Narang, A., Maiti, S. and Brahmachari, V. (2020). The regulatory function of dIno80 correlates with its DNA binding activity. Gene 732: 144368. PubMed ID: 31954859

    Sharma, V., Kohli, S. and Brahmachari, V. (2017). Correlation between desiccation stress response and epigenetic modifications of genes in Drosophila melanogaster: An example of environment-epigenome interaction. Biochim Biophys Acta. PubMed ID: 28801151

    Maini, J., Ghasemi, M., Yandhuri, D., Thakur, S. S. and Brahmachari, V. (2017). Human PRE-PIK3C2B, an intronic cis-element with dual function of activation and repression. Biochim Biophys Acta 1860(2): 196-204. PubMed ID: 27932267

    Mendiratta, S., Bhatia, S., Jain, S., Kaur, T. and Brahmachari, V. (2016). Interaction of the Chromatin Remodeling Protein hINO80 with DNA. PLoS One 11(7): e0159370. PubMed ID: 27428271

  • Andrea Brand Gurdon Institute and The Department of Physiology, University of Cambridge
    Tang, J. L. Y., Krautz, R., Llorà-Batlle, O., Hakes, A. E., Fox, P. M. and Brand, A. H. (2022). In vivo, genome-wide profiling of endogenously tagged chromatin-binding proteins with spatial and temporal resolution using NanoDam in Drosophila. STAR Protoc 3(4): 101788. PubMed ID: 36345375

    Senos Demarco, R., Stack, B. J., Tang, A. M., Voog, J., Sandall, S. L., Southall, T. D., Brand, A. H. and Jones, D. L. (2022). Escargot controls somatic stem cell maintenance through the attenuation of the insulin receptor pathway in Drosophila. Cell Rep 39(3): 110679. PubMed ID: 35443165

    Agrawal, N., Lawler, K., Davidson, C. M., Keogh, J. M., Legg, R., Barroso, I., Farooqi, I. S. and Brand, A. H. (2021). Predicting novel candidate human obesity genes and their site of action by systematic functional screening in Drosophila. PLoS Biol 19(11): e3001255. PubMed ID: 34748544

    Contreras, E. G., Glavic, Á., Brand, A. H. and Sierralta, J. (2021). The serine protease homologue, Scarface, is sensitive to nutrient availability and modulates the development of the Drosophila blood brain barrier. J Neurosci. PubMed ID: 34210781

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

    Hakes, A. E. and Brand, A. H. (2020). Tailless/TLX reverts intermediate neural progenitors to stem cells driving tumourigenesis via repression of asense/ASCL1. Elife 9. PubMed ID: 32073402

    van den Ameele, J. and Brand, A. (2019). Neural stem cell temporal patterning and brain tumour growth rely on oxidative phosphorylation. Elife 8. PubMed ID: 31513013

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

    Otsuki, L. and Brand, A. H. (2019). Dorsal-Ventral Differences in Neural Stem Cell Quiescence Are Induced by p57(KIP2)/Dacapo. Dev Cell 49(2): 293-300.e293. PubMed ID: 30905769

    Jorg, D. J., Caygill, E. E., Hakes, A. E., Contreras, E. G., Brand, A. H. and Simons, B. D. (2019). The proneural wave in the Drosophila optic lobe is driven by an excitable reaction-diffusion mechanism. Elife 8. PubMed ID: 30794154

  • Marko Brankatschk Technische Universität Dresden, Germany
    Prince, E., Kretzschmar, J., Trautenberg, L. C., Broschk, S. and Brankatschk, M. (2021). DIlp7-Producing Neurons Regulate Insulin-Producing Cells in Drosophila. Front Physiol 12: 630390. PubMed ID: 34385929

    Trautenberg, L. C., Knittelfelder, O., Hofmann, C., Shevchenko, A., Brankatschk, M. and Prince, E. (2020). How to use the development of individual Drosophila larvae as a metabolic sensor. J Insect Physiol 126: 104095. PubMed ID: 32783958

    Knittelfelder, O., Prince, E., Sales, S., Fritzsche, E., Wohner, T., Brankatschk, M. and Shevchenko, A. (2020). Sterols as dietary markers for Drosophila melanogaster. Biochim Biophys Acta Mol Cell Biol Lipids 1865(7): 158683. PubMed ID: 32169653

    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

  • Sarah Bray Department of Physiology, Development and Neuroscience, University of Cambridge
    Falo-Sanjuan, J. and Bray, S. (2022). Notch-dependent and -independent transcription are modulated by tissue movements at gastrulation. Elife 11. PubMed ID: 35583918

    Logeay, R., Geminard, C., Lassus, P., Rodriguez-Vazquez, M., Kantar, D., Heron-Milhavet, L., Fischer, B., Bray, S. J., Colinge, J. and Djiane, A. (2022). Mechanisms underlying the cooperation between loss of epithelial polarity and Notch signaling during neoplastic growth in Drosophila. Development. PubMed ID: 35005772

    Martins, T., Meng, Y., Korona, B., Suckling, R., Johnson, S., Handford, P. A., Lea, S. M. and Bray, S. J. (2021). The conserved C2 phospholipid-binding domain in Delta contributes to robust Notch signalling. EMBO Rep: e52729. PubMed ID: 34347930

    Falo-Sanjuan, J. and Bray, S. J. (2021). Membrane architecture and adherens junctions contribute to strong Notch pathway activation. Development. PubMed ID: 34486648

    Feng, S., Zacharioudaki, E., Millen, K. and Bray, S. J. (2020). The SLC36 transporter Pathetic is required for neural stem cell proliferation and for brain growth under nutrition restriction. Neural Dev 15(1): 10. PubMed ID: 32741363

    Boukhatmi, H., Martins, T., Pillidge, Z., Kamenova, T. and Bray, S. (2020). Notch mediates inter-tissue communication to promote tumorigenesis. Curr Biol. PubMed ID: 32275875

    Koca, Y., Housden, B. E., Gault, W. J., Bray, S. J. and Mlodzik, M. (2019). Notch signaling coordinates ommatidial rotation in the Drosophila eye via transcriptional regulation of the EGF-Receptor ligand Argos. Sci Rep 9(1): 18628. PubMed ID: 31819141

    Falo-Sanjuan, J., Lammers, N. C., Garcia, H. G. and Bray, S. J. (2019). Enhancer priming enables fast and sustained transcriptional responses to Notch nignaling. Dev Cell 50(4): 411-425.e418. PubMed ID: 31378591

    Zacharioudaki, E., Falo Sanjuan, J. and Bray, S. (2019). Mi-2/NuRD complex protects stem cell progeny from mitogenic Notch signalling. Elife 8. PubMed ID: 30694174

    Boukhatmi, H. and Bray, S. (2018). A population of adult satellite-like cells in Drosophila is maintained through a switch in RNA-isoforms. Elife 7. PubMed ID: 29629869

    Chan, S. K. K., Cerda-Moya, G., Stojnic, R., Millen, K., Fischer, B., Fexova, S., Skalska, L., Gomez-Lamarca, M., Pillidge, Z., Russell, S. and Bray, S. J. (2017). Role of co-repressor genomic landscapes in shaping the Notch response. PLoS Genet 13(11): e1007096. PubMed ID: 29155828

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

    Terriente-Felix, A., Perez, L., Bray, S. J., Nebreda, A. R. and Milan, M. (2017). Drosophila model of myeloproliferative neoplasm reveals a feed-forward loop in the JAK pathway mediated by p38 MAPK signalling. Dis Model Mech. PubMed ID: 28237966

  • Emilie Brasset Faculté de Médecine, Clermont-Ferrand, France
    Duc, C., Yoth, M., Jensen, S., Mouniee, N., Bergman, C. M., Vaury, C. and Brasset, E. (2019). Trapping a somatic endogenous retrovirus into a germline piRNA cluster immunizes the germline against further invasion. Genome Biol 20(1): 127. PubMed ID: 31227013

    Casier, K., Delmarre, V., Gueguen, N., Hermant, C., Viode, E., Vaury, C., Ronsseray, S., Brasset, E., Teysset, L. and Boivin, A. (2019). Environmentally-induced epigenetic conversion of a piRNA cluster. Elife 8. PubMed ID: 30875295

    Theron, E., Maupetit-Mehouas, S., Pouchin, P., Baudet, L., Brasset, E. and Vaury, C. (2018). The interplay between the Argonaute proteins Piwi and Aub within Drosophila germarium is critical for oogenesis, piRNA biogenesis and TE silencing. Nucleic Acids Res 46(19): 10052-10065. PubMed ID: 30113668

    Duc, C., Yoth, M., Jensen, S., Mouniee, N., Bergman, C. M., Vaury, C. and Brasset, E. (2019). Trapping a somatic endogenous retrovirus into a germline piRNA cluster immunizes the germline against further invasion. Genome Biol 20(1): 127. PubMed ID: 31227013

  • Alexander Brehm Institute for Molecular Biology and Tumour Research, Philipps-University of Marburg
    Ullah, I., Tholken, C., Zhong, Y., John, M., Rossbach, O., Lenz, J., Gossringer, M., Nist, A., Albert, L., Stiewe, T., Hartmann, R., Vazquez, O., Chung, H. R., Mackay, J. P. and Brehm, A. (2022). RNA inhibits dMi-2/CHD4 chromatin binding and nucleosome remodeling. Cell Rep 39(9): 110895. PubMed ID: 35649367
    Lenz, J., Liefke, R., Funk, J., Shoup, S., Nist, A., Stiewe, T., Schulz, R., Tokusumi, Y., Albert, L., Raifer, H., Förstemann, K., Vázquez, O., Tokusumi, T., Fossett, N. and Brehm, A. (2021). Ush regulates hemocyte-specific gene expression, fatty acid metabolism and cell cycle progression and cooperates with dNuRD to orchestrate hematopoiesis. PLoS Genet 17(2): e1009318. PubMed ID: 33600407

    Macinkovic, I., Theofel, I., Hundertmark, T., Kovac, K., Awe, S., Lenz, J., Forne, I., Lamp, B., Nist, A., Imhof, A., Stiewe, T., Renkawitz-Pohl, R., Rathke, C. and Brehm, A. (2019). Distinct CoREST complexes act in a cell-type-specific manner. Nucleic Acids Res. PubMed ID: 31701127

    Kovac, K., Sauer, A., Macinkovic, I., Awe, S., Finkernagel, F., Hoffmeister, H., Fuchs, A., Muller, R., Rathke, C., Langst, G. and Brehm, A. (2018). Tumour-associated missense mutations in the dMi-2 ATPase alters nucleosome remodelling properties in a mutation-specific manner. Nat Commun 9(1): 2112. PubMed ID: 29844320

    Kreher, J., Kovac, K., Bouazoune, K., Macinkovic, I., Ernst, A. L., Engelen, E., Pahl, R., Finkernagel, F., Murawska, M., Ullah, I. and Brehm, A. (2017). EcR recruits dMi-2 and increases efficiency of dMi-2-mediated remodelling to constrain transcription of hormone-regulated genes. Nat Commun 8: 14806. PubMed ID: 28378812

    Theofel, I., Bartkuhn, M., Boettger, T., Gartner, S. M., Kreher, J., Brehm, A. and Rathke, C. (2017). tBRD-1 and tBRD-2 regulate expression of genes necessary for spermatid differentiation. Biol Open. PubMed ID: 28235844

    Fasulo, B., Deuring, R., Murawska, M., Gause, M., Dorighi, K. M., Schaaf, C. A., Dorsett, D., Brehm, A. and Tamkun, J. W. (2012). The Drosophila MI-2 chromatin-remodeling factor regulates higher-order chromatin structure and cohesin dynamics in vivo. PLoS Genet 8: e1002878. PubMed ID: 22912596

    Meier, K., Mathieu, E. L., Finkernagel, F., Reuter, L. M., Scharfe, M., Doehlemann, G., Jarek, M. and Brehm, A. (2012). LINT, a novel dL(3)mbt-containing complex, represses malignant brain tumour signature genes. PLoS Genet 8: e1002676. PubMed ID: 22570633

    Mathieu, E. L., Finkernagel, F., Murawska, M., Scharfe, M., Jarek, M. and Brehm, A. (2012). Recruitment of the ATP-dependent chromatin remodeler dMi-2 to the transcribed region of active heat shock genes. Nucleic Acids Res 40: 4879-4891. PubMed ID: 22362736

  • Björn Brembs Institute of Zoology - Neurogenetics, Universität Regensburg
    Steymans, I., Pujol-Lereis, L. M., Brembs, B. and Gorostiza, E. A. (2021). Collective action or individual choice: Spontaneity and individuality contribute to decision-making in Drosophila. PLoS One 16(8): e0256560. PubMed ID: 34437617

    Damrau, C., Colomb, J. and Brembs, B. (2021). Sensitivity to expression levels underlies differential dominance of a putative null allele of the Drosophila tßh gene in behavioral phenotypes. PLoS Biol 19(5): e3001228. PubMed ID: 33970909

    Palazzo, O., Rass, M. and Brembs, B. (2020). Identification of FoxP circuits involved in locomotion and object fixation in Drosophila. Open Biol 10(12): 200295. PubMed ID: 33321059

    Damrau, C., Toshima, N., Tanimura, T., Brembs, B. and Colomb, J. (2017). Octopamine and tyramine contribute separately to the counter-regulatory response to sugar deficit in Drosophila. Front Syst Neurosci 11: 100. PubMed ID: 29379421

    Gorostiza, E. A., Colomb, J. and Brembs, B. (2016). A decision underlies phototaxis in an insect. Open Biol 6(12). PubMed ID: 28003472

    Colomb, J. and Brembs, B. (2016). PKC in motorneurons underlies self-learning, a form of motor learning in Drosophila. PeerJ 4: e1971. PubMed ID: 27168980

    Colomb, J. and Brembs, B. (2014). Sub-strains of Drosophila Canton-S differ markedly in their locomotor behavior. F1000Res 3: 176. PubMed ID: 25210619

    Mendoza, E., Colomb, J., Rybak, J., Pfluger, H. J., Zars, T., Scharff, C. and Brembs, B. (2014). Drosophila FoxP mutants are deficient in operant self-learning. PLoS One 9: e100648. PubMed ID: 24964149

    Colomb, J., Reiter, L., Blaszkiewicz, J., Wessnitzer, J. and Brembs, B. (2012). Open source tracking and analysis of adult Drosophila locomotion in Buridan's paradigm with and without visual targets. PLoS One 7: e42247. PubMed ID: 22912692

  • Jay Brenman Department of Cell Biology and Physiology, UNC Chapel Hill
    Swick, L. L., Kazgan, N., Onyenwoke, R. U. and Brenman, J. E. (2013). Isolation of AMP-activated protein kinase (AMPK) alleles required for neuronal maintenance in Drosophila melanogaster. Biol Open 2: 1321-1323. PubMed ID: 24337116

    Kern, J. V., Zhang, Y. V., Kramer, S., Brenman, J. E. and Rasse, T. M. (2013). The kinesin-3, unc-104 regulates dendrite morphogenesis and synaptic development in Drosophila. Genetics 195: 59-72. PubMed ID: 23770702

    Braco, J. T., Gillespie, E. L., Alberto, G. E., Brenman, J. E. and Johnson, E. C. (2012). Energy-dependent modulation of glucagon-like signaling in Drosophila via the AMP-activated protein kinase. Genetics 192: 457-466. PubMed ID: 22798489

  • Catherine Brennan California State University, Fullerton
    Myers, A. L., Harris, C. M., Choe, K. M. and Brennan, C. A. (2018). Inflammatory production of reactive oxygen species by Drosophila hemocytes activates cellular immune defenses. Biochem Biophys Res Commun 505(3): 726-732. PubMed ID: 30292413

    Park, S. H., Lee, C. W., Lee, J. H., Park, J. Y., Roshandell, M., Brennan, C. A. and Choe, K. M. (2018). Requirement for and polarized localization of integrin proteins during Drosophila wound closure. Mol Biol Cell 29(18): 2137-2147. PubMed ID: 29995573

    Brennan, C. A., Delaney, J. R., Schneider, D. S. and Anderson, K. V. (2007). Psidin is required in Drosophila blood cells for both phagocytic degradation and immune activation of the fat body. Curr Biol 17(1): 67-72. PubMed ID: 17208189

  • Julius Brennecke Institute of Molecular Biotechnology, Vienna
    Baumgartner, L., Handler, D., Platzer, S. W., Yu, C., Duchek, P. and Brennecke, J. (2022). The Drosophila ZAD zinc finger protein Kipferl guides Rhino to piRNA clusters. Elife 11. PubMed ID: 36193674

    Andreev, V. I., Yu, C., Wang, J., Schnabl, J., Tirian, L., Gehre, M., Handler, D., Duchek, P., Novatchkova, M., Baumgartner, L., Meixner, K., Sienski, G., Patel, D. J. and Brennecke, J. (2022). Panoramix SUMOylation on chromatin connects the piRNA pathway to the cellular heterochromatin machinery. Nat Struct Mol Biol 29(2): 130-142. PubMed ID: 35173350

    ElMaghraby, M. F., Tirian, L., Senti, K. A., Meixner, K. and Brennecke, J. (2021). A genetic toolkit for studying transposon control in the Drosophila melanogaster ovary. Genetics. PubMed ID: 34718559

    ElMaghraby, M. F., Tirian, L., Senti, K. A., Meixner, K. and Brennecke, J. (2021). A genetic toolkit for studying transposon control in the Drosophila melanogaster ovary. Genetics. PubMed ID: 34718559

    Schnabl, J., Wang, J., Hohmann, U., Gehre, M., Batki, J., Andreev, V. I., Purkhauser, K., Fasching, N., Duchek, P., Novatchkova, M., Mechtler, K., Plaschka, C., Patel, D. J. and Brennecke, J. (2021). Molecular principles of Piwi-mediated cotranscriptional silencing through the dimeric SFiNX complex. Genes Dev 35(5-6): 392-409. PubMed ID: 33574069

    ElMaghraby, M. F., Andersen, P. R., Puhringer, F., Hohmann, U., Meixner, K., Lendl, T., Tirian, L. and Brennecke, J. (2019). A heterochromatin-specific RNA export pathway facilitates piRNA production. Cell 178(4): 964-979.e920. PubMed ID: 31398345

    Hayashi, R., Schnabl, J., Handler, D., Mohn, F., Ameres, S. L. and Brennecke, J. (2016). Genetic and mechanistic diversity of piRNA 3'-end formation. Nature [Epub ahead of print]. PubMed ID: 27851737

    Sienski, G., Batki, J., Senti, K. A., Donertas, D., Tirian, L., Meixner, K. and Brennecke, J. (2015). Silencio/CG9754 connects the Piwi-piRNA complex to the cellular heterochromatin machinery. Genes Dev 29: 2258-2271. PubMed ID: 26494711

    Senti, K. A., Jurczak, D., Sachidanandam, R. and Brennecke, J. (2015). piRNA-guided slicing of transposon transcripts enforces their transcriptional silencing via specifying the nuclear piRNA repertoire. Genes Dev 29: 1747-1762. PubMed ID: 26302790

    Mohn, F., Handler, D. and Brennecke, J. (2015). Noncoding RNA. piRNA-guided slicing specifies transcripts for Zucchini-dependent, phased piRNA biogenesis. Science 348: 812-817. PubMed ID: 25977553

    Hayashi, R., Handler, D., Ish-Horowicz, D. and Brennecke, J. (2014). The exon junction complex is required for definition and excision of neighboring introns in Drosophila. Genes Dev. PubMed ID: 25081352

    Mohn, F., Sienski, G., Handler, D. and Brennecke, J. (2014). The Rhino-Deadlock-Cutoff Complex Licenses Noncanonical Transcription of Dual-Strand piRNA Clusters in Drosophila. Cell 157: 1364-1379. PubMed ID: 24906153

  • Amanda Bretman Faculty of Biological Sciences, University of Leeds
    Leech, T., McDowall, L., Hopkins, K. P., Sait, S. M., Harrison, X. A. and Bretman, A. (2021). Social environment drives sex and age-specific variation in Drosophila melanogaster microbiome composition and predicted function. Mol Ecol. PubMed ID: 34494339

    Dore, A. A., Bretman, A. and Chapman, T. (2020). Fitness consequences of redundant cues of competition in male Drosophila melanogaster. Ecol Evol 10(12): 5517-5526. PubMed ID: 32607171

    Leech, T., Evison, S. E. F., Armitage, S. A. O., Sait, S. M. and Bretman, A. (2019). Interactive effects of social environment, age and sex on immune responses in Drosophila melanogaster. J Evol Biol 32(10): 1082-1092. PubMed ID: 31313398

    Rouse, J., Watkinson, K. and Bretman, A. (2018). Flexible memory controls sperm competition responses in male Drosophila melanogaster. Proc Biol Sci 285(1879). PubMed ID: 29848652

    Rouse, J., McDowall, L., Mitchell, Z., Duncan, E. J. and Bretman, A. (2020). Social competition stimulates cognitive performance in a sex-specific manner. Proc Biol Sci 287(1935): 20201424. PubMed ID: 32933446

  • Julie Brill Hospital for Sick Children, University of Toronto
    Shao, L., Fingerhut, J. M., Falk, B. L., Han, H., Maldonado, G., Qiao, Y., Lee, V., Hall, E., Chen, L., Polevoy, G., Hernandez, G., Lasko, P. and Brill, J. A. (2023). Eukaryotic translation initiation factor eIF4E-5 is required for spermiogenesis in Drosophila melanogaster. Development. PubMed ID: 36695474

    Ma, C. J. and Brill, J. A. (2021). Endosomal Rab GTPases regulate secretory granule maturation in Drosophila larval salivary glands. Commun Integr Biol 14(1): 15-20. PubMed ID: 33628358

    Griffiths, N. W., Del Bel, L. M., Wilk, R. and Brill, J. A. (2020). Cellular homeostasis in the Drosophila retina requires the lipid phosphatase Sac1. Mol Biol Cell: mbcE20020161. PubMed ID: 32186963

    Gupta, A., Fabian, L. and Brill, J. A. (2018). Phosphatidylinositol 4,5-bisphosphate regulates cilium transition zone maturation in Drosophila melanogaster. J Cell Sci. PubMed ID: 30054387

    Del Bel, L. M., Griffiths, N., Wilk, R., Wei, H. C., Blagoveshchenskaya, A., Burgess, J., Polevoy, G., Price, J. V., Mayinger, P. and Brill, J. A. (2018). The phosphoinositide phosphatase Sac1 regulates cell shape and microtubule stability in the developing Drosophila eye. Development 145(11). PubMed ID: 29752385

    Tan, J., Oh, K., Burgess, J., Hipfner, D. R. and Brill, J. A. (2014). PI4KIIIalpha is required for cortical integrity and cell polarity during Drosophila oogenesis. J Cell Sci. PubMed ID: 24413170

    Fabian, L. and Brill, J. A. (2012). Drosophila spermiogenesis: Big things come from little packages. Spermatogenesis 2: 197-212. PubMed ID: 23087837

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

    Hernandez, G., Han, H., Gandin, V., Fabian, L., Ferreira, T., Zuberek, J., Sonenberg, N., Brill, J. A. and Lasko, P. (2012). Eukaryotic initiation factor 4E-3 is essential for meiotic chromosome segregation, cytokinesis and male fertility in Drosophila. Development 139: 3211-3220. PubMed ID: 22833128

  • Steve Britt Neurology, Dell Medical School, University of Texas at Austin
    Tan, H., Fulton, R. E., Chou, W. H., Birkholz, D. A., Mannino, M. P., Yamaguchi, D. M., Aldrich, J. C., Jacobsen, T. L. and Britt, S. G. (2020). Drosophila R8 photoreceptor cell subtype specification requires hibris. PLoS One 15(10): e0240451. PubMed ID: 33052948

    Tan, H., Fulton, R. E., Chou, W. H., Birkholz, D. A., Mannino, M. P., Yamaguchi, D. M., Aldrich, J. C., Jacobsen, T. L. and Britt, S. G. (2020). Drosophila R8 photoreceptor cell subtype specification requires hibris. PLoS One 15(10): e0240451. PubMed ID: 33052948

    Earl, J. B., Vanderlinden, L. A., Jacobsen, T. L., Aldrich, J. C., Saba, L. M. and Britt, S. G. (2020). Identification of Genes Involved in the Differentiation of R7y and R7p Photoreceptor Cells in Drosophila. G3 (Bethesda) 10(11): 3949-3958. PubMed ID: 32972998

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

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

  • Kendal Broadie Vanderbilt University
    Song, C. and Broadie, K. (2023). Fragile X mental retardation protein coordinates neuron-to-glia communication for clearance of developmentally transient brain neurons. Proc Natl Acad Sci U S A 120(12): e2216887120. PubMed ID: 36920921

    Leahy, S. N., Song, C., Vita, D. J. and Broadie, K. (2023). FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 21(1): e3001969. PubMed ID: 36701299

    Sears, J. C. and Broadie, K. (2022). Temporally and Spatially Localized PKA Activity within Learning and Memory Circuitry Regulated by Network Feedback. eNeuro 9(2). PubMed ID: 35301221

    Song, C., Leahy, S. N., Rushton, E. M. and Broadie, K. (2022). RNA-binding FMRP and Staufen sequentially regulate the coracle scaffold to control synaptic glutamate receptor and bouton development. Development. PubMed ID: 35394012

    Bhimreddy, M., Rushton, E., Kopke, D. L. and Broadie, K. (2021). Secreted C-type lectin regulation of neuromuscular junction synaptic vesicle dynamics modulates coordinated movement. J Cell Sci 134(9). PubMed ID: 33973638

    Vita, D. J., Meier, C. J. and Broadie, K. (2021). Neuronal fragile X mental retardation protein activates glial insulin receptor mediated PDF-Tri neuron developmental clearance. Nat Commun 12(1): 1160. PubMed ID: 33608547

    Golovin, R. M., Vest, J. and Broadie, K. (2021). Neuron-Specific FMRP Roles in Experience-Dependent Remodeling of Olfactory Brain Innervation During an Early-life Critical Period. J Neurosci. PubMed ID: 33402421

    Sears, J. C. and Broadie, K. (2020). FMRP-PKA Activity Negative Feedback Regulates RNA Binding-Dependent Fibrillation in Brain Learning and Memory Circuitry. Cell Rep 33(2): 108266. PubMed ID: 33053340

    Rushton, E., Kopke, D. L. and Broadie, K. (2020). Extracellular heparan sulfate proteoglycans and glycan-binding lectins orchestrate trans-synaptic signaling. J Cell Sci 133(15). PubMed ID: 32788209

    Kennedy, T., Rinker, D. and Broadie, K. (2020). Genetic background mutations drive neural circuit hyperconnectivity in a fragile X syndrome model. BMC Biol 18(1): 94. PubMed ID: 32731855

  • Nichole Broderick Department of Molecular and Cell Biology, University of Connecticut, Storrs
    Barron, A. J., Lesperance, D. N. A., Doucette, J., Calle, S. and Broderick, N. A. (2023). Microbiome derived acidity protects against microbial invasion in Drosophila. bioRxiv. PubMed ID: 36711873

    Lesperance, D. N. A. and Broderick, N. A. (2020). Gut bacteria mediate nutrient availability in Drosophila diets. Appl Environ Microbiol. PubMed ID: 33067193'

    Fischer, C., Trautman, E. P., Crawford, J. M., Stabb, E. V., Handelsman, J. and Broderick, N. A. (2017). Metabolite exchange between microbiome members produces compounds that influence Drosophila behavior. Elife 6. PubMed ID: 28068220

    Broderick, N. A. (2016). Friend, foe or food? Recognition and the role of antimicrobial peptides in gut immunity and Drosophila-microbe interactions. Philos Trans R Soc Lond B Biol Sci 371(1695). PubMed ID: 27160597

    Broderick, N. A., Buchon, N. and Lemaitre, B. (2014). Microbiota-induced changes in drosophila melanogaster host gene expression and gut morphology. MBio 5(3): e01117-01114. PubMed ID: 24865556

  • Heather Broihier Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio
    Jindal, D. A., Leier, H. C., Salazar, G., Foden, A. J., Seitz, E. A., Wilkov, A. J., Coutinho-Budd, J. C. and Broihier, H. T. (2023). Early Draper-mediated glial refinement of neuropil architecture and synapse number in the Drosophila antennal lobe. Front Cell Neurosci 17: 1166199. PubMed ID: 37333889

    Hoover, K. M., Gratz, S. J., Qi, N., Herrmann, K. A., Liu, Y., Perry-Richardson, J. J., Vanderzalm, P. J., O'Connor-Giles, K. M. and Broihier, H. T. (2019). The calcium channel subunit alpha2delta-3 organizes synapses via an activity-dependent and autocrine BMP signaling pathway. Nat Commun 10(1): 5575. PubMed ID: 31811118

    Sears, J. C. and Broihier, H. T. (2016). FoxO regulates microtubule dynamics and polarity to promote dendrite branching in Drosophila sensory neurons. Dev Biol [Epub ahead of print]. PubMed ID: 27546375

    McLaughlin, C. N., Nechipurenko, I. V., Liu, N. and Broihier, H. T. (2016). A Toll receptor-FoxO pathway represses Pavarotti/MKLP1 to promote microtubule dynamics in motoneurons. J Cell Biol 214: 459-474. PubMed ID: 27502486

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

    Broihier, H. (2012). Whole-mount fluorescence in situ hybridization and antibody staining of Drosophila embryos. Cold Spring Harb Protoc 2012. PubMed ID: 22854564

    James, R. E. and Broihier, H. T. (2011). Crimpy inhibits the BMP homolog Gbb in motoneurons to enable proper growth control at the Drosophila neuromuscular junction. Development 138: 3273-3286. PubMed ID: 21750037

    Miller, C. M., Liu, N., Page-McCaw, A. and Broihier, H. T. (2011). Drosophila MMP2 regulates the matrix molecule faulty attraction (Frac) to promote motor axon targeting in Drosophila. J Neurosci 31: 5335-5347. PubMed ID: 21471368

    Weng, Y. L., Liu, N., DiAntonio, A. and Broihier, H. T. (2011). The cytoplasmic adaptor protein Caskin mediates Lar signal transduction during Drosophila motor axon guidance. J Neurosci 31: 4421-4433. PubMed ID: 21430143

  • William Brook Department of Biochemistry and Molecular Biology, University of Calgary
    Phillips, L. A., Atienza, M. L., Ryu, J. R., Svendsen, P. C., Kelemen, L. K. and Brook, W. J. (2022). midline represses Dpp signaling and target gene expression in Drosophila ventral leg development. Biol Open 11(5). PubMed ID: 35608103

    Svendsen, P. C., Phillips, L. A., Deshwar, A. R., Ryu, J. R., Najand, N. and Brook, W. J. (2019). The selector genes midline and H15 control ventral leg pattern by both inhibiting Dpp signaling and specifying ventral fate. Dev Biol. PubMed ID: 31299230

    Najand, N., Ryu, J. R. and Brook, W. J. (2012). In vitro site selection of a consensus binding site for the Drosophila melanogaster Tbx20 homolog midline. PLoS One 7(10): e48176. PubMed ID: 23133562

    Ryu, J. R., Najand, N. and Brook, W. J. (2011). Tinman is a direct activator of midline in the Drosophila dorsal vessel. Dev Dyn 240(1): 86-95. PubMed ID: 21108319

    Svendsen, P. C., Formaz-Preston, A., Leal, S. M. and Brook, W. J. (2009). The Tbx20 homologs midline and H15 specify ventral fate in the Drosophila melanogaster leg. Development 136(16): 2689-2693. PubMed ID: 19605497

  • Nick Brown Wellcome Trust/Cancer Research UK, Gurdon Institute, Department of Physiology, Development and Neuroscience, University of Cambridge
    Green, H. J., Griffiths, A. G. M., Ylanne, J. and Brown, N. H. (2018). Novel functions for integrin-associated proteins revealed by analysis of myofibril attachment in Drosophila. Elife 7. PubMed ID: 30028294

    Bulgakova, N. A., Wellmann, J. and Brown, N. H. (2017). Diverse integrin adhesion stoichiometries caused by varied actomyosin activity. Open Biol 7(4). PubMed ID: 28446705

    Maartens, A.P., Wellmann, J., Wictome, E., Klapholz, B., Green, H. and Brown, N.H. (2016). Drosophila vinculin is more harmful when hyperactive than absent, and can circumvent integrin to form adhesion complexes. J Cell Sci [Epub ahead of print]. PubMed ID: 27737911

    Huelsmann, S., Rintanen, N., Sethi, R., Brown, N. H. and Ylanne, J. (2016). Evidence for the mechanosensor function of filamin in tissue development. Sci Rep 6: 32798. PubMed ID: 27597179

    Klapholz, B., Herbert, S. L., Wellmann, J., Johnson, R., Parsons, M. and Brown, N. H. (2015). Alternative mechanisms for Talin to mediate integrin function. Curr Biol [Epub ahead of print]. PubMed ID: 25754646

    Huelsmann, S., Ylanne, J. and Brown, N. H. (2013). Filopodia-like Actin Cables Position Nuclei in Association with Perinuclear Actin in Drosophila Nurse Cells. Dev Cell 26: 604-615. PubMed ID: 24091012

    Bulgakova, N. A., Grigoriev, I., Yap, A. S., Akhmanova, A. and Brown, N. H. (2013). Dynamic microtubules produce an asymmetric E-cadherin-Bazooka complex to maintain segment boundaries. J Cell Biol 201: 887-901. PubMed ID: 23751496

    Bulgakova, N. A., Klapholz, B. and Brown, N. H. (2012). Cell adhesion in Drosophila: versatility of cadherin and integrin complexes during development. Curr Opin Cell Biol 24: 702-712. PubMed ID: 22938782

    Zervas, C. G., Psarra, E., Williams, V., Solomon, E., Vakaloglou, K. M. and Brown, N. H. (2011). A central multifunctional role of integrin-linked kinase at muscle attachment sites. J Cell Sci 124: 1316-1327. PubMed ID: 21444757

    Sabino, D., Brown, N. H. and Basto, R. (2011). Drosophila Ajuba is not an Aurora-A activator but is required to maintain Aurora-A at the centrosome. J Cell Sci 124: 1156-1166. PubMed ID: 21402878

  • Katja Brückner Cell & Tissue Biology, University of California, San Francisco
    Sanchez Bosch, P., Makhijani, K., Herboso, L., Gold, K. S., Baginsky, R., Woodcock, K. J., Alexander, B., Kukar, K., Corcoran, S., Jacobs, T., Ouyang, D., Wong, C., Ramond, E. J. V., Rhiner, C., Moreno, E., Lemaitre, B., Geissmann, F. and Bruckner, K. (2019). Adult Drosophila lack hematopoiesis but rely on a blood cell reservoir at the respiratory epithelia to relay infection signals to surrounding tissues. Dev Cell. PubMed ID: 31735669

    Makhijani, K., Alexander, B., Rao, D., Petraki, S., Herboso, L., Kukar, K., Batool, I., Wachner, S., Gold, K. S., Wong, C., O'Connor, M. B. and Bruckner, K. (2017). Regulation of Drosophila hematopoietic sites by Activin-beta from active sensory neurons. Nat Commun 8: 15990. PubMed ID: 28748922

    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

    Sopko, R., Lin, Y. B., Makhijani, K., Alexander, B., Perrimon, N. and Bruckner, K. (2015). A systems-level interrogation identifies regulators of Drosophila blood cell number and survival. PLoS Genet 11: e1005056. PubMed ID: 25749252

    Gold, K. S. and Bruckner, K. (2014). Drosophila as a model for the two myeloid blood cell systems in vertebrates. Exp Hematol 42: 717-727. PubMed ID: 24946019

    Makhijani, K. and Bruckner, K. (2012). Of blood cells and the nervous system: hematopoiesis in the Drosophila larva. Fly (Austin) 6: 254-260. PubMed ID: 23022764

  • Erich Buchner Institute of Clinical Neurobiology, Wuerzburg
    Blanco-Redondo, B., Nuwal, N., Kneitz, S., Nuwal, T., Halder, P., Liu, Y., Ehmann, N., Scholz, N., Mayer, A., Kleber, J., Kahne, T., Schmitt, D., Sadanandappa, M. K., Funk, N., Albertova, V., Helfrich-Forster, C., Ramaswami, M., Hasan, G., Kittel, R. J., Langenhan, T., Gerber, B. and Buchner, E. (2019). Implications of the Sap47 null mutation for synapsin phosphorylation, longevity, climbing, and behavioural plasticity in adult Drosophila. J Exp Biol. PubMed ID: 31488622

    Redondo, B. B., Bunz, M., Halder, P., Sadanandappa, M. K., Muhlbauer, B., Erwin, F., Hofbauer, A., Rodrigues, V., Vijayraghavan, K., Ramaswami, M., Rieger, D., Wegener, C., Forster, C. and Buchner, E. (2013). Identification and structural characterization of interneurons of the Drosophila brain by monoclonal antibodies of the wurzburg hybridoma library. PLoS One 8: e75420. PubMed ID: 24069413

    Nuwal, T., Kropp, M., Wegener, S., Racic, S., Montalban, I. and Buchner, E. (2012). The Drosophila homologue of tubulin-specific chaperone E-like protein is required for synchronous sperm individualization and normal male fertility. J Neurogenet 26: 374-381. PubMed ID: 23121292

    Nuwal, N., Stock, P., Hiemeyer, J., Schmid, B., Fiala, A. and Buchner, E. (2012). Avoidance of heat and attraction to optogenetically induced sugar sensation as operant behavior in adult Drosophila. J Neurogenet 26: 298-305. PubMed ID: 22834571

    Halder, P., Chen, Y. C., Brauckhoff, J., Hofbauer, A., Dabauvalle, M. C., Lewandrowski, U., Winkler, C., Sickmann, A. and Buchner, E. (2011). Identification of Eps15 as antigen recognized by the monoclonal antibodies aa2 and ab52 of the Wuerzburg Hybridoma Library against Drosophila brain. PLoS One 6: e29352. PubMed ID: 22206011

  • Nicolas Buchon Department of Entomology, Cornell University, NY
    Bonfini, A., Dobson, A. J., Duneau, D., Revah, J., Liu, X., Houtz, P. and Buchon, N. (2021). Multiscale analysis reveals that diet-dependent midgut plasticity emerges from alterations in both stem cell niche coupling and enterocyte size. Elife 10. PubMed ID: 34553686

    White, M. A., Bonfini, A., Wolfner, M. F. and Buchon, N. (2021). Drosophila melanogaster sex peptide regulates mated female midgut morphology and physiology. Proc Natl Acad Sci U S A 118(1). PubMed ID: 33443193

    Troha, K., Nagy, P., Pivovar, A., Lazzaro, B. P., Hartley, P. S. and Buchon, N. (2019). Nephrocytes remove microbiota-derived peptidoglycan from systemic circulation to maintain immune homeostasis. Immunity 51(4): 625-637.e623. PubMed ID: 31564469

    Houtz, P., Bonfini, A., Bing, X. and Buchon, N. (2019). Recruitment of adult precursor cells underlies limited repair of the infected larval midgut in Drosophila. Cell Host Microbe 26(3): 412-425. PubMed ID: 31492656

    Houtz, P., Bonfini, A., Bing, X. and Buchon, N. (2019). Recruitment of Adult Precursor Cells Underlies Limited Repair of the Infected Larval Midgut in Drosophila. Cell Host Microbe. PubMed ID: 31492656

    Duneau, D., Sun, H., Revah, J., San Miguel, K., Kunerth, H. D., Caldas, I. V., Messer, P. W., Scott, J. G. and Buchon, N. (2018). Signatures of insecticide selection in the genome of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 30190420

    Troha, K., Im, J. H., Revah, J., Lazzaro, B. P. and Buchon, N. (2018). Comparative transcriptomics reveals CrebA as a novel regulator of infection tolerance in D. melanogaster. PLoS Pathog 14(2): e1006847. PubMed ID: 29394281

    Sannino, D. R., Dobson, A. J., Edwards, K., Angert, E. R. and Buchon, N. (2018). The Drosophila melanogaster gut microbiota provisions thiamine to its host. MBio 9(2). PubMed ID: 29511074

    Troha, K., Im, J. H., Revah, J., Lazzaro, B. P. and Buchon, N. (2018). Comparative transcriptomics reveals CrebA as a novel regulator of infection tolerance in D. melanogaster. PLoS Pathog 14(2): e1006847. PubMed ID: 29394281

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

  • Graham Budd Department of Earth Sciences, Uppsala University
    Janssen, R. and Budd, G. E. (2022). Expression of netrin and its receptors uncoordinated-5 and frazzled in arthropods and onychophorans suggests conserved and diverged functions in neuronal pathfinding and synaptogenesis. Dev Dyn. PubMed ID: 35112412

    Janssen, R., Schomburg, C., Prpic, N. M. and Budd, G. E. (2022). A comprehensive study of arthropod and onychophoran Fox gene expression patterns. PLoS One 17(7): e0270790. PubMed ID: 35802758

    Medina-Jimenez, B. I., Budd, G. E. and Janssen, R. (2021). Panarthropod tiptop/teashirt and spalt orthologs and their potential role as "trunk"-selector genes. Evodevo 12(1): 7. PubMed ID: 34078450

    Panara, V., Budd, G. E. and Janssen, R. (2019). Phylogenetic analysis and embryonic expression of panarthropod Dmrt genes. Front Zool 16: 23. PubMed ID: 31303887

  • Vivian Budnik University of Massachusetts Worcester Campus
    Ashley, J., Cordy, B., Lucia, D., Fradkin, L. G., Budnik, V. and Thomson, T. (2018). Retrovirus-like Gag Protein Arc1 Binds RNA and Traffics across Synaptic Boutons. Cell 172(1-2): 262-274.e211. PubMed ID: 29328915

    Li, Y., Hassinger, L., Thomson, T., Ding, B., Ashley, J., Hassinger, W. and Budnik, V. (2016). Lamin Mutations Accelerate Aging via Defective Export of Mitochondrial mRNAs through Nuclear Envelope Budding. Curr Biol [Epub ahead of print]. PubMed ID: 27451905

    Packard, M., Jokhi, V., Ding, B., Ruiz-Canada, C., Ashley, J. and Budnik, V. (2015). Nucleus to Synapse Nesprin1 Railroad Tracks Direct Synapse Maturation through RNA Localization. Neuron [Epub ahead of print]. PubMed ID: 25959729

    Kerr, K. S., Fuentes-Medel, Y., Brewer, C., Barria, R., Ashley, J., Abruzzi, K. C., Sheehan, A., Tasdemir-Yilmaz, O. E., Freeman, M. R. and Budnik, V. (2014). Glial wingless/wnt regulates glutamate receptor clustering and synaptic physiology at the Drosophila neuromuscular junction. J Neurosci 34: 2910-2920. PubMed ID: 24553932

    Jokhi, V., Ashley, J., Nunnari, J., Noma, A., Ito, N., Wakabayashi-Ito, N., Moore, M. J. and Budnik, V. (2013). Torsin mediates primary envelopment of large ribonucleoprotein granules at the nuclear envelope. Cell Rep 3: 988-995. PubMed ID: 23583177

    Koles, K. and Budnik, V. (2012). Exosomes go with the Wnt. Cell Logist 2: 169-173. PubMed ID: 23739155

    Korkut, C., Li, Y., Koles, K., Brewer, C., Ashley, J., Yoshihara, M., Budnik, V. (2013) Regulation of postsynaptic retrograde signaling by presynaptic exosome release. Neuron 77: 1039-1046. PubMed ID: 23522040

    Fuentes-Medel, Y., Ashley, J., Barria, R., Maloney, R., Freeman, M. and Budnik, V. (2012). Integration of a retrograde signal during synapse formation by glia-secreted TGF-beta ligand. Curr Biol 22: 1831-1838. PubMed ID: 22959350

    Koles, K., et al. (2012). Mechanism of Evenness interrupted (Evi)-exosome release at synaptic boutons. J. Biol. Chem 287: 16820-34. PubMed ID: 22437826

    Speese, S. D., Ashley, J., Jokhi, V., Nunnari, J., Barria, R., Li, Y., Ataman, B., Koon, A., Chang, Y. T., Li, Q., Moore, M. J. and Budnik, V. (2012). Nuclear envelope budding enables large ribonucleoprotein particle export during synaptic Wnt signaling. Cell 149: 832-846. PubMed ID: 22579286

  • Natalia Bulgakova Department of Biomedical Science, United Kingdom
    Moreno, M. R., Boswell, K., Casbolt, H. L. and Bulgakova, N. A. (2022). Multifaceted control of E-cadherin dynamics by the Adaptor Protein Complex 1 during epithelial morphogenesis. Mol Biol Cell: mbcE21120598. PubMed ID: 35609212

    Greig, J. and Bulgakova, N. A. (2020). Interplay between actomyosin and E-cadherin dynamics regulates cell shape in the Drosophila embryonic epidermis. J Cell Sci 133(15). PubMed ID: 32665321

    Bulgakova, N. A., Wellmann, J. and Brown, N. H. (2017). Diverse integrin adhesion stoichiometries caused by varied actomyosin activity. Open Biol 7(4). PubMed ID: 28446705

    Gomez, J. M., Chumakova, L., Bulgakova, N. A. and Brown, N. H. (2016). Microtubule organization is determined by the shape of epithelial cells. Nat Commun 7: 13172. PubMed ID: 27779189

  • Lori Buhlman
    Houlihan, K. L., Keoseyan, P. P., Juba, A. N., Margaryan, T., Voss, M. E., Babaoghli, A. M., Norris, J. M., Adrian, G. J., Tovmasyan, A. and Buhlman, L. M. (2022). Folic Acid Improves Parkin-Null Drosophila Phenotypes and Transiently Reduces Vulnerable Dopaminergic Neuron Mitochondrial Hydrogen Peroxide Levels and Glutathione Redox Equilibrium. Antioxidants (Basel) 11(10). PubMed ID: 36290790

    Cackovic, J., Gutierrez-Luke, S., Call, G. B., Juba, A., O'Brien, S., Jun, C. H. and Buhlman, L. M. (2018). Vulnerable parkin loss-of-function Drosophila dopaminergic neurons have advanced mitochondrial aging, mitochondrial network loss and transiently reduced autophagosome recruitment. Front Cell Neurosci 12: 39. PubMed ID: 29497364

    Chambers, R. P., Call, G. B., Meyer, D., Smith, J., Techau, J. A., Pearman, K. and Buhlman, L. M. (2013). Nicotine increases lifespan and rescues olfactory and motor deficits in a Drosophila model of Parkinson's disease. Behav Brain Res 253: 95-102. PubMed ID: 23871228

  • Raymond BujdosoDepartment of Veterinary Medicine, Cambridge Veterinary School, Cambridge, U.K.
    Thackray, A. M., Lam, B., McNulty, E. E., Nalls, A. V., Mathiason, C. K., Magadi, S. S., Jackson, W. S., Andreoletti, O., Marrero-Winkens, C., Schatzl, H. and Bujdoso, R. (2022). Clearance of variant Creutzfeldt-Jakob disease prions in vivo by the Hsp70 disaggregase system. Brain 145(9): 3236-3249. PubMed ID: 35446941

    Thackray, A. M., Andreoletti, O., Spiropoulos, J. and Bujdoso, R. (2021). A new model for sensitive detection of zoonotic prions by PrP transgenic Drosophila. J Biol Chem 297(2): 100878. PubMed ID: 34270959

    Thackray, A. M., Lam, B., Shahira Binti Ab Razak, A., Yeo, G. and Bujdoso, R. (2020). Transcriptional signature of prion-induced neurotoxicity in a Drosophila model of transmissible mammalian prion disease. Biochem J 477(4): 833-852. PubMed ID: 32108870

  • Simon Bullock MRC Laboratory of Molecular Biology, Cambridge
    Salvador-Garcia, D., Jin, L., Hensley, A., Golcuk, M., Gallaud, E., Chaaban, S., Port, F., Vagnoni, A., Planelles-Herrero, V. J., McClintock, M. A., Derivery, E., Carter, A. P., Giet, R., Gur, M., Yildiz, A. and Bullock, S. L. (2023). A force-sensitive mutation reveals a spindle assembly checkpoint-independent role for dynein in anaphase progression. bioRxiv. PubMed ID: 37577480

    Vagnoni, A. and Bullock, S. L. (2018). A cAMP/PKA/Kinesin-1 axis promotes the axonal transport of mitochondria in aging Drosophila neurons. Curr Biol 28(8): 1265-1272.e1264. PubMed ID: 29606421

    Vagnoni, A., Hoffmann, P. C. and Bullock, S. L. (2015). Reducing Lissencephaly-1 levels augments mitochondrial transport and has a protective effect in adult Drosophila neurons. J Cell Sci [Epub ahead of print]. PubMed ID: 26598558

    Port, F., Chen, H. M., Lee, T. and Bullock, S. L. (2014). Optimized CRISPR/Cas tools for efficient germline and somatic genome engineering in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 25002478

    Soundararajan, H. C. and Bullock, S. L. (2014). The influence of dynein processivity control, MAPs, and microtubule ends on directional movement of a localising mRNA. Elife 3: e01596. PubMed ID: 24737859

    Dix, C. I., Soundararajan, H. C., Dzhindzhev, N. S., Begum, F., Suter, B., Ohkura, H., Stephens, E. and Bullock, S. L. (2013). Lissencephaly-1 promotes the recruitment of dynein and dynactin to transported mRNAs. J Cell Biol 202: 479-494. PubMed ID: 23918939

    Liu, Y., Salter, H. K., Holding, A. N., Johnson, C. M., Stephens, E., Lukavsky, P. J., Walshaw, J. and Bullock, S. L. (2013). Bicaudal-D uses a parallel, homodimeric coiled coil with heterotypic registry to coordinate recruitment of cargos to dynein. Genes Dev. PubMed ID: 23723415

    Amrute-Nayak, M. and Bullock, S. L. (2012). Single-molecule assays reveal that RNA localization signals regulate dynein-dynactin copy number on individual transcript cargoes. Nat Cell Biol 14: 416-423. PubMed ID: 22366687

    Bullock, S. L. (2011). Messengers, motors and mysteries: sorting of eukaryotic mRNAs by cytoskeletal transport. Biochem Soc Trans 39: 1161-1165. PubMed ID: 21936782

    Bianco, A., Dienstbier, M., Salter, H. K., Gatto, G. and Bullock, S. L. (2010). Bicaudal-D regulates fragile X mental retardation protein levels, motility, and function during neuronal morphogenesis. Curr Biol 20: 1487-1492. PubMed ID: 20691595

  • Martha Bulyk Medicine and Pathology, Harvard Medical School, Boston
    Waters, C. T., Gisselbrecht, S. S., Sytnikova, Y. A., Cafarelli, T. M., Hill, D. E. and Bulyk, M. L. (2021). Quantitative-enhancer-FACS-seq (QeFS) reveals epistatic interactions among motifs within transcriptional enhancers in developing Drosophila tissue. Genome Biol 22(1): 348. PubMed ID: 34930411

    Gisselbrecht, S. S., Palagi, A., Kurland, J. V., Rogers, J. M., Ozadam, H., Zhan, Y., Dekker, J. and Bulyk, M. L. (2019). Transcriptional silencers in Drosophila serve a dual role as transcriptional enhancers in alternate cellular vontexts. Mol Cell. PubMed ID: 31704182

    Shokri, L., Inukai, S., Hafner, A., Weinand, K., Hens, K., Vedenko, A., Gisselbrecht, S. S., Dainese, R., Bischof, J., Furger, E., Feuz, J. D., Basler, K., Deplancke, B. and Bulyk, M. L. (2019). A comprehensive Drosophila melanogaster transcription factor interactome. Cell Rep 27(3): 955-970. PubMed ID: 30995488

    Gisselbrecht, S. S., Barrera, L. A., Porsch, M., Aboukhalil, A., Estep, P. W., 3rd, Vedenko, A., Palagi, A., Kim, Y., Zhu, X., Busser, B. W., Gamble, C. E., Iagovitina, A., Singhania, A., Michelson, A. M. and Bulyk, M. L. (2013). Highly parallel assays of tissue-specific enhancers in whole Drosophila embryos. Nat Methods. PubMed ID: 23852450

    Busser, B. W., Huang, D., Rogacki, K. R., Lane, E. A., Shokri, L., Ni, T., Gamble, C. E., Gisselbrecht, S. S., Zhu, J., Bulyk, M. L., Ovcharenko, I. and Michelson, A. M. (2012). Integrative analysis of the zinc finger transcription factor Lame duck in the Drosophila myogenic gene regulatory network. Proc Natl Acad Sci U S A 109: 20768-20773. PubMed ID: 23184988

    Zhu, X., Ahmad, S. M., Aboukhalil, A., Busser, B. W., Kim, Y., Tansey, T. R., Haimovich, A., Jeffries, N., Bulyk, M. L. and Michelson, A. M. (2012). Differential regulation of mesodermal gene expression by Drosophila cell type-specific Forkhead transcription factors. Development 139: 1457-1466. PubMed ID: 22378636

    Busser, B. W., Shokri, L., Jaeger, S. A., Gisselbrecht, S. S., Singhania, A., Berger, M. F., Zhou, B., Bulyk, M. L. and Michelson, A. M. (2012). Molecular mechanism underlying the regulatory specificity of a Drosophila homeodomain protein that specifies myoblast identity. Development 139: 1164-1174. PubMed ID: 22296846

  • Richard Burke Faculty of Science, Monash University, Melbourne
    Zhang, B., Binks, T. and Burke, R. (2020). The E3 ubiquitin ligase Slimb/beta-TrCP is required for normal copper homeostasis in Drosophila. Biochim Biophys Acta Mol Cell Res: 118768. PubMed ID: 32502619

    Richards, C. D., Warr, C. G. and Burke, R. (2017). A role for the Drosophila zinc transporter Zip88E in protecting against dietary zinc toxicity. PLoS One 12(7): e0181237. PubMed ID: 28704512

    Mercer, S. W., La Fontaine, S., Warr, C. G. and Burke, R. (2016). Reduced glutathione biosynthesis in Drosophila melanogaster causes neuronal defects linked to copper deficiency. J Neurochem [Epub ahead of print]. PubMed ID: 26851457

    Richards, C. D. and Burke, R. (2015). Local and systemic effects of targeted zinc redistribution in Drosophila neuronal and gastrointestinal tissues. Biometals [Epub ahead of print]. PubMed ID: 26411574

    Jones, M. W., de Jonge, M. D., James, S. A. and Burke, R. (2015). Elemental mapping of the entire intact Drosophila gastrointestinal tract. J Biol Inorg Chem. PubMed ID: 26153547

    Dechen, K., Richards, C. D., Lye, J. C., Hwang, J. E. and Burke, R. (2015). Compartmentalized zinc deficiency and toxicities caused by ZnT and Zip gene over expression result in specific phenotypes in Drosophila. Int J Biochem Cell Biol. PubMed ID: 25562517

    Hwang, J. E., de Bruyne, M., Warr, C. G. and Burke, R. (2014). Copper overload and deficiency both adversely affect the central nervous system of Drosophila. Metallomics. PubMed ID: 25322772

    Wang, J., Binks, T., Warr, C. G. and Burke, R. (2014). Vacuolar-type H-ATPase subunits and the neurogenic protein Big brain are required for optimal copper and zinc uptake. Metallomics. PubMed ID: 25209718

    Lye, J. C., Richards, C. D., Dechen, K., Warr, C. G. and Burke, R. (2013). In vivo zinc toxicity phenotypes provide a sensitized background that suggests zinc transport activities for most of the Drosophila Zip and ZnT genes. J Biol Inorg Chem 18: 323-332. PubMed ID: 23322169

    Lye, J. C., Richards, C. D., Dechen, K., Paterson, D., de Jonge, M. D., Howard, D. L., Warr, C. G. and Burke, R. (2012). Systematic functional characterization of putative zinc transport genes and identification of zinc toxicosis phenotypes in Drosophila melanogaster. J Exp Biol 215: 3254-3265. PubMed ID: 22693027

  • Ansgar Büschges Institute of Zoology, Biocenter Cologne, University of Cologne, Cologne, Germany.
    Chockley, A. S., Dinges, G. F., Di Cristina, G., Ratican, S., Bockemuhl, T. and Buschges, A. (2022). Subsets of leg proprioceptors influence leg kinematics but not interleg coordination in Drosophila melanogaster walking. J Exp Biol 225(20). PubMed ID: 36268799

    Dinges, G. F., Bockemuhl, T., Iacoviello, F., Shearing, P. R., Buschges, A. and Blanke, A. (2022). Ultra high-resolution biomechanics suggest that substructures within insect mechanosensors decisively affect their sensitivity. J R Soc Interface 19(190): 20220102. PubMed ID: 35506211

    Liessem, S., Kowatschew, D., Dippel, S., Blanke, A., Korsching, S., Guschlbauer, C., Hooper, S. L., Predel, R. and Buschges, A. (2021). Neuromodulation Can Be Simple: Myoinhibitory Peptide, Contained in Dedicated Regulatory Pathways, Is the Only Neurally-Mediated Peptide Modulator of Stick Insect Leg Muscle. J Neurosci 41(13): 2911-2929. PubMed ID: 33531417

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

  • Ana Maria Busturia Centro de Biologia Molecular 'Severo Ochoa', Madrid
    Solorzano, J., Carrillo-de Santa Pau, E., Laguna, T. and Busturia, A. (2022). A genome-wide computational approach to define microRNA-Polycomb/trithorax gene regulatory circuits in Drosophila. Dev Biol 495: 63-75. PubMed ID: 36596335

    Simoes da Silva, C. J., Sospedra, I., Aparicio, R. and Busturia, A. (2019). The microRNA-306/abrupt regulatory axis controls wing and haltere growth in Drosophila. Mech Dev: 103555. PubMed ID: 31112748

    Simoes da Silva, C. J., Fereres, S., Simon, R. and Busturia, A. (2017). Drosophila SCE/dRING E3-ligase inhibits apoptosis in a Dp53 dependent manner. Dev Biol 429(1): 81-91. PubMed ID: 28712876

    Simon, R., Aparicio, R., Housden, B. E., Bray, S. and Busturia, A. (2014). Drosophila p53 controls Notch expression and balances apoptosis and proliferation. Apoptosis. PubMed ID: 24858703

    Fereres, S., Simon, R. and Busturia, A. (2013). A novel dRYBP-SCF complex functions to inhibit apoptosis in Drosophila. Apoptosis. PubMed ID: 23979704

    Aparicio, R., Neyen, C., Lemaitre, B., Busturia, A. (2013) dRYBP Contributes to the Negative Regulation of the Drosophila Imd Pathway. PLoS One 8: e62052. PubMed ID: 23596533

    Rodriguez-Jato, S., Busturia, A. and Herr, W. (2011). Drosophila melanogaster dHCF interacts with both PcG and TrxG epigenetic regulators. PLoS One 6: e27479. PubMed ID: 22174740

  • Michael Buszczak University of Texas, Southwestern Medical Center, Dallas
    McCarthy, A., Sarkar, K., Martin, E. T., Upadhyay, M., Jang, S., Williams, N. D., Forni, P. E., Buszczak, M. and Rangan, P. (2022). Msl3 promotes germline stem cell differentiation in female Drosophila. Development 149(1). PubMed ID: 34878097

    Jang, S., Lee, J., Mathews, J., Ruess, H., Williford, A. O., Rangan, P., Betran, E. and Buszczak, M. (2021). The Drosophila ribosome protein S5 paralog RpS5b promotes germ cell and follicle cell differentiation during oogenesis. Development 148(19). PubMed ID: 34495316

    Palacios, V., Kimble, G. C., Tootle, T. L. and Buszczak, M. (2021). Importin-9 regulates chromosome segregation and packaging in Drosophila germ cells. J Cell Sci. PubMed ID: 33632744

    Shalaby, N. A., Pinzon, J. H., Narayanan, A. S., Jin, E. J., Ritz, M. P., Dove, R. J., Wolfenberg, H., Rodan, A. R., Buszczak, M. and Rothenfluh, A. (2018). JmjC domain proteins modulate circadian behaviors and sleep in Drosophila. Sci Rep 8(1): 815. PubMed ID: 29339751

    Shalaby, N. A., Sayed, R., Zhang, Q., Scoggin, S., Eliazer, S., Rothenfluh, A. and Buszczak, M. (2017). Systematic discovery of genetic modulation by Jumonji histone demethylases in Drosophila. Sci Rep 7(1): 5240. PubMed ID: 28701701

    Chaturvedi, D., Inaba, M., Scoggin, S. and Buszczak, M. (2016). Drosophila CG2469 encodes a homolog of human CTR9 and is essential for development. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27678520

    Mottier-Pavie, V. I., Palacios, V., Eliazer, S., Scoggin, S. and Buszczak, M. (2016). The Wnt pathway limits BMP signaling outside of the germline stem cell niche in Drosophila ovaries. Dev Biol [Epub ahead of print]. PubMed ID: 27364467

    Carreira-Rosario, A., Bhargava, V., Hillebrand, J., Kollipara, R. K., Ramaswami, M. and Buszczak, M. (2016). Repression of Pumilio protein expression by Rbfox1 promotes germ cell differentiation. Dev Cell 36: 562-571. PubMed ID: 26954550

    Inaba, M., Buszczak, M. and Yamashita, Y.M. (2015). Nanotubes mediate niche-stem-cell signalling in the Drosophila testis. Nature [Epub ahead of print]. PubMed ID: 26131929

    Eliazer, S., Palacios, V., Wang, Z., Kollipara, R. K., Kittler, R. and Buszczak, M. (2014). Lsd1 restricts the number of germline stem cells by regulating multiple targets in escort cells. PLoS Genet 10: e1004200. PubMed ID: 24625679

    Zhang, Q., Shalaby, N. A. and Buszczak, M. (2014). Changes in rRNA transcription influence proliferation and cell fate within a stem cell lineage. Science 343: 298-301. PubMed ID: 24436420

    Carreira-Rosario, A., Scoggin, S., Shalaby, N. A., Williams, N. D., Hiesinger, P. R. and Buszczak, M. (2013). Recombineering homologous recombination constructs in Drosophila. J Vis Exp. PubMed ID: 23893070

  • Laura Buttitta Department of Molecular, Cellular and Developmental Biology, University of Michigan
    Nandakumar, S., Grushko, O. and Buttitta, L. A. (2020). Polyploidy in the adult Drosophila brain. Elife 9. PubMed ID: 32840209

    Ma, Y., McKay, D. J. and Buttitta, L. (2019). Changes in chromatin accessibility ensure robust cell cycle exit in terminally differentiated cells. PLoS Biol 17(9): e3000378. PubMed ID: 31479438

    Ma, Y. and Buttitta, L. (2017). Chromatin organization changes during the establishment and maintenance of the postmitotic state. Epigenetics Chromatin 10(1): 53. PubMed ID: 29126440

    Guo, Y., Flegel, K., Kumar, J., McKay, D.J. and Buttitta, L.A. (2016). Ecdysone signaling induces two phases of cell cycle exit in Drosophila cells. Biol Open [Epub ahead of print]. PubMed ID: 27737823

    Flegel, K., Grushko, O., Bolin, K., Griggs, E. and Buttitta, L. (2016). The role of the histone modifying and exchange complex NuA4 in cell cycle progression in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 27184390

    Bolin, K., Rachmaninoff, N., Moncada, K., Pula, K., Kennell, J. and Buttitta, L. (2016). miR-8 modulates cytoskeletal regulators to influence cell survival and epithelial organization in Drosophila wings. Dev Biol [Epub ahead of print]. PubMed ID: 26902111

    Sun, D. and Buttitta, L. (2015). Protein phosphatase 2A promotes the transition to G0 during terminal differentiation in Drosophila. Development [Epub ahead of print]. PubMed ID: 26253406

    O'Keefe, D. D., Thomas, S., Edgar, B. A. and Buttitta, L. (2014). Temporal regulation of Dpp signaling output in the Drosophila wing. Dev Dyn. PubMed ID: 24591046

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

    O'Keefe, D. D., Thomas, S., Edgar, B. A. and Buttitta, L. (2014). Temporal regulation of Dpp signaling output in the Drosophila wing. Dev Dyn. PubMed ID: 24591046

  • Maria Bykhovskaia Neurology, Wayne State University, Detroit
    Astacio, H., Vasin, A. and Bykhovskaia, M. (2022). Stochastic Properties of Spontaneous Synaptic Transmission at Individual Active Zones. J Neurosci 42(6): 1001-1019. PubMed ID: 34969867

    Brady, J., Vasin, A. and Bykhovskaia, M. (2021). The Accessory Helix of Complexin Stabilizes a Partially Unzippered State of the SNARE Complex and Mediates the Complexin Clamping Function In Vivo. eNeuro 8(2). PubMed ID: 33692090

    Vasin, A., Sabeva, N., Torres, C., Phan, S., Bushong, E. A., Ellisman, M. H. and Bykhovskaia, M. (2019). Two Pathways for the Activity-Dependent Growth and Differentiation of Synaptic Boutons in Drosophila. eNeuro 6(4). PubMed ID: 31387877


  • Return to The Interactive Fly