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


  • Fei Ma Nanjing Normal University, China
    Zhou, H., Li, S., Wu, S., Jin, P. and Ma, F. (2021). LncRNA-CR11538 Decoys Dif/Dorsal to Reduce Antimicrobial Peptide Products for Restoring Drosophila Toll Immunity Homeostasis. Int J Mol Sci 22(18). PubMed ID: 34576280

    Li, R., Yao, X., Zhou, H., Jin, P. and Ma, F. (2021). The Drosophila miR-959-962 Cluster Members Repress Toll Signaling to Regulate Antibacterial Defense during Bacterial Infection. Int J Mol Sci 22(2). PubMed ID: 33477373

    Li, R., Zhou, H., Jia, C., Jin, P. and Ma, F. (2020). Drosophila Myc restores immune homeostasis of Imd pathway via activating miR-277 to inhibit imd/Tab2. PLoS Genet 16(8): e1008989. PubMed ID: 32810129

    Li, R., Huang, Y., Zhang, Q., Zhou, H., Jin, P. and Ma, F. (2019). The miR-317 functions as a negative regulator of Toll immune response and influences Drosophila survival. Dev Comp Immunol 95: 19-27. PubMed ID: 30708026

    Li, S., Xu, J., Sun, L., Li, R., Jin, P. and Ma, F. (2017). Drosophila miR-964 modulates Toll signaling pathway in response to bacterial infection. Dev Comp Immunol 77: 252-258. PubMed ID: 28823799

    Li, S., Shen, L., Sun, L., Xu, J., Jin, P., Chen, L. and Ma, F. (2017). Small RNA-Seq analysis reveals microRNA-regulation of the Imd pathway during Escherichia coli infection in Drosophila. Dev Comp Immunol [Epub ahead of print]. PubMed ID: 28069431

    Li, S., Li, Y., Shen, L., Jin, P., Chen, L. and Ma, F. (2017). miR-958 inhibits Toll signaling and Drosomycin expression via direct targeting of Toll and Dif in Drosophila melanogaster. Am J Physiol Cell Physiol 312(2): C103-C110. PubMed ID: 27974298

    Li, Y., Li, S., Li, R., Xu, J., Jin, P., Chen, L. and Ma, F. (2017). Genome-wide miRNA screening reveals miR-310 family members negatively regulate the immune response in Drosophila melanogaster via co-targeting Drosomycin. Dev Comp Immunol 68: 34-45. PubMed ID: 27871832

    Li, Y., Li, S., Jin, P., Chen, L. and Ma, F. (2017). miR-11 regulates pupal size of Drosophila melanogaster via directly targeting Ras85D. Am J Physiol Cell Physiol 312(1): C71-C82. PubMed ID: 27733364

  • Jun Ma Cincinnati Childrens, Department of Pediatrics
    Liu, J., Xiao, Y., Zhang, T. and Ma, J. (2016). Time to move on: modeling transcription dynamics during an embryonic transition away from maternal control. Fly (Austin): [Epub ahead of print]. PubMed ID: 27172244

    Wu, H., Manu, Jiao, R. and Ma, J. (2015). Temporal and spatial dynamics of scaling-specific features of a gene regulatory network in Drosophila. Nat Commun 6: 10031. PubMed ID: 26644070

    Liu, J. and Ma, J. (2015). Modulation of temporal dynamics of gene transcription by activator potency in the Drosophila embryo. Development [Epub ahead of print]. PubMed ID: 26395487

    Cheung, D. and Ma, J. (2015). Probing the impact of temperature on molecular events in a developmental system. Sci Rep 5: 13124. PubMed ID: 26286011

    He, F., Wei, C., Wu, H., Cheung, D., Jiao, R. and Ma, J. (2015). Fundamental origins and limits for scaling a maternal morphogen gradient. Nat Commun 6: 6679. PubMed ID: 25809405

    Liu, J. and Ma, J. (2013). Dampened regulates the activating potency of Bicoid and the embryonic patterning outcome in Drosophila. Nat Commun 4: 2968. PubMed ID: 24336107

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

    Liu, J. and Ma, J. (2013). Uncovering a dynamic feature of the transcriptional regulatory network for anterior-posterior patterning in the Drosophila embryo. PLoS One 8: e62641. PubMed ID: 23646132

    He, F., Ma, J. (2013) A spatial point pattern analysis in Drosophila blastoderm embryos evaluating the potential inheritance of transcriptional States. PLoS One 8: e60876. PubMed ID: 23593336

  • David MacAlpine Duke University Medical Center
    Li, Y., Armstrong, R. L., Duronio, R. J. and MacAlpine, D. M. (2016). Methylation of histone H4 lysine 20 by PR-Set7 ensures the integrity of late replicating sequence domains in Drosophila. Nucleic Acids Res. PubMed ID: 27131378

    Kim, J. C., Nordman, J., Xie, F., Kashevsky, H., Eng, T., Li, S., MacAlpine, D. M. and Orr-Weaver, T. L. (2011). Integrative analysis of gene amplification in Drosophila follicle cells: parameters of origin activation and repression. Genes Dev 25: 1384-1398. PubMed ID: 21724831

    Negre, N., et al. (2011). A cis-regulatory map of the Drosophila genome. Nature 471: 527-531. PubMed ID: 21430782

    Kharchenko, P. V., et al. (2011). Comprehensive analysis of the chromatin landscape in Drosophila melanogaster. Nature 471: 480-485. PubMed ID: 21179089

  • Paul Macdonald Section of Molecular Cell and Developmental Biology, University of Texas at Austin
    Kenny, A., Morgan, M. B., Mohr, S. and Macdonald, P. M. (2021). Knock down analysis reveals critical phases for specific oskar noncoding RNA functions during Drosophila oogenesis. G3 (Bethesda). PubMed ID: 34586387

    Mohr, S., Kenny, A., Lam, S. T. Y., Morgan, M. B., Smibert, C. A., Lipshitz, H. D. and Macdonald, P. M. (2021). Opposing roles for Egalitarian and Staufen in transport, anchoring and localization of oskar mRNA in the Drosophila oocyte. PLoS Genet 17(4): e1009500. PubMed ID: 33798193

    Kenny, A., Morgan, M. B. and Macdonald, P. M. (2021). Different roles for the adjoining and structurally similar A-rich and poly(A) domains of oskar mRNA: Only the A-rich domain is required for oskar noncoding RNA function, which includes MTOC positioning. Dev Biol 476: 117-127. PubMed ID: 33798537

    Everman, E. R., Cloud-Richardson, K. M. and Macdonald, S. J. (2021). Characterizing the genetic basis of copper toxicity in Drosophila reveals a complex pattern of allelic, regulatory, and behavioral variation. Genetics 217(1): 1-20. PubMed ID: 33683361

    Everman, E. R., McNeil, C. L., Hackett, J. L., Bain, C. L. and Macdonald, S. J. (2019). Dissection of Complex, Fitness-Related Traits in Multiple Drosophila Mapping Populations Offers Insight into the Genetic Control of Stress Resistance. Genetics 211(4): 1449-1467. PubMed ID: 30760490

    Ryu, Y. H., Kenny, A., Gim, Y., Snee, M. and Macdonald, P. M. (2017). Multiple cis-acting signals, some weak by necessity, collectively direct robust transport of oskar mRNA to the oocyte. J Cell Sci 130(18): 3060-3071. PubMed ID: 28760927

    Highfill, C. A., Tran, J. H., Nguyen, S. K. T., Moldenhauer, T. R., Wang, X. and Macdonald, S. J. (2017). Naturally-segregating variation at Ugt86Dd contributes to nicotine resistance in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 28743761

    Ryu, Y. H. and Macdonald, P. M. (2015). RNA sequences required for the noncoding function of oskar RNA also mediate regulation of Oskar protein expression by Bicoid Stability Factor. Dev Biol. PubMed ID: 26433064

    Jones, J. and Macdonald, P.M. (2015). Neurl4 contributes to germ cell formation and integrity in Drosophila. Biol Open [Epub ahead of print]. PubMed ID: 26116656

    Kanke, M. and Macdonald, P. M. (2015). Translational activation of Oskar mRNA: Reevaluation of the role and importance of a 5' regulatory element. PLoS One 10: e0125849. PubMed ID: 25938537

    Kanke, M., Jambor, H., Reich, J., Marches, B., Gstir, R., Ryu, Y.H., Ephrussi, A. and Macdonald, P.M. (2015). oskar RNA plays multiple noncoding roles to support oogenesis and maintain integrity of the germline/soma distinction. RNA [Epub ahead of print]. PubMed ID: 25862242

  • Stuart Macdonald Department of Molecular Biosciences, University of Kansas, Lawrence
    Smith, B. R. and Macdonald, S. J. (2020). Dissecting the Genetic Basis of Variation in Drosophila Sleep Using a Multiparental QTL Mapping Resource. Genes (Basel) 11(3). PubMed ID: 32168738

    Chakraborty, M., Emerson, J. J., Macdonald, S. J. and Long, A. D. (2019). Structural variants exhibit widespread allelic heterogeneity and shape variation in complex traits. Nat Commun 10(1): 4872. PubMed ID: 31653862

    Everman, E. R., McNeil, C. L., Hackett, J. L., Bain, C. L. and Macdonald, S. J. (2019). Dissection of complex, fitness-related traits in multiple Drosophila mapping populations offers Insight into the genetic control of stress resistance. Genetics. PubMed ID: 30760490

    Hackett, J. L., Wang, X., Smith, B. R. and Macdonald, S. J. (2016). Mapping QTL Contributing to Variation in Posterior Lobe Morphology between Strains of Drosophila melanogaster. PLoS One 11: e0162573. PubMed ID: 27606594

    Cloud-Richardson, K. M., Smith, B. R. and Macdonald, S. J. (2016). Genetic dissection of intraspecific variation in a male-specific sexual trait in Drosophila melanogaster. Heredity (Edinb) [Epub ahead of print]. PubMed ID: 27530909

    Marriage, T. N., King, E. G., Long, A. D. and Macdonald, S. J. (2014). Fine-mapping nicotine resistance Loci in Drosophila using a multiparent advanced generation inter-cross population. Genetics 198: 45-57. PubMed ID: 25236448

    King, E. G., Sanderson, B. J., McNeil, C. L., Long, A. D. and Macdonald, S. J. (2014). Genetic Dissection of the Drosophila melanogaster Female Head Transcriptome Reveals Widespread Allelic Heterogeneity. PLoS Genet 10: e1004322. PubMed ID: 24810915

    Cridland, J. M., Macdonald, S. J., Long, A. D. and Thornton, K. R. (2013). Abundance and distribution of transposable elements in two Drosophila QTL mapping resources. Mol Biol Evol 30: 2311-2327. PubMed ID: 23883524

  • Trudy Mackay North Caroline State University, Raleigh
    Huggett, S. B., Hatfield, J. S., Walters, J. D., McGeary, J. E., Welsh, J. W., Mackay, T. F. C., Anholt, R. R. H. and Palmer, R. H. C. (2021). Ibrutinib as a potential therapeutic for cocaine use disorder. Transl Psychiatry 11(1): 623. PubMed ID: 34880215

    Ozsoy, E. D., Yilmaz, M., Patlar, B., Emecen, G., Durmaz, E., Magwire, M. M., Zhou, S., Huang, W., Anholt, R. R. H. and Mackay, T. F. C. (2021). Epistasis for head morphology in Drosophila melanogaster. G3 (Bethesda) 11(10). PubMed ID: 34568933

    Nazario-Yepiz, N. O., Fernandez Sobaberas, J., Lyman, R., Campbell, M. R., Shankar, V., Anholt, R. R. H. and Mackay, T. F. C. (2021). Physiological and metabolomic consequences of reduced expression of the Drosophila brummer triglyceride Lipase. PLoS One 16(9): e0255198. PubMed ID: 34547020

    Mokashi, S. S., Shankar, V., MacPherson, R. A., Hannah, R. C., Mackay, T. F. C. and Anholt, R. R. H. (2021). Developmental Alcohol Exposure in Drosophila: Effects on Adult Phenotypes and Gene Expression in the Brain. Front Psychiatry 12: 699033. PubMed ID: 34366927

    Tallo, C. A., Duncan, L. H., Yamamoto, A. H., Slaydon, J. D., Arya, G. H., Turlapati, L., Mackay, T. F. C. and Carbone, M. A. (2021). Heat shock proteins and small nucleolar RNAs are dysregulated in a Drosophila model for feline hypertrophic cardiomyopathy. G3 (Bethesda) 11(1). PubMed ID: 33561224

    Johnstun, J. A., Shankar, V., Mokashi, S. S., Sunkara, L. T., Ihearahu, U. E., Lyman, R. L., Mackay, T. F. C. and Anholt, R. R. H. (2021). Functional Diversification, Redundancy and Epistasis among Paralogs of the Drosophila melanogaster Obp50a-d Gene Cluster. Mol Biol Evol. PubMed ID: 33560417

    Morgante, F., Huang, W., Sørensen, P., Maltecca, C. and Mackay, T. F. C. (2020). Leveraging multiple layers of data to predict Drosophila complex traits. G3 (Bethesda). PubMed ID: 33106232

    Huang, W., Carbone, M. A., Lyman, R. F., Anholt, R. R. H. and Mackay, T. F. C. (2020). Genotype by environment interaction for gene expression in Drosophila melanogaster. Nat Commun 11(1): 5451. PubMed ID: 33116142

    Yanagawa, A., Huang, W., Yamamoto, A., Wada-Katsumata, A., Schal, C. and Mackay, T. F. C. (2020). Genetic Basis of Natural Variation in Spontaneous Grooming in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32727922

    Huang, W., Campbell, T., Carbone, M. A., Jones, W. E., Unselt, D., Anholt, R. R. H. and Mackay, T. F. C. (2020). Context-dependent genetic architecture of Drosophila life span. PLoS Biol 18(3): e3000645. PubMed ID: 32134916

    Everett, L. J., Huang, W., Zhou, S., Carbone, M. A., Lyman, R. F., Arya, G. H., Geisz, M. S., Ma, J., Morgante, F., St Armour, G., Turlapati, L., Anholt, R. R. H. and Mackay, T. F. C. (2020). Gene expression networks in the Drosophila Genetic Reference Panel. Genome Res. PubMed ID: 32144088

    Parker, G. A., Kohn, N., Spirina, A., McMillen, A., Huang, W. and Mackay, T. F. C. (2020). Genetic Basis of Increased Lifespan and Postponed Senescence in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 31969430

  • Greg Macleod Florida Atlantic University
    Justs, K. A., Lu, Z., Chouhan, A. K., Borycz, J. A., Lu, Z., Meinertzhagen, I. A. and Macleod, G. T. (2021). Presynaptic Mitochondrial Volume and Packing Density Scale with Presynaptic Power Demand. J Neurosci. PubMed ID: 34907026

    Feghhi, T., Hernandez, R. X., Stawarski, M., Thomas, C. I., Kamasawa, N., Lau, A. W. C. and Macleod, G. T. (2021). Computational modeling predicts ephemeral acidic microdomains followed by prolonged alkalinization in the glutamatergic synaptic cleft. Biophys J. PubMed ID: 34774503

    Stawarski, M., Hernandez, R. X., Feghhi, T., Borycz, J. A., Lu, Z., Agarwal, A., Reihl, K., Tavora, R., Lau, A. W. C., Meinertzhagen, I. A., Renden, R. and Macleod, G. T. (2020). Neuronal glutamatergic synaptic clefts alkalinize rather than acidify during neurotransmission. J Neurosci. PubMed ID: 31964719

    Lu, Z., Chouhan, A. K., Borycz, J. A., Lu, Z., Rossano, A. J., Brain, K. L., Zhou, Y., Meinertzhagen, I. A. and Macleod, G. T. (2016). High-probability neurotransmitter release sites represent an energy-efficient design. Curr Biol 26: 2562-2571. PubMed ID: 27593375

    Rossano, A. J., Kato, A., Minard, K. I., Romero, M. F. and Macleod, G. T. (2016). Na+ /H+ -exchange via the Drosophila vesicular glutamate transporter (DVGLUT) mediates activity-induced acid efflux from presynaptic terminals. J Physiol [Epub ahead of print]. PubMed ID: 27641622

    Ivannikov, M. V. and Macleod, G. T. (2013). Mitochondrial free ca(2+) levels and their effects on energy metabolism in Drosophila motor nerve terminals. Biophys J 104: 2353-2361. PubMed ID: 23746507

    Rossano, A. J., Chouhan, A. K. and Macleod, G. T. (2013). Genetically-Encoded pH-Indicators (GEpHIs) Reveal Activity-Dependent Cytosolic Acidification of Drosophila Motor Nerve Termini in vivo. J Physiol. PubMed ID: 23401611

    Macleod, G. T. (2012). Forward-filling of dextran-conjugated indicators for calcium imaging at the Drosophila larval neuromuscular junction. Cold Spring Harb Protoc 2012: 791-796. PubMed ID: 22753611

  • Heath MacMillan Department of Biology Carleton University, Ottawa, ON
    Cheslock, A., Andersen, M. K. and MacMillan, H. A. (2021). Thermal acclimation alters Na(+)/K(+)-ATPase activity in a tissue-specific manner in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol: 110934. PubMed ID: 33684554

    El-Saadi, M. I., Ritchie, M. W., Davis, H. E. and MacMillan, H. A. (2020). Warm periods in repeated cold stresses protect Drosophila against ionoregulatory collapse, chilling injury, and reproductive deficits. J Insect Physiol 123: 104055. PubMed ID: 32380094

    Livingston, D. B. H., Patel, H., Donini, A. and MacMillan, H. A. (2020). Active transport of brilliant blue FCF across the Drosophila midgut and Malpighian tubule epithelia. Comp Biochem Physiol A Mol Integr Physiol 239: 110588. PubMed ID: 31648063

    MacMillan, H. A., Nazal, B., Wali, S., Yerushalmi, G. Y., Misyura, L., Donini, A. and Paluzzi, J. P. (2018). Anti-diuretic activity of a CAPA neuropeptide can compromise Drosophila chill tolerance. J Exp Biol 221(Pt 19). PubMed ID: 30104306

    Yerushalmi, G. Y., Misyura, L., MacMillan, H. A. and Donini, A. (2018). Functional plasticity of the gut and the Malpighian tubules underlies cold acclimation and mitigates cold-induced hyperkalemia in Drosophila melanogaster. J Exp Biol 221(Pt 6). PubMed ID: 29367271

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

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

    Ishibashi, T., Hatori, R., Maeda, R., Nakamura, M., Taguchi, T., Matsuyama, Y. and Matsuno, K. (2019). E and ID proteins regulate cell chirality and left-right asymmetric development in Drosophila. Genes Cells 24(3): 214-230. PubMed ID: 30624823

    Prince, E., Kroeger, B., Gligorov, D., Wilson, C., Eaton, S., Karch, F., Brankatschk, M. and Maeda, R. K. (2019). Rab-mediated trafficking in the secondary cells of Drosophila male accessory glands and its role in fecundity. Traffic 20(2): 137-151. PubMed ID: 30426623

    Maharjan, M., Maeda, R. K., Karch, F. and Hart, C. M. (2018). Using a phiC31 "Disintegrase" to make new attP sites in the Drosophila genome at locations showing chromosomal position effects. PLoS One 13(10): e0205538. PubMed ID: 30296303

    Fedotova, A., Aoki, T., Rossier, M., Mishra, R. K., Clendinen, C., Kyrchanova, O., Wolle, D., Bonchuk, A., Maeda, R. K., Mutero, A., Cleard, F., Mogila, V., Karch, F., Georgiev, P. and Schedl, P. (2018). The BEN Domain Protein Insensitive Binds to the Fab-7 Chromatin Boundary To Establish Proper Segmental Identity in Drosophila. Genetics 210(2): 573-585. PubMed ID: 30082280

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

  • Keith Maggert Department of Biology, Texas A & M, College Station, TX
    Bughio, F. J. and Maggert, K. A. (2022). Live analysis of position-effect variegation in Drosophila reveals different modes of action for HP1a and Su(var)3-9. Proc Natl Acad Sci U S A 119(25): e2118796119. PubMed ID: 35704756

    Bughio, F., Huckell, G. R. and Maggert, K. A. (2019). Monitoring of switches in heterochromatin-induced silencing shows incomplete establishment and developmental instabilities. Proc Natl Acad Sci U S A 116(40): 20043-20053. PubMed ID: 31527269

    Aldrich, J. C. and Maggert, K. A. (2015). Transgenerational inheritance of diet-induced genome rearrangements in Drosophila. PLoS Genet 11: e1005148. PubMed ID: 25885886

    Aldrich, J. C. and Maggert, K. A. (2014). Simple quantitative PCR approach to reveal naturally occurring and mutation-induced repetitive sequence variation on the Drosophila Y chromosome. PLoS One 9: e109906. PubMed ID: 25285439

    Maggert, K. A. (2014). Reduced rDNA copy number does not affect "competitive" chromosome pairing in XYY males of Drosophila melanogaster. G3 (Bethesda) 4: 497-507. PubMed ID: 24449686

    Guerrero, P. A. and Maggert, K. A. (2011). The CCCTC-binding factor (CTCF) of Drosophila contributes to the regulation of the ribosomal DNA and nucleolar stability. PLoS One 6: e16401. PubMed ID: 21283722

  • Anthony Mahowald Department of Molecular Genetics and Cell Biology, The University of Chicago
    Schoenfelder, K. P., Montague, R. A., Paramore, S. V., Lennox, A. L., Mahowald, A. P. and Fox, D. T. (2014). Indispensable pre-mitotic endocycles promote aneuploidy in the Drosophila rectum. Development. PubMed ID: 25142462

    Lerner, D. W., McCoy, D., Isabella, A. J., Mahowald, A. P., Gerlach, G. F., Chaudhry, T. A., Horne-Badovinac, S. (2013) A Rab10-Dependent Mechanism for Polarized Basement Membrane Secretion during Organ Morphogenesis. Dev Cell 24: 159-168. PubMed ID: 23369713

    Srinivasan, S., Mahowald, A. P. and Fuller, M. T. (2012). The receptor tyrosine phosphatase Lar regulates adhesion between Drosophila male germline stem cells and the niche. Development 139: 1381-1390. PubMed ID: 22378638

    Chatterjee, N., Rollins, J., Mahowald, A. P. and Bazinet, C. (2011). Neurotransmitter Transporter-Like: a male germline-specific SLC6 transporter required for Drosophila spermiogenesis. PLoS One 6: e16275. PubMed ID: 21298005

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

  • Helder Maiato Chromosome Instability and Dynamics Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
    Afonso, O., Castellani, C. M., Cheeseman, L. P., Ferreira, J. G., Orr, B., Ferreira, L. T., Chambers, J. J., Morais-de-Sa, E., Maresca, T. J. and Maiato, H. (2019). Spatiotemporal control of mitotic exit during anaphase by an aurora B-Cdk1 crosstalk. Elife 8. PubMed ID: 31424385

    Jana, S. C., Mendonca, S., Machado, P., Werner, S., Rocha, J., Pereira, A., Maiato, H. and Bettencourt-Dias, M. (2018). Differential regulation of transition zone and centriole proteins contributes to ciliary base diversity. Nat Cell Biol 20(8): 928-941. PubMed ID: 30013109

    Drpic, D., Pereira, A. J., Barisic, M., Maresca, T. J. and Maiato, H. (2015). Polar ejection forces promote the conversion from lateral to end-on kinetochore-microtubule attachments on mono-oriented chromosomes. Cell Rep 13: 460-468. PubMed ID: 26456825

    Schweizer, N., Pawar, N., Weiss, M. and Maiato, H. (2015). An organelle-exclusion envelope assists mitosis and underlies distinct molecular crowding in the spindle region. J Cell Biol 210: 695-704. PubMed ID: 26304726

    Afonso, O., Matos, I., Pereira, A. J., Aguiar, P., Lampson, M. A. and Maiato, H. (2014). Feedback control of chromosome separation by a midzone Aurora B gradient. Science 345: 332-336. PubMed ID: 24925910

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

    Conde, C., Osswald, M., Barbosa, J., Moutinho-Santos, T., Pinheiro, D., Guimaraes, S., Matos, I., Maiato, H. and Sunkel, C. E. (2013). Drosophila Polo regulates the spindle assembly checkpoint through Mps1-dependent BubR1 phosphorylation. EMBO J 32: 1761-1777. PubMed ID: 23685359

    Drpic, D., Barisic, M., Pinheiro, D. and Maiato, H. (2013). Selective tracking of template DNA strands after induction of mitosis with unreplicated genomes (MUGs) in Drosophila S2 cells. Chromosome Res 21: 329-337. PubMed ID: 23681663

  • Dieter Maier Institut für Genetik, Universität Hohenheim
    Maier, D. (2020). Membrane-Anchored Hairless Protein Restrains Notch Signaling Activity. Genes (Basel) 11(11). PubMed ID: 33171957

    Wolf, D., Smylla, T. K., Reichmuth, J., Hoffmeister, P., Kober, L., Zimmermann, M., Turkiewicz, A., Borggrefe, T., Nagel, A. C., Oswald, F., Preiss, A. and Maier, D. (2019). Nucleo-cytoplasmic shuttling of Drosophila Hairless/Su(H) heterodimer as a means of regulating Notch dependent transcription. Biochim Biophys Acta Mol Cell Res 1866(10): 1520-1532. PubMed ID: 31326540

    Maier, D. (2019). The evolution of transcriptional repressors in the Notch signaling pathway: a computational analysis. Hereditas 156: 5. PubMed ID: 30679936

    Smylla, T. K., Meier, M., Preiss, A. and Maier, D. (2019). The Notch repressor complex in Drosophila: in vivo analysis of Hairless mutants using overexpression experiments. Dev Genes Evol. PubMed ID: 30612166

    Preiss, A., Nagel, A. C., Praxenthaler, H. and Maier, D. (2018). Complex genetic interactions of novel Suppressor of Hairless alleles deficient in co-repressor binding. PLoS One 13(3): e0193956. PubMed ID: 29509808

    Praxenthaler, H., Nagel, A. C., Schulz, A., Zimmermann, M., Meier, M., Schmid, H., Preiss, A. and Maier, D. (2017). Hairless-binding deficient Suppressor of Hairless alleles reveal Su(H) protein levels are dependent on complex formation with Hairless. PLoS Genet 13(5): e1006774. PubMed ID: 28475577

    Smylla, T.K., Preiss, A. and Maier, D. (2016). In vivo analysis of internal ribosome entry at the Hairless locus by genome engineering in Drosophila. Sci Rep 6: 34881. PubMed ID: 27713501

    Yuan, Z., Praxenthaler, H., Tabaja, N., Torella, R., Preiss, A., Maier, D. and Kovall, R. A. (2016). Structure and Function of the Su(H)-Hairless Repressor Complex, the Major Antagonist of Notch Signaling in Drosophila melanogaster. PLoS Biol 14: e1002509. PubMed ID: 27404588

    Praxenthaler, H., Smylla, T. K., Nagel, A. C., Preiss, A. and Maier, D. (2015). Generation of new hairless alleles by genomic engineering at the Hairless locus in Drosophila melanogaster. PLoS One 10: e0140007. PubMed ID: 26448463

    Panneton, V., Nath, A., Sader, F., Delaunay, N., Pelletier, A., Maier, D., Oh, K. and Hipfner, D. R. (2015). Regulation of Catalytic and Non-catalytic Functions of the Drosophila Ste20 Kinase Slik by Activation Segment Phosphorylation. J Biol Chem 290: 20960-20971. PubMed ID: 26170449

  • Gaby Maimon Laboratory of Integrative Brain Function, Rockefeller University
    Lyu, C., Abbott, L. F. and Maimon, G. (2022). Building an allocentric travelling direction signal via vector computation. Nature 601(7891): 92-97. PubMed ID: 34912112

    Fenk, L. M., Kim, A. J. and Maimon, G. (2021). Suppression of motion vision during course-changing, but not course-stabilizing, navigational turns. Curr Biol. PubMed ID: 34644548

    Green, J., Vijayan, V., Mussells Pires, P., Adachi, A. and Maimon, G. (2019). A neural heading estimate is compared with an internal goal to guide oriented navigation. Nat Neurosci. PubMed ID: 31332373

    Ferris, B. D., Green, J. and Maimon, G. (2018). Abolishment of Spontaneous Flight Turns in Visually Responsive Drosophila. Curr Biol 28(2): 170-180.e175. PubMed ID: 29337081

    Green, J., Adachi, A., Shah, K. K., Hirokawa, J. D., Magani, P. S. and Maimon, G. (2017). A neural circuit architecture for angular integration in Drosophila. Nature 546(7656): 101-106. PubMed ID: 28538731

    Kim, A. J., Fenk, L. M., Lyu, C. and Maimon, G. (2017). Quantitative Predictions Orchestrate Visual Signaling in Drosophila. Cell 168(1-2): 280-294 e212. PubMed ID: 28065412

    Kim, A. J., Fitzgerald, J. K. and Maimon, G. (2015). Cellular evidence for efference copy in Drosophila visuomotor processing. Nat Neurosci 18(9): 1247-1255. PubMed ID: 26237362

  • John Manak Department of Biology, University of Iowa, Iowa City
    Santana, J. F., Parida, M., Long, A., Wankum, J., Lilienthal, A. J., Nukala, K. M. and Manak, J. R. (2020). The Dm-Myb Oncoprotein Contributes to Insulator Function and Stabilizes Repressive H3K27me3 PcG Domains. Cell Rep 30(10): 3218-3228.e3215. PubMed ID: 32160531

    Ehaideb, S. N., Iyengar, A., Ueda, A., Iacobucci, G. J., Cranston, C., Bassuk, A. G., Gubb, D., Axelrod, J. D., Gunawardena, S., Wu, C. F. and Manak, J. R. (2014). prickle modulates microtubule polarity and axonal transport to ameliorate seizures in flies. Proc Natl Acad Sci U S A. PubMed ID: 25024231

    Soshnev, A. A., Baxley, R. M., Manak, J. R., Tan, K. and Geyer, P. K. (2013). The insulator protein Suppressor of Hairy-wing is an essential transcriptional repressor in the Drosophila ovary. Development 140: 3613-3623. PubMed ID: 23884443

    Pezzulo, A. A., Hornick, E. E., Rector, M. V., Estin, M., Reisetter, A. C., Taft, P. J., Butcher, S. C., Carter, A. B., Manak, J. R., Stoltz, D. A. and Zabner, J. (2012). Expression of human paraoxonase 1 decreases superoxide levels and alters bacterial colonization in the gut of Drosophila melanogaster. PLoS One 7: e43777. PubMed ID: 22952763

  • Harmit Malik Fred Hutchinson Cancer Research Center, Seattle
    Schroeder, C. M., Tomlin, S. A., Mejia Natividad, I., Valenzuela, J. R., Young, J. M. and Malik, H. S. (2021). An actin-related protein that is most highly expressed in Drosophila testes is critical for embryonic development. Elife 10. PubMed ID: 34282725

    Kursel, L. E., McConnell, H., de la Cruz, A. F. A. and Malik, H. S. (2021). Gametic specialization of centromeric histone paralogs in Drosophila virilis. Life Sci Alliance 4(7). PubMed ID: 33986021

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

    Kursel, L. E., Welsh, F. C. and Malik, H. S. (2020). Ancient co-retention of paralogs of Cid centromeric histones and Cal1 chaperones in mosquito species. Mol Biol Evol. PubMed ID: 32125433

    Schroeder, C. M., Valenzuela, J. R., Natividad, I. M., Hocky, G. M. and Malik, H. S. (2019). A burst of genetic innovation in Drosophila actin-related proteins for testis-specific function. Mol Biol Evol. PubMed ID: 31697328

    Levin, T. C. and Malik, H. S. (2017). Rapidly evolving Toll-3/4 genes encode male-specific Toll-like receptors in Drosophila. Mol Biol Evol. PubMed ID: 28541576

    Kursel, L. E. and Malik, H. S. (2017). Recurrent gene duplication leads to diverse repertoires of centromeric histones in Drosophila species. Mol Biol Evol [Epub ahead of print]. PubMed ID: 28333217

    Levine, M. T., Vander Wende, H. M., Hsieh, E., Baker, E. P. and Malik, H. S. (2016). Recurrent gene duplication diversifies genome defense repertoire in Drosophila. Mol Biol Evol [Epub ahead of print]. PubMed ID: 26979388

    Phadnis, N., Baker, E. P., Cooper, J. C., Frizzell, K. A., Hsieh, E., de la Cruz, A. F., Shendure, J., Kitzman, J. O. and Malik, H. S. (2015). An essential cell cycle regulation gene causes hybrid inviability in Drosophila. Science 350: 1552-1555. PubMed ID: 26680200

    Levine, M. T., Vander Wende, H. M. and Malik, H. S. (2015). Mitotic fidelity requires transgenerational action of a testis-restricted HP1. Elife 4. PubMed ID: 26151671

  • Lolitika Mandal Indian Institute of Science Education and Research, Mohali
    Ramesh, P., Ghosh, S. and Mandal, L. (2022). Combination of Immunofluorescence and Quantitative Fluorescence In-situ Hybridization for Analysing Differential Gene Expression in the Niche Cells of the Drosophila Lymph Gland. Bio Protoc 12(2): e4290. PubMed ID: 35127980

    Gera, J., Budakoti, P., Suhag, M., Mandal, L. and Mandal, S. (2022). Physiological ROS controls Upd3-dependent modeling of ECM to support cardiac function in Drosophila. Sci Adv 8(7): eabj4991. PubMed ID: 35179958

    Ghosh, S., Ghosh, S. and Mandal, L. (2020). Drosophila metamorphosis involves hemocyte mediated macroendocytosis and efferocytosis. Int J Dev Biol 64(4-5-6): 329-339. PubMed ID: 32658992

    Tiwari, S. K., Toshniwal, A. G., Mandal, S. and Mandal, L. (2020). Fatty acid β-oxidation is required for the differentiation of larval hematopoietic progenitors in Drosophila. Elife 9. PubMed ID: 32530419

    Kaur, H., Sharma, S. K., Mandal, S. and Mandal, L. (2019). Lar maintains the homeostasis of the hematopoietic organ in Drosophila by regulating insulin signaling in the niche. Development 146(24). PubMed ID: 31784462

    Sharma, S. K., Ghosh, S., Geetha, A. R., Mandal, S. and Mandal, L. (2019). Cell adhesion-mediated actomyosin assembly regulates the activity of Cubitus interruptus for hematopoietic progenitor maintenance in Drosophila. Genetics. PubMed ID: 31138608

    Toshniwal, A. G., Gupta, S., Mandal, L. and Mandal, S. (2019). ROS inhibits cell growth by regulating 4EBP and S6K, independent of TOR, during development. Dev Cell 49(3): 473-489. PubMed ID: 31063760

    Aggarwal, P., Gera, J., Ghosh, S., Mandal, L. and Mandal, S. (2017). Non-canonical Decapentaplegic signaling activates Matrix Metalloproteinase 1 to restrict Hedgehog activity and limit ectopic eye differentiation in Drosophila. Genetics. PubMed ID: 28696218

    Aggarwal, P., Gera, J., Mandal, L. and Mandal, S. (2016). The morphogen Decapentaplegic employs a two-tier mechanism to activate target retinal determining genes during ectopic eye formation in Drosophila. Sci Rep 6: 27270. PubMed ID: 27270790

    Ghosh, S., Singh, A., Mandal, S. and Mandal, L. (2015). Active hematopoietic hubs in Drosophila adults generate hemocytes and contribute to immune response. Dev Cell 33: 478-488. PubMed ID: 25959225

  • Mollie Manier Department of Biological Sciences, George Washington University, Washington, DC
    Chebbo, S., Josway, S., Belote, J. M. and Manier, M. K. (2020). A putative novel role for Eip74EF in male reproduction in promoting sperm elongation at the cost of male fecundity. J Exp Zool B Mol Dev Evol. PubMed ID: 32725718

    Zajitschek, S., Zajitschek, F., Josway, S., Al Shabeeb, R., Weiner, H. and Manier, M. K. (2019). Costs and benefits of giant sperm and sperm storage organs in Drosophila melanogaster. J Evol Biol 32(11): 1300-1309. PubMed ID: 31465604

    Lupold, S., Manier, M. K., Puniamoorthy, N., Schoff, C., Starmer, W. T., Luepold, S. H., Belote, J. M. and Pitnick, S. (2016). How sexual selection can drive the evolution of costly sperm ornamentation. Nature 533(7604): 535-538. PubMed ID: 27225128

  • Richard Mann Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center
    Guan, W., Bellemin, S., Bouchet, M., Venkatasubramanian, L., Guillermin, C., Laurençon, A., Kabir, C., Darnas, A., Godin, C., Urdy, S., Mann, R. S. and Enriquez, J. (2022). Post-transcriptional regulation of transcription factor codes in immature neurons drives neuronal diversity. Cell Rep 39(13): 110992. PubMed ID: 35767953

    Feng, S., Rastogi, C., Loker, R., Glassford, W. J., Tomas Rube, H., Bussemaker, H. J. and Mann, R. S. (2022). Transcription factor paralogs orchestrate alternative gene regulatory networks by context-dependent cooperation with multiple cofactors. Nat Commun 13(1): 3808. PubMed ID: 35778382

    Feng, S. and Mann, R. S. (2022). SpyChIP identifies cell type-specific transcription factor occupancy from complex tissues. Proc Natl Acad Sci U S A 119(25): e2122900119. PubMed ID: 35696584 Mishra, A. K., Fritsch, C., Voutev, R., Mann, R. S. and Sprecher, S. G. (2021). Homothorax controls a binary Rhodopsin switch in Drosophila ocelli. PLoS Genet 17(7): e1009460. PubMed ID: 34314427

    Loker, R., Sanner, J. E. and Mann, R. S. (2021). Cell-type-specific Hox regulatory strategies orchestrate tissue identity. Curr Biol. PubMed ID: 34358443

    Feng, S., Lu, S., Grueber, W. B. and Mann, R. S. (2021). Scarless engineering of the Drosophila genome near any site-specific integration site. Genetics 217(3). PubMed ID: 33772309

    Diaz-de-la-Loza, M. D., Loker, R., Mann, R. S. and Thompson, B. J. (2020). Control of tissue morphogenesis by the HOX gene Ultrabithorax. Development 147(5). PubMed ID: 32122911

    Howard, C. E., Chen, C. L., Tabachnik, T., Hormigo, R., Ramdya, P. and Mann, R. S. (2019). Serotonergic Modulation of Walking in Drosophila. Curr Biol 29(24): 4218-4230.e4218. PubMed ID: 31786064

    Delker, R. K., Ranade, V., Loker, R., Voutev, R. and Mann, R. S. (2019). Low affinity binding sites in an activating CRM mediate negative autoregulation of the Drosophila Hox gene Ultrabithorax. PLoS Genet 15(10): e1008444. PubMed ID: 31589607

    Sanchez-Higueras, C., Rastogi, C., Voutev, R., Bussemaker, H. J., Mann, R. S. and Hombria, J. C. (2019). In vivo Hox binding specificity revealed by systematic changes to a single cis regulatory module. Nat Commun 10(1): 3597. PubMed ID: 31399572

    Venkatasubramanian, L., Guo, Z., Xu, S., Tan, L., Xiao, Q., Nagarkar-Jaiswal, S. and Mann, R. S. (2019). Stereotyped terminal axon branching of leg motor neurons mediated by IgSF proteins DIP-alpha and Dpr10. Elife 8. PubMed ID: 30714901

    Newcomb, S., Voutev, R., Jory, A., Delker, R. K., Slattery, M. and Mann, R. S. (2018). cis-regulatory architecture of a short-range EGFR organizing center in the Drosophila melanogaster leg. PLoS Genet 14(8): e1007568. PubMed ID: 30142157

    Enriquez, J., Rio, L. Q., Blazeski, R., Bellemin, S., Godement, P., Mason, C. and Mann, R. S. (2018). Differing strategies despite shared lineages of motor neurons and glia to achieve robust development of an adult neuropil in Drosophila. Neuron 97(3): 538-554.e535. PubMed ID: 29395908

  • Mattias Mannervik Wenner-Gren Institute for Experimental Biology, Stockholms Universitet
    Ing-Simmons, E., Vaid, R., Bing, X. Y., Levine, M., Mannervik, M. and Vaquerizas, J. M. (2021). Independence of chromatin conformation and gene regulation during Drosophila dorsoventral patterning. Nat Genet 53(4): 487-499. PubMed ID: 33795866

    Regadas, I., Dahlberg, O., Vaid, R., Ho, O., Belikov, S., Dixit, G., Deindl, S., Wen, J. and Mannervik, M. (2021). A unique histone 3 lysine 14 chromatin signature underlies tissue-specific gene regulation. Mol Cell. PubMed ID: 33631105

    Sajwan, S. and Mannervik, M. (2019). Gene activation by dCas9-CBP and the SAM system differ in target preference. Sci Rep 9(1): 18104. PubMed ID: 31792240

    Boija, A., Mahat, D. B., Zare, A., Holmqvist, P. H., Philip, P., Meyers, D. J., Cole, P. A., Lis, J. T., Stenberg, P. and Mannervik, M. (2017). CBP regulates recruitment and release of promoter-proximal RNA polymerase II. Mol Cell 68(3): 491-503.e495. PubMed ID: 29056321

    Yeung, K., Boija, A., Karlsson, E., Holmqvist, P. H., Tsatskis, Y., Nisoli, I., Yap, D. B., Lorzadeh, A., Moksa, M., Hirst, M., Aparicio, S., Fanto, M., Stenberg, P., Mannervik, M. and McNeill, H. (2017). Atrophin controls developmental signaling pathways via interactions with Trithorax-like. Elife 6. PubMed ID: 28327288

    Boija, A. and Mannervik, M. (2016). Initiation of diverse epigenetic states during nuclear programming of the Drosophila body plan. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27439862

    Philip, P., Boija, A., Vaid, R., Churcher, A. M., Meyers, D. J., Cole, P. A., Mannervik, M. and Stenberg, P. (2015). CBP binding outside of promoters and enhancers in Drosophila melanogaster. Epigenetics Chromatin 8: 48. PubMed ID: 26604986

    Crona, F., Holmqvist, P.H., Tang, M., Singla, B., Vakifahmetoglu-Norberg, H., Fantur, K. and Mannervik, M. (2015). The Brakeless co-regulator can directly activate and repress transcription in early Drosophila embryos. Dev Biol [Epub ahead of print]. PubMed ID: 26260775

    Boija, A. and Mannervik, M. (2015). A time of change: Dynamics of chromatin and transcriptional regulation during nuclear programming in early Drosophila development. Mol Reprod Dev. PubMed ID: 26099731

    Shir-Shapira, H., Sharabany, J., Filderman, M., Ideses, D., Ovadia-Shochat, A., Mannervik, M. and Juven-Gershon, T. (2015). Structure-Function Analysis of the Drosophila melanogaster Caudal Provides Insights into Core Promoter-Preferential Activation. J Biol Chem. PubMed ID: 26018075

    Dahlberg, O., Shilkova, O., Tang, M., Holmqvist, P. H. and Mannervik, M. (2015). P-TEFb, the Super Elongation Complex and Mediator Regulate a Subset of Non-paused Genes during Early Drosophila Embryo Development. PLoS Genet 11: e1004971. PubMed ID: 25679530

  • Stefan Marciniak Cambridge Institute for Medical Research
    Nichols, R., Pittala, K., Leander, M., Maynard, B., Nikolaou, P. and Marciniak, P. (2021). The myosuppressin structure-activity relationship for cardiac contractility and its receptor interactions support the presence of a ligand-directed signaling pathway in heart. Peptides: 170641. PubMed ID: 34453985

    Malzer, E., Dominicus, C. S., Chambers, J. E., Dickens, J. A., Mookerjee, S. and Marciniak, S. J. (2018). The integrated stress response regulates BMP signalling through effects on translation. BMC Biol 16(1): 34. PubMed ID: 29609607

    Malzer, E., Szajewska-Skuta, M., Dalton, L. E., Thomas, S. E., Hu, N., Skaer, H., Lomas, D. A., Crowther, D. C., Marciniak, S. J. (2013) Coordinate regulation of eIF2alpha phosphorylation by dPPP1R15 and dGCN2 is required during development. J Cell Sci. PubMed ID: 23418347

    Liu, B., Moloney, A., Meehan, S., Morris, K., Thomas, S. E., Serpell, L. C., Hider, R., Marciniak, S. J., Lomas, D. A. and Crowther, D. C. (2011). Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem 286: 4248-4256. PubMed ID: 21147772

    Malzer, E., Daly, M. L., Moloney, A., Sendall, T. J., Thomas, S. E., Ryder, E., Ryoo, H. D., Crowther, D. C., Lomas, D. A. and Marciniak, S. J. (2010). Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in the integrated stress response. J Cell Sci 123: 2892-2900. PubMed ID: 20682638

  • Graeme Mardon Developmental Biology, Baylor College of Medicine
    Yeung, K., Wang, F., Li, Y., Wang, K., Mardon, G. and Chen, R. (2018). Integrative genomic analysis reveals novel regulatory mechanisms of eyeless during Drosophila eye development. Nucleic Acids Res. PubMed ID: 30295802

    Jin, M., Eblimit, A., Pulikkathara, M., Corr, S., Chen, R. and Mardon, G. (2016). Conditional knockout of retinal determination genes in differentiating cells in Drosophila. FEBS J [Epub ahead of print]. PubMed ID: 27257739

    Jin, M. and Mardon, G. (2016). Distinct biochemical activities of Eyes absent during Drosophila eye development. Sci Rep 6: 23228. PubMed ID: 26980695

    Jusiak, B., Wang, F., Karandikar, U. C., Kwak, S. J., Wang, H., Chen, R. and Mardon, G. (2014). Genome-wide DNA binding pattern of the homeodomain transcription factor Sine oculis (So) in the developing eye of. Genom Data 2: 153-155. PubMed ID: 25126519

    Karandikar, U. C., Jin, M., Jusiak, B., Kwak, S., Chen, R. and Mardon, G. (2014). Drosophila eyes absent is required for normal cone and pigment cell development. PLoS One 9: e102143. PubMed ID: 25057928

    Jusiak, B., Karandikar, U. C., Kwak, S. J., Wang, F., Wang, H., Chen, R. and Mardon, G. (2014). Regulation of Drosophila Eye Development by the Transcription Factor Sine oculis. PLoS One 9: e89695. PubMed ID: 24586968

    Atkins, M., Jiang, Y., Sansores-Garcia, L., Jusiak, B., Halder, G. and Mardon, G. (2013). Dynamic rewiring of the Drosophila retinal determination network switches its function from selector to differentiation. PLoS Genet 9: e1003731. PubMed ID: 24009524

    Haase Gilbert, E., Kwak, S. J., Chen, R., Mardon, G. (2013) Drosophila signal peptidase complex member spase12 is required for development and cell differentiation. PLoS One 8: e60908. PubMed ID: 23573290

    Jin, M., Jusiak, B., Bai, Z., Mardon, G. (2013) Eyes absent tyrosine phosphatase activity is not required for Drosophila development or survival. PLoS One 8: e58818. PubMed ID: 23554934

    Jusiak, B., Abulimiti, A., Haelterman, N., Chen, R. and Mardon, G. (2012). MAPK target sites of eyes absent are not required for eye development or survival in Drosophila. PLoS One 7: e50776. PubMed ID: 23251383

  • Daniel Marenda Department of Biology, Drexel University, Philadelphia
    Barrett, M., Fiocca, K., Waddell, E. A., McNair, C., O'Donnell, S. and Marenda, D. R. (2020). Larval mannitol diets increase mortality, prolong development and decrease adult body sizes in fruit flies (Drosophila melanogaster). Biol Open 8(12). PubMed ID: 31822472

    Fiocca, K., Barrett, M., Waddell, E. A., Viveiros, J., McNair, C., O'Donnell, S. and Marenda, D. R. (2019). Mannitol ingestion causes concentration-dependent, sex-biased mortality in adults of the fruit fly (Drosophila melanogaster). PLoS One 14(5): e0213760. PubMed ID: 31150400

    D'Rozario, M., Zhang, T., Waddell, E.A., Zhang, Y., Sahin, C., Sharoni, M., Hu, T., Nayal, M., Kutty, K., Liebl, F., Hu, W. and Marenda, D.R. (2016). Type I bHLH proteins Daughterless and Tcf4 restrict neurite branching and synapse formation by repressing Neurexin in postmitotic neurons. Cell Rep [Epub ahead of print]. PubMed ID: 27050508

    Mhatre, S. D., Satyasi, V., Killen, M., Paddock, B. E., Moir, R. D., Saunders, A. J. and Marenda, D. R. (2014). Altered synapses in a Drosophila model of Alzheimer's disease. Dis Model Mech. PubMed ID: 24487408

    Reza, M. A., Mhatre, S. D., Morrison, J. C., Utreja, S., Saunders, A. J., Breen, D. E. and Marenda, D. R. (2013). Automated analysis of courtship suppression learning and memory in Drosophila melanogaster. Fly (Austin) 7: 105-111. PubMed ID: 23644900

    Distefano, G. M., Gangemi, A. J., Khandelwal, P. J., Saunders, A. J. and Marenda, D. R. (2012). Drosophila lilliputian is required for proneural gene expression in retinal development. Dev Dyn 241: 553-562. PubMed ID: 22275119

  • Thomas Maresca, Biology Department, University of Massachusetts Amherst
    Audett, M. R., Johnson, E. L., McGory, J. M., Barcelos, D. M., Szalai, E. O., Przewloka, M. R. and Maresca, T. J. (2021). The microtubule- and PP1-binding activities of Drosophila melanogaster Spc105 control the kinetics of SAC satisfaction. Mol Biol Cell: mbcE21060307T. PubMed ID: 34705493

    Verma, V. and Maresca, T. J. (2019). Direct observation of branching MT nucleation in living animal cells. J Cell Biol. PubMed ID: 31340987

    Verma, V. and Maresca, T. J. (2019). Microtubule plus-ends act as physical signaling hubs to activate RhoA during cytokinesis. Elife 8. PubMed ID: 30758285

    Ye, A. A., Verma, V. and Maresca, T. J. (2018). NOD is a plus end-directed motor that binds EB1 via a new microtubule tip localization sequence. J Cell Biol. PubMed ID: 29899040

    Ye, A. A., Torabi, J. and Maresca, T. J. (2016). Aurora A Kinase Amplifies a Midzone Phosphorylation Gradient to Promote High-Fidelity Cytokinesis. Biol Bull 231: 61-72. PubMed ID: 27638695

    Cane, S., Ye, A. A., Luks-Morgan, S. J., Maresca, T. J. (2013) Elevated polar ejection forces stabilize kinetochore-microtubule attachments. J Cell Biol 200: 203-218. PubMed ID: 23337118

    Maresca, T. J. (2011). Chromosome segregation: a kinetochore missing link is found. Curr Biol 21: R261-263. PubMed ID: 21481764

    Maresca, T. J. and Salmon, E. D. (2009). Intrakinetochore stretch is associated with changes in kinetochore phosphorylation and spindle assembly checkpoint activity. J Cell Biol 184: 373-381. PubMed ID: 19193623

  • Theresa Markow Cell and Developmental Biology, U. C. San Diego
    Ranz, J. M., González, P. M., Su, R. N., Bedford, S. J., Abreu-Goodger, C. and Markow, T. (2022). Multiscale analysis of the randomization limits of the chromosomal gene organization between Lepidoptera and Diptera. Proc Biol Sci 289(1967): 20212183. PubMed ID: 35042416

    Mateus, P. R., Nazario-Yepiz, N. O., Ibarra-Laclette, E., Ramirez Loustalot Laclette, M. and Markow, T. A. (2018). Developmental and transcriptomal responses to seasonal dietary shifts in the cactophilic Drosophila mojavensis of North America. J Hered. PubMed ID: 30371801

    Martinson, V. G., Carpinteyro-Ponce, J., Moran, N. A. and Markow, T. A. (2017). A distinctive and host-restricted gut microbiota in populations of a cactophilic Drosophila species. Appl Environ Microbiol [Epub ahead of print]. PubMed ID: 28939605

    Castro Vargas, C., Richmond, M. P., Ramirez Loustalot Laclette, M. and Markow, T. A. (2017). Early events in speciation: Cryptic species of Drosophila aldrichi. Ecol Evol 7(12): 4220-4228. PubMed ID: 28649335

    Markow, T. A. (2015). Drosophila reproduction: Molecules meet morphology. Proc Natl Acad Sci U S A 112: 8168-8169. PubMed ID: 26124147

    Markow, T. A. (2015). The secret lives of Drosophila flies. Elife 4. PubMed ID: 26041333

    Haselkorn, T. S., Watts, T. D. and Markow, T. A. (2013). Density dynamics of diverse Spiroplasma strains naturally infecting different species of Drosophila. Fly (Austin) 7. PubMed ID: 23846301

    Matzkin, L. M., Johnson, S., Paight, C., Markow, T. A. (2013) Preadult Parental Diet Affects Offspring Development and Metabolism in Drosophila melanogaster. PLoS One 8: e59530. PubMed ID: 23555695

    Markow, T. A. (2013) Parents without Partners: Drosophila as a Model for Understanding the Mechanisms and Evolution of Parthenogenesis. G3 (Bethesda). PubMed ID: 23550124

    Markow, T. A., Beall, S. and Castrezana, S. (2012). The wild side of life: Drosophila reproduction in nature. Fly (Austin) 6: 98-101. PubMed ID: 22627903

  • Frédéric Marion-Poll Laboratoire Evolution, Génomes, Comportement, Ecologie, UMR, CNRS, Gif sur Yvette
    Yanagawa, A., Couto, A., Sandoz, J. C., Hata, T., Mitra, A., Ali Agha, M. and Marion-Poll, F. (2018). LPS perception through taste-induced reflex in Drosophila melanogaster. J Insect Physiol. PubMed ID: 30528842

    Yanagawa, A., Neyen, C., Lemaitre, B. and Marion-Poll, F. (2017). The gram-negative sensing receptor PGRP-LC contributes to grooming induction in Drosophila. PLoS One 12(11): e0185370. PubMed ID: 29121087

    Yanagawa, A., Chabaud, M. A., Imai, T. and Marion-Poll, F. (2017). Olfactory cues play a significant role in removing fungus from the body surface of Drosophila melanogaster. J Invertebr Pathol. PubMed ID: 29175531

    French, A. S., Sellier, M. J., Moutaz, A. A., Guigue, A., Chabaud, M. A., Reeb, P. D., Mitra, A., Grau, Y., Soustelle, L. and Marion-Poll, F. (2015). Dual mechanism for bitter avoidance in Drosophila. J Neurosci 35: 3990-4004. PubMed ID: 25740527

    Yanagawa, A., Guigue, A. M. and Marion-Poll, F. (2014). Hygienic grooming is induced by contact chemicals in Drosophila melanogaster. Front Behav Neurosci 8: 254. PubMed ID: 25100963

    Devambez, I., Ali Agha, M., Mitri, C., Bockaert, J., Parmentier, M. L., Marion-Poll, F., Grau, Y. and Soustelle, L. (2013). Galphao is required for L-canavanine detection in Drosophila. PLoS One 8: e63484. PubMed ID: 23671680

  • Michael Marr Life Sciences at Brandeis
    Spellberg, M. J. and Marr, M. T. (2015). FOXO regulates RNA interference in Drosophila and protects from RNA virus infection. Proc Natl Acad Sci U S A 112: 14587-14592. PubMed ID: 26553999

    Marr, S. K., Lis, J. T., Treisman, J. E. and Marr, M. T. (2014). The Metazoan-Specific Mediator Subunit 26 (Med26) Is Essential For Viability And Is Found At Both Active Genes And Pericentric Heterochromatin In Drosophila. Mol Cell Biol. PubMed ID: 24820420

    Olson, C. M., Donovan, M. R., Spellberg, M. J. and Marr, M. T. (2013). The insulin receptor cellular IRES confers resistance to eIF4A inhibition. Elife 2: e00542. PubMed ID: 23878722

    Marr, S. K., Pennington, K. L. and Marr, M. T. (2012). Efficient metal-specific transcription activation by Drosophila MTF-1 requires conserved cysteine residues in the carboxy-terminal domain. Biochim Biophys Acta 1819: 902-912. PubMed ID: 22484022

    Khodor, Y. L., Rodriguez, J., Abruzzi, K. C., Tang, C. H., Marr, M. T. and Rosbash, M. (2011). Nascent-seq indicates widespread cotranscriptional pre-mRNA splicing in Drosophila. Genes Dev 25: 2502-2512. PubMed ID: 22156210

  • Larry Marsh Developmental and Cell Biology, University of California Irvine
    Chongtham, A., Bornemann, D. J., Barbaro, B. A., Lukacsovich, T., Agrawal, N., Syed, A., Worthge, S., Purcell, J., Burke, J., Chin, T. M. and Marsh, J. L. (2020). Effects of flanking sequences and cellular context on subcellular behavior and pathology of mutant HTT. Hum Mol Genet. PubMed ID: 31943010

    Barbaro, B. A., Lukacsovich, T., Agrawal, N., Burke, J., Bornemann, D. J., Purcell, J. M., Worthge, S. A., Caricasole, A., Weiss, A., Song, W., Morozova, O. A., Colby, D. W. and Marsh, J. L. (2014). Comparative study of naturally occurring huntingtin fragments in Drosophila points to exon 1 as the most pathogenic species in Huntington's disease. Hum Mol Genet. PubMed ID: 25305076

    Song, W., Smith, M. R., Syed, A., Lukacsovich, T., Barbaro, B. A., Purcell, J., Bornemann, D. J., Burke, J., Marsh, J. L. (2013) Morphometric Analysis of Huntington's Disease Neurodegeneration in Drosophila. Methods Mol Biol 1017: 41-57. PubMed ID: 23719906

    Bodai, L., Pallos, J., Thompson, L. M. and Marsh, J. L. (2012). Pcaf modulates polyglutamine pathology in a Drosophila model of Huntington's disease. Neurodegener Dis 9: 104-106. PubMed ID: 21912091

    Sinadinos, C., Burbidge-King, T., Soh, D., Thompson, L. M., Marsh, J. L., Wyttenbach, A. and Mudher, A. K. (2009). Live axonal transport disruption by mutant huntingtin fragments in Drosophila motor neuron axons. Neurobiol Dis 34: 389-395. PubMed ID: 19268537

  • Adam Martin Lab of Morphogenesis, MIT
    Denk-Lobnig, M., Totz, J. F., Heer, N. C., Dunkel, J. and Martin, A. C. (2021). Combinatorial patterns of graded RhoA activation and uniform F-actin depletion promote tissue curvature. Development 148(11). PubMed ID: 34124762

    Denk-Lobnig, M., Totz, J. F., Heer, N. C., Dunkel, J. and Martin, A. C. (2021). Combinatorial patterns of graded RhoA activation and uniform F-actin depletion promote tissue curvature. Development. PubMed ID: 33983369

    Imran Alsous, J., Romeo, N., Jackson, J. A., Mason, F. M., Dunkel, J. and Martin, A. C. (2021). Dynamics of hydraulic and contractile wave-mediated fluid transport during Drosophila oogenesis. Proc Natl Acad Sci U S A 118(10). PubMed ID: 33658367

    Martin, A. C. (2020). The Physical Mechanisms of Drosophila Gastrulation: Mesoderm and Endoderm Invagination. Genetics 214(3): 543-560. PubMed ID: 32132154

    Ko, C. S., Kalakuntla, P. and Martin, A. C. (2020). Apical constriction reversal upon mitotic entry underlies different morphogenetic outcomes of cell division. Mol Biol Cell: mbcE19120673. PubMed ID: 32129704

    Yevick, H. G., Miller, P. W., Dunkel, J. and Martin, A. C. (2019). Structural redundancy in supracellular actomyosin networks enables robust tissue folding. Dev Cell. PubMed ID: 31353314

    Ko, C. S., Tserunyan, V. and Martin, A. C. (2019). Microtubules promote intercellular contractile force transmission during tissue folding. J Cell Biol. PubMed ID: 31227595

    Chanet, S., Miller, C. J., Vaishnav, E. D., Ermentrout, B., Davidson, L. A. and Martin, A. C. (2017). Actomyosin meshwork mechanosensing enables tissue shape to orient cell force. Nat Commun 8: 15014. PubMed ID: 28504247

    Vasquez, C. G., Heissler, S. M., Billington, N., Sellers, J. R. and Martin, A. C. (2016). Drosophila non-muscle myosin II motor activity determines the rate of tissue folding. Elife 5. PubMed ID: 28035903

    Xie, S., Mason, F. M. and Martin, A. C. (2016). Loss of Galpha12/13 exacerbates apical area dependence of actomyosin contractility. Mol Biol Cell 27: 3526-3536. PubMed ID: 27489340

    Mason, F. M., Xie, S., Vasquez, C. G., Tworoger, M. and Martin, A. C. (2016). RhoA GTPase inhibition organizes contraction during epithelial morphogenesis. J Cell Biol 214: 603-617. PubMed ID: 27551058

    Xie, S., Mason, F. M. and Martin, A. C. (2016). Loss of Galpha12/13 exacerbates apical area-dependence of actomyosin contractility. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27489340

  • David Martin Institute of Evolutionary Biology, Barcelona
    Cruz, J., Martin, D. and Franch-Marro, X. (2020). Egfr Signaling Is a Major Regulator of Ecdysone Biosynthesis in the Drosophila Prothoracic Gland. Curr Biol 30(8): 1547-1554.e1544. PubMed ID: 32220314

    Mansilla, A., Martín, F.A., Martín, D. and Ferrús, A. (2015). Ligand-independent requirements of steroid receptors EcR and USP for cell survival. Cell Death Differ [Epub ahead of print]. PubMed ID: 26250909

    Urena, E., Manjon, C., Franch-Marro, X. and Martin, D. (2014). Transcription factor E93 specifies adult metamorphosis in hemimetabolous and holometabolous insects. Proc Natl Acad Sci U S A. PubMed ID: 24778249

    Takayama, S., Dhahbi, J., Roberts, A., Mao, G., Heo, S. J., Pachter, L., Martin, D. and Boffelli, D. (2014). Genome methylation in D. melanogaster is found at specific short motifs and is independent of DNMT2 activity. Genome Res. PubMed ID: 24558263

    Esslinger, S. M., Schwalb, B., Helfer, S., Michalik, K. M., Witte, H., Maier, K. C., Martin, D., Michalke, B., Tresch, A., Cramer, P. and Forstemann, K. (2013). Drosophila miR-277 controls branched-chain amino acid catabolism and affects lifespan. RNA Biol 10: 1042-1056. PubMed ID: 23669073

    Pankotai, T., Popescu, C., Martin, D., Grau, B., Zsindely, N., Bodai, L., Tora, L., Ferrus, A. and Boros, I. (2010). Genes of the ecdysone biosynthesis pathway are regulated by the dATAC histone acetyltransferase complex in Drosophila. Mol Cell Biol 30: 4254-4266. PubMed ID: 20584983

  • Francisco Martin Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Madrid, Spain
    Jarabo, P., Barredo, C. G., de Pablo, C., Casas-Tinto, S. and Martin, F. A. (2022). Alignment between glioblastoma internal clock and environmental cues ameliorates survival in Drosophila. Commun Biol 5(1): 644. PubMed ID: 35773327

    Jarabo, P., de Pablo, C., Herranz, H., Martin, F. A. and Casas-Tinto, S. (2021). Insulin signaling mediates neurodegeneration in glioma. Life Sci Alliance 4(3). PubMed ID: 33526430

    Barredo, C. G., Gil-Marti, B., Deveci, D., Romero, N. M. and Martin, F. A. (2020). Timing the Juvenile-Adult Neurohormonal Transition: Functions and Evolution. Front Endocrinol (Lausanne) 11: 602285. PubMed ID: 33643219

    Jarabo, P. and Martin, F. A. (2017). Neurogenetics of Drosophila circadian clock: expect the unexpected. J Neurogenet 31(4): 250-265. PubMed ID: 28868955

  • Jean René Martin Imagerie Cérébrale Fonctionnelle et Comportements, Neurobiologie et Developpement, Gif-sur-Yvette, France.
    Soule, S., Mellottee, L., Arab, A., Chen, C. and Martin, J. R. (2020). Jouvence a small nucleolar RNA required in the gut extends lifespan in Drosophila. Nat Commun 11(1): 987. PubMed ID: 32080190

    Lark, A., Kitamoto, T. and Martin, J. R. (2017). Modulation of neuronal activity in the Drosophila mushroom body by DopEcR, a unique dual receptor for ecdysone and dopamine. Biochim Biophys Acta. PubMed ID: 28554773

    Murmu, M. S. and Martin, J. R. (2016). Interaction between cAMP and intracellular Ca-signaling pathways during odor-perception and adaptation in Drosophila. Biochim Biophys Acta 1863: 2156-2174. PubMed ID: 27212269

    Lark, A. R., Kitamoto, T. and Martin, J. R. (2016). In Vivo Functional Brain Imaging Approach Based on Bioluminescent Calcium Indicator GFP-aequorin. J Vis Exp. PubMed ID: 26779599

    Pavot, P., Carbognin, E. and Martin, J. R. (2015). PKA and cAMP/CNG channels independently regulate the cholinergic Ca(2+)-response of Drosophila mushroom body neurons. eNeuro 2. PubMed ID: 26464971

    Minocci, D., Carbognin, E., Murmu, M. S. and Martin, J. R. (2013). In vivo functional calcium imaging of induced or spontaneous activity in the fly brain using a GFP-apoaequorin-based bioluminescent approach. Biochim Biophys Acta 1833: 1632-1640. PubMed ID: 23287020

    Murmu, M. S., Stinnakre, J., Real, E. and Martin, J. R. (2011). Calcium-stores mediate adaptation in axon terminals of olfactory receptor neurons in Drosophila. BMC Neurosci 12: 105. PubMed ID: 22024464

    Inoshita, T., Martin, J. R., Marion-Poll, F. and Ferveur, J. F. (2011). Peripheral, central and behavioral responses to the cuticular pheromone bouquet in Drosophila melanogaster males. PLoS One 6: e19770. PubMed ID: 21625481

  • Paul Martin Department of Biochemistry, University of Bristol
    Weavers, H., Wood, W. and Martin, P. (2019). Injury activates a dynamic cytoprotective network to confer stress resilience and drive repair. Curr Biol 29(22): 3851-3862 PubMed ID: 31668626

    Weavers, H., Franz, A., Wood, W. and Martin, P. (2018). Long-term in vivo tracking of inflammatory cell dynamics within Drosophila pupae. J Vis Exp(136). PubMed ID: 29985351

    Thuma, L., Carter, D., Weavers, H. and Martin, P. (2018). Drosophila immune cells extravasate from vessels to wounds using Tre1 GPCR and Rho signaling. J Cell Biol. PubMed ID: 29941473

    Franz, A., Wood, W. and Martin, P. (2018). Fat body cells are motile and actively migrate to wounds to drive repair and prevent infection. Dev Cell 44(4): 460-470.e463. PubMed ID: 29486196

    Weavers, H., Evans, I. R., Martin, P. and Wood, W. (2016). Corpse Engulfment Generates a Molecular Memory that Primes the Macrophage Inflammatory Response. Cell. PubMed ID: 27212238

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

    Razzell, W., Wood, W. and Martin, P. (2014). Recapitulation of morphogenetic cell shape changes enables wound re-epithelialisation. Development. PubMed ID: 24718989

    Foronda, D., Martin, P. and Sanchez-Herrero, E. (2012). Drosophila Hox and sex-determination genes control segment elimination through EGFR and extramacrochetae activity. PLoS Genet 8: e1002874. PubMed ID:
    Razzell, W., Wood, W. and Martin, P. (2011). Swatting flies: modelling wound healing and inflammation in Drosophila. Dis Model Mech 4: 569-574. PubMed ID:
    21810906

  • Maria Martin-Bermudo Centro Andaluz de Biologia del Desarrollo
    Perez-Moreno, J. J., Santa-Cruz Mateos, C., Martin-Bermudo, M. D. and Estrada, B. (2021). LanB1 Cooperates With Kon-Tiki During Embryonic Muscle Migration in Drosophila. Front Cell Dev Biol 9: 749723. PubMed ID: 35047493

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

    Santa-Cruz Mateos, C., Valencia-Exposito, A., Palacios, I. M. and Martin-Bermudo, M. D. (2020). Integrins regulate epithelial cell shape by controlling the architecture and mechanical properties of basal actomyosin networks. PLoS Genet 16(6): e1008717. PubMed ID: 32479493

    Dai, J., Estrada, B., Jacobs, S., Sanchez-Sanchez, B. J., Tang, J., Ma, M., Magadan-Corpas, P., Pastor-Pareja, J. C. and Martin-Bermudo, M. D. (2018). Dissection of Nidogen function in Drosophila reveals tissue-specific mechanisms of basement membrane assembly. PLoS Genet 14(9): e1007483. PubMed ID: 30260959

    Sanchez-Sanchez, B. J., Urbano, J. M., Comber, K., Dragu, A., Wood, W., Stramer, B. and Martin-Bermudo, M. D. (2017). Drosophila embryonic hemocytes produce laminins to strengthen migratory response. Cell Rep 21(6): 1461-1470. PubMed ID: 29117553

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

    Ng, B. F., Selvaraj, G. K., Mateos, C. S., Grosheva, I., Alvarez-Garcia, I., Martin-Bermudo, M. D. and Palacios, I. M. (2016). Alpha-Spectrin and Integrins act together to regulate actomyosin and columnarization, and to maintain a mono-layered follicular epithelium. Development. PubMed ID: 26952981

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

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

    Gomez-Lamarca, M. J., Cobreros-Reguera, L., Ibanez-Jimenez, B., Palacios, I. M. and Martin-Bermudo, M. D. (2014). Integrins regulate epithelial cell differentiation by modulating Notch activity. J Cell Sci. PubMed ID: 25179603

  • Enrique Martín-Blanco Molecular Biology Institute of Barcelona
    Karkali, K. and Martin-Blanco, E. (2021). Dissection of the Regulatory Elements of the Complex Expression Pattern of Puckered, a Dual-Specificity JNK Phosphatase. Int J Mol Sci 22(22). PubMed ID: 34830088

    Karkali, K., Tiwari, P., Singh, A., Tlili, S., Jorba, I., Navajas, D., Munoz, J. J., Saunders, T. E. and Martin-Blanco, E. (2022). Condensation of the Drosophila nerve cord is oscillatory and depends on coordinated mechanical interactions. Dev Cell 57(7): 867-882.e865. PubMed ID: 35413236

    Prat-Rojo, C., Pouille, P. A., Buceta, J. and Martin-Blanco, E. (2019). Mechanical coordination is sufficient to promote tissue replacement during metamorphosis in Drosophila. Embo j: e103594. PubMed ID: 31858605

    Mangione, F. and Martin-Blanco, E. (2018). The Dachsous/Fat/Four-Jointed pathway directs the uniform axial orientation of epithelial cells in the Drosophila abdomen. Cell Rep 25(10): 2836-2850.e2834. PubMed ID: 30517870

    Alvarez-Fernandez, C., Tamirisa, S., Prada, F., Chernomoretz, A., Podhajcer, O., Blanco, E. and Martin-Blanco, E. (2015). Identification and functional analysis of healing regulators in Drosophila. PLoS Genet 11: e1004965. PubMed ID: 25647511

    Rebollo, E., Karkali, K., Mangione, F. and Martin-Blanco, E. (2014). Live imaging in Drosophila: The optical and genetic toolkits. Methods. PubMed ID: 24814031

    Pereira, A. M., Tudor, C., Kanger, J. S., Subramaniam, V. and Martin-Blanco, E. (2011). Integrin-dependent activation of the JNK signaling pathway by mechanical stress. PLoS One 6: e26182. PubMed ID: 22180774

    Ninov, N., Menezes-Cabral, S., Prat-Rojo, C., Manjon, C., Weiss, A., Pyrowolakis, G., Affolter, M. and Martin-Blanco, E. (2010). Dpp signaling directs cell motility and invasiveness during epithelial morphogenesis. Curr Biol 20: 513-520. PubMed ID: 20226662

    Ninov, N. and Martin-Blanco, E. (2009). Changing gears in the cell cycle: histoblasts and beyond. Fly (Austin) 3: 286-289. PubMed ID: 19934653

  • Julian Martinez-Agosto Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles
    Ferguson, G. B. and Martinez-Agosto, J. A. (2017). The TEAD family transcription factor Scalloped regulates blood progenitor maintenance and proliferation in Drosophila through PDGF/VEGFR receptor (Pvr) signaling. Dev Biol. PubMed ID: 28322737

    Ferguson, G. B. and Martinez-Agosto, J. A. (2014). Kicking it up a Notch for the best in show: Scalloped leads Yorkie into the haematopoietic arena. Fly (Austin) 8: 206-217. PubMed ID: 26151599

    Wong, D. M., Shen, Z., Owyang, K. E. and Martinez-Agosto, J. A. (2014). Insulin- and Warts-dependent regulation of tracheal plasticity modulates systemic larval growth during hypoxia in Drosophila melanogaster. PLoS One 9: e115297. PubMed ID: 25541690

    Ferguson, G. B. and Martinez-Agosto, J. A. (2014). Yorkie and Scalloped Signaling Regulates Notch-Dependent Lineage Specification during Drosophila Hematopoiesis. Curr Biol 24: 2665-2672. PubMed ID: 25454586

    Dragojlovic-Munther, M. and Martinez-Agosto, J. A. (2013). Extracellular matrix-modulated Heartless signaling in Drosophila blood progenitors regulates their differentiation via a Ras/ETS/FOG pathway and target of rapamycin function. Dev Biol 384: 313-330. PubMed ID: 23603494

    Dragojlovic-Munther, M. and Martinez-Agosto, J. A. (2012). Multifaceted roles of PTEN and TSC orchestrate growth and differentiation of Drosophila blood progenitors. Development 139: 3752-3763. PubMed ID: 22951642

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

    Dragojlovic-Munther, M., Martinez-Agosto, J. A. (2013) Extracellular matrix-modulated Heartless signaling in Drosophila blood progenitors regulates their differentiation via a Ras/ETS/FOG pathway and Target of Rapamycin function. Dev Biol. PubMed ID: 23603494

    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

    Mateus, A. M. and Martinez Arias, A. (2011). Patterned cell adhesion associated with tissue deformations during dorsal closure in Drosophila. PLoS One 6: e27159. PubMed ID: 22076130

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

  • Rui Martinho Department of Biomedical Sciences and Medicine, University of Algarve Campus de Gambelas, Faro Portugal
    Rathore, O. S., Silva, R. D., Ascensao-Ferreira, M., Matos, R., Carvalho, C., Marques, B., Tiago, M. N., Prudencio, P., Andrade, R. P., Roignant, J. Y., Barbosa-Morais, N. L. and Martinho, R. G. (2020). NineTeen Complex-subunit Salsa is required for efficient splicing of a subset of introns and dorsal-ventral patterning. Rna. PubMed ID: 32963109

    Ribeiro, A. L., Silva, R. D., Foyn, H., Tiago, M. N., Rathore, O. S., Arnesen, T. and Martinho, R. G. (2016). Naa50/San-dependent N-terminal acetylation of Scc1 is potentially important for sister chromatid cohesion. Sci Rep 6: 39118. PubMed ID: 27996020

    Ferreira, T., Prudencio, P. and Martinho, R. G. (2014). Drosophila protein kinase N (Pkn) is a negative regulator of actin-myosin activity during oogenesis. Dev Biol 394(2): 277-291. PubMed ID: 25131196

    Guilgur, L. G., Prudencio, P., Sobral, D., Liszekova, D., Rosa, A. and Martinho, R. G. (2014). Requirement for highly efficient pre-mRNA splicing during Drosophila early embryonic development. Elife 3: e02181. PubMed ID: 24755291

    Alves, E., Henriques, B. J., Rodrigues, J. V., Prudencio, P., Rocha, H., Vilarinho, L., Martinho, R. G. and Gomes, C. M. (2012). Mutations at the flavin binding site of ETF:QO yield a MADD-like severe phenotype in Drosophila. Biochim Biophys Acta 1822(8): 1284-1292. PubMed ID: 22580358

  • L. Miguel Martins MRC Toxicology Unit, University of Leicester
    Redhai, S., Pilgrim, C., Gaspar, P., Giesen, L. V., Lopes, T., Riabinina, O., Grenier, T., Milona, A., Chanana, B., Swadling, J. B., Wang, Y. F., Dahalan, F., Yuan, M., Wilsch-Brauninger, M., Lin, W. H., Dennison, N., Capriotti, P., Lawniczak, M. K. N., Baines, R. A., Warnecke, T., Windbichler, N., Leulier, F., Bellono, N. W. and Miguel-Aliaga, I. (2020). An intestinal zinc sensor regulates food intake and developmental growth. Nature 580(7802): 263-268. PubMed ID: 32269334

    Garrido-Maraver, J., Loh, S. H. Y. and Martins, L. M. (2019). Forcing contacts between mitochondria and the endoplasmic reticulum extends lifespan in a Drosophila model of Alzheimer's disease. Biol Open. PubMed ID: 31822473

    Celardo, I., Lehmann, S., Costa, A.C., Loh, S.H. and Martins, L. M. (2017).dATF4 regulation of mitochondrial folate-mediated one-carbon metabolism is neuroprotective. Cell Death Differ [Epub ahead of print]. PubMed ID: 28211874

    Lehmann, S., Loh, S. H. and Martins, L. M. (2016). Enhancing NAD+ salvage metabolism is neuroprotective in a PINK1 model of Parkinson's disease. Biol Open. PubMed ID: 28011627

    Lehmann, S., Loh, S. H. and Martins, L. M. (2016). Enhancing NAD+ salvage metabolism is neuroprotective in a PINK1 model of Parkinson's disease. Biol Open [Epub ahead of print]. PubMed ID: 28011627

    Celardo, I., Costa, A. C., Lehmann, S., Jones, C., Wood, N., Mencacci, N. E., Mallucci, G. R., Loh, S. H. and Martins, L. M. (2016). Mitofusin-mediated ER stress triggers neurodegeneration in pink1/parkin models of Parkinson's disease. Cell Death Dis 7: e2271. PubMed ID: 27336715

    Lehmann, S., Costa, A. C., Celardo, I., Loh, S. H. and Martins, L. M. (2016). Parp mutations protect against mitochondrial dysfunction and neurodegeneration in a PARKIN model of Parkinson's disease. Cell Death Dis 7: e2166. PubMed ID: 27031963

    Tufi, R., Gandhi, S., de Castro, I. P., Lehmann, S., Angelova, P. R., Dinsdale, D., Deas, E., Plun-Favreau, H., Nicotera, P., Abramov, A. Y., Willis, A. E., Mallucci, G. R., Loh, S. H. and Martins, L. M. (2014). Enhancing nucleotide metabolism protects against mitochondrial dysfunction and neurodegeneration in a PINK1 model of Parkinson's disease. Nat Cell Biol. PubMed ID: 24441527

    de Castro, I. P., Costa, A. C., Celardo, I., Tufi, R., Dinsdale, D., Loh, S. H. and Martins, L. M. (2013). Drosophila ref(2)P is required for the parkin-mediated suppression of mitochondrial dysfunction in pink1 mutants. Cell Death Dis 4: e873. PubMed ID: 24157867

    Costa, A. C., Loh, S. H., Martins, L. M. (2013) Drosophila Trap1 protects against mitochondrial dysfunction in a PINK1/parkin model of Parkinson's disease. Cell Death Dis 4: e467. PubMed ID: 23328674

  • John Masly Department of Biology, University of Oklahoma, Norman
    Frazee, S. R., Harper, A. R., Afkhami, M., Wood, M. L., McCrory, J. C. and Masly, J. P. (2021). Interspecific introgression reveals a role of male genital morphology during the evolution of reproductive isolation in Drosophila. Evolution. PubMed ID: 33433903

    Frazee, S. R. and Masly, J. P. (2015). Multiple sexual selection pressures drive the rapid evolution of complex morphology in a male secondary genital structure. Ecol Evol 5: 4437-4450. PubMed ID: 26664690

    Smith, B. N., Ghazanfari, A. M., Bohm, R. A., Welch, W. P., Zhang, B. and Masly, J. P. (2015). A Flippase-Mediated GAL80/GAL4 Intersectional Resource for Dissecting Appendage Development in Drosophila. G3 (Bethesda) 5: 2105-2112. PubMed ID: 26276385

    Masly, J. P. and Kamimura, Y. (2014). Asymmetric mismatch in strain-specific genital morphology causes increased harm to Drosophila females. Evolution 68: 2401-2411. PubMed ID: 24758304

  • James Mason University of Rochester Medical Center
    Brar, S. S., Petrovich, R. M., Williams, J. G. and Mason, J. M. (2013). Phosphorylation at Serines 216 and 221 Is Important for Drosophila HeT-A Gag Protein Stability. PLoS One 8: e75381. PubMed ID: 24058682

    Takacs, S., Biessmann, H., Reddy, H. M., Mason, J. M. and Torok, T. (2012). Protein interactions on telomeric retrotransposons in Drosophila. Int J Biol Sci 8: 1055-1061. PubMed ID: 22949888

    Antao, J. M., Mason, J. M., Dejardin, J. and Kingston, R. E. (2012). Protein landscape at Drosophila melanogaster telomere-associated sequence repeats. Mol Cell Biol 32: 2170-2182. PubMed ID: 22493064

    Dronamraju, R. and Mason, J. M. (2011). MU2 and HP1a regulate the recognition of double strand breaks in Drosophila melanogaster. PLoS One 6: e25439. PubMed ID: 21966530

  • Gregory Matera Department of Genetics, University of North Carolina, Chapel Hill
    Schmidt, C. A., Min, L. Y., McVay, M. H., Giusto, J. D., Brown, J. C., Salzler, H. R. and Matera, A. G. (2022). Mutations in Drosophila tRNA processing factors cause phenotypes similar to Pontocerebellar Hypoplasia. Biol Open 11(3). PubMed ID: 35132432

    Raimer, A. C., Singh, S. S., Edula, M. R., Paris-Davila, T., Vandadi, V., Spring, A. M. and Matera, A. G. (2020). Temperature-sensitive spinal muscular atrophy-causing point mutations lead to SMN instability, locomotor defects and premature lethality in Drosophila. Dis Model Mech 13(5). PubMed ID: 32501283

    Schmidt, C. A., Giusto, J. D., Bao, A., Hopper, A. K. and Matera, A. G. (2019). Molecular determinants of metazoan tricRNA biogenesis. Nucleic Acids Res. PubMed ID: 31032518

    Meers, M. P., Adelman, K., Duronio, R. J., Strahl, B. D., McKay, D. J. and Matera, A. G. (2018). Transcription start site profiling uncovers divergent transcription and enhancer-associated RNAs in Drosophila melanogaster. BMC Genomics 19(1): 157. PubMed ID: 29466941

    Gray, K. M., Kaifer, K. A., Baillat, D., Wen, Y., Bonacci, T. R., Ebert, A. D., Raimer, A. C., Spring, A. M., Have, S. T., Glascock, J. J., Gupta, K., Van Duyne, G. D., Emanuele, M. J., Lamond, A. I., Wagner, E. J., Lorson, C. L. and Matera, A. G. (2017). Self-oligomerization regulates stability of Survival Motor Neuron (SMN) protein isoforms by sequestering an SCF(Slmb) degron. Mol Biol Cell. PubMed ID: 29167380

    Meers, M. P., Henriques, T., Lavender, C. A., McKay, D. J., Strahl, B. D., Duronio, R. J., Adelman, K. and Matera, A. G. (2017). Histone gene replacement reveals a post-transcriptional role for H3K36 in maintaining metazoan transcriptome fidelity. Elife 6 [Epub ahead of print]. PubMed ID: 28346137

    Garcia, E. L., Wen, Y., Praveen, K. and Matera, A. G. (2016). Transcriptomic comparison of Drosophila snRNP biogenesis mutants reveals mutant-specific changes in pre-mRNA processing: implications for spinal muscular atrophy. RNA [Epub ahead of print]. PubMed ID: 27268418

    Lu, Z. and Matera, A. G. (2014). Developmental Analysis of Spliceosomal snRNA Isoform Expression. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25416704

    Praveen, K., Wen, Y., Gray, K. M., Noto, J. J., Patlolla, A. R., Van Duyne, G. D. and Matera, A. G. (2014). SMA-Causing Missense Mutations in Survival motor neuron (Smn) Display a Wide Range of Phenotypes When Modeled in Drosophila. PLoS Genet 10: e1004489. PubMed ID: 25144193

    Lu, Z., Guan, X., Schmidt, C. A. and Matera, A. G. (2014). RIP-seq analysis of eukaryotic Sm proteins identifies three major categories of Sm-containing ribonucleoproteins. Genome Biol 15: R7. PubMed ID: 24393626

    Garcia, E. L., Lu, Z., Meers, M. P., Praveen, K. and Matera, A. G. (2013). Developmental arrest of Drosophila survival motor neuron (Smn) mutants accounts for differences in expression of minor intron-containing genes. RNA. PubMed ID: 24006466

    Natalizio, A. H. and Matera, A. G. (2013). Identification and characterization of Drosophila Snurportin reveals a role for the import receptor Moleskin/Importin7 in snRNP biogenesis. Mol Biol Cell. PubMed ID: 23885126

  • Maja Matis Institute of Cell Biology, Münster
    Panzade, S. and Matis, M. (2021). The Microtubule Minus-End Binding Protein Patronin Is Required for the Epithelial Remodeling in the Drosophila Abdomen. Front Cell Dev Biol 9: 682083. PubMed ID: 34368132

    Singh, A., Saha, T., Begemann, I., Ricker, A., Nusse, H., Thorn-Seshold, O., Klingauf, J., Galic, M. and Matis, M. (2018). Polarized microtubule dynamics directs cell mechanics and coordinates forces during epithelial morphogenesis. Nat Cell Biol 20(10): 1126-1133. PubMed ID: 30202051

    Galic, M. and Matis, M. (2015). Polarized trafficking provides spatial cues for planar cell polarization within a tissue. Bioessays 37(6): 678-686. PubMed ID: 25845311

    Olofsson, J., Sharp, K. A., Matis, M., Cho, B. and Axelrod, J. D. (2014). Prickle/spiny-legs isoforms control the polarity of the apical microtubule network in planar cell polarity. Development 141(14): 2866-2874. PubMed ID: 25005476

  • Margarida Matos Centre for Ecology, Evolution and Environmental Changes, Lisbon University, Portugal
    Simoes, P., Santos, M. A., Carromeu-Santos, A., Quina, A. S., Santos, M. and Matos, M. (2020). Beneficial developmental acclimation in reproductive performance under cold but not heat stress. J Therm Biol 90: 102580. PubMed ID: 32479384

    Simoes, P., Fragata, I., Santos, J., Santos, M. A., Santos, M., Rose, M. R. and Matos, M. (2019). How phenotypic convergence arises in experimental evolution. Evolution. PubMed ID: 31329268

    Seabra, S. G., Fragata, I., Antunes, M. A., Faria, G. S., Santos, M. A., Sousa, V. C., Simoes, P. and Matos, M. (2017). Different genomic changes underlie adaptive evolution in populations of contrasting history. Mol Biol Evol. PubMed ID: 29029198

    Simoes, P., Fragata, I., Seabra, S. G., Faria, G. S., Santos, M. A., Rose, M. R., Santos, M. and Matos, M. (2017). Predictable phenotypic, but not karyotypic, evolution of populations with contrasting initial history. Sci Rep 7(1): 913. PubMed ID: 28424494

    Simoes, P., Fragata, I., Lopes-Cunha, M., Lima, M., Kellen, B., Barbaro, M., Santos, M. and Matos, M. (2015). Wing trait-inversion associations in Drosophila subobscura can be generalized within continents, but may change through time. J Evol Biol 28(12): 2163-2174. PubMed ID: 26302686

    Fragata, I., Lopes-Cunha, M., Barbaro, M., Kellen, B., Lima, M., Santos, M. A., Faria, G. S., Santos, M., Matos, M. and Simoes, P. (2014). How much can history constrain adaptive evolution? A real-time evolutionary approach of inversion polymorphisms in Drosophila subobscura. J Evol Biol 27(12): 2727-2738. PubMed ID: 25430492

    Simoes, P., Pascual, M., Coelho, M. M. and Matos, M. (2010). Divergent evolution of molecular markers during laboratory adaptation in Drosophila subobscura. Genetica 138(9-10): 999-1009. PubMed ID: 20803349

  • Kenji Matsuno Department of Biological Sciences, Osaka University
    Das, P., Salazar, J. L., Li-Kroeger, D., Yamamoto, S., Nakamura, M., Sasamura, T., Inaki, M., Masuda, W., Kitagawa, M., Yamakawa, T. and Matsuno, K. (2019). Maternal almondex, a neurogenic gene, is required for proper subcellular Notch distribution in early Drosophila embryogenesis. Dev Growth Differ. PubMed ID: 31782145

    Ishibashi, T., Hatori, R., Maeda, R., Nakamura, M., Taguchi, T., Matsuyama, Y. and Matsuno, K. (2019). E and ID proteins regulate cell chirality and left-right asymmetric development in Drosophila. Genes Cells. PubMed ID: 30624823

    Inatomi, M., Shin, D., Lai, Y. T. and Matsuno, K. (2019). Proper direction of male genitalia is prerequisite for copulation in Drosophila, implying cooperative evolution between genitalia rotation and mating behavior. Sci Rep 9(1): 210. PubMed ID: 30659250

    Yamakawa, T., Atsumi, Y., Kubo, S., Yamagishi, A., Morita, I. and Matsuno, K. (2018). Insight into Notch signaling steps that involve pecanex from dominant-modifier screens in Drosophila. Genetics. Pubmed ID: 29853475

    Matsumoto, K., Ayukawa, T., Ishio, A., Sasamura, T., Yamakawa, .T and Matsuno, K. (2016). Dual roles of O-glucose glycans redundant with monosaccharide O-fucose on Notch in Notch trafficking. J Biol Chem [Epub ahead of print]. PubMed ID: 27129198

    Okumura, T., Sasamura, T., Inatomi, M., Hozumi, S., Nakamura, M., Hatori, R., Taniguchi, K., Nakazawa, N., Suzuki, E., Maeda, R., Yamakawa, T. and Matsuno, K. (2015). Class I myosins have overlapping and specialized gunctions in left-right asymmetric development in Drosophila. Genetics. PubMed ID: 25659376

    Ishio, A., Sasamura, T., Ayukawa, T., Kuroda, J., Ishikawa, H. O., Aoyama, N., Matsumoto, K., Gushiken, T., Okajima, T., Yamakawa, T. and Matsuno, K. (2014). O-fucose monosaccharide of Drosophila Notch has a temperature-sensitive function and cooperates with O-glucose glycan in Notch transport and Notch signaling activation. J Biol Chem. PubMed ID: 25378397

    Sawamura, K., Maehara, K., Keira, Y., Ishikawa, H. O., Sasamura, T., Yamakawa, T. and Matsuno, K. (2014). A Test of Double Interspecific Introgression of Nucleoporin Genes in Drosophila. G3 (Bethesda). PubMed ID: 25172915

    Hatori, R., Ando, T., Sasamura, T., Nakazawa, N., Nakamura, M., Taniguchi, K., Hozumi, S., Kikuta, J., Ishii, M. and Matsuno, K. (2014). Left-right asymmetry is formed in individual cells by intrinsic cell chirality. Mech Dev. PubMed ID: 24800645

    Nakayama, M., et al. (2014). A gain-of-function screen to identify genes that reduce lifespan in the adult of Drosophila melanogaster. BMC Genet 15: 46. PubMed ID: 24739137


  • Takashi Matsuo Department of Agricultural and Environmental Biology, University of Tokyo, Yayoi, Tokyo, Japan
    Minekawa, K., Amino, K. and Matsuo, T. (2020). A courtship behavior that makes monandrous females polyandrous. Evolution. PubMed ID: 32944943

    Tomioka, S., Aigaki, T., Matsuo, T. (2012) Conserved cis-regulatory elements of two odorant-binding protein genes, Obp57d and Obp57e, in Drosophila. Genes Genet Syst 87: 323-329. PubMed ID: 23412634

    Harada, E., Nakagawa, J., Asano, T., Taoka, M., Sorimachi, H., Ito, Y., Aigaki, T. and Matsuo, T. (2012). Functional evolution of duplicated odorant-binding protein genes, Obp57d and Obp57e, in Drosophila. PLoS One 7: e29710. PubMed ID: 22238638

    Mitaka, H., Matsuo, T., Miura, N. and Ishikawa, Y. (2011). Identification of odorant-binding protein genes from antennal expressed sequence tags of the onion fly, Delia antiqua. Mol Biol Rep 38: 1787-1792. PubMed ID: 20848218

  • Fumio Matsuzaki Riken Center for Developmental Biology, Kobe
    Kono, K., Yoshiura, S., Fujita, I., Okada, Y., Shitamukai, A., Shibata, T. and Matsuzaki, F. (2019). Reconstruction of Par-dependent polarity in apolar cells reveals a dynamic process of cortical polarization. Elife 8. PubMed ID: 31172945

    Saadaoui, M., Konno, D., Loulier, K., Goiame, R., Jadhav, V., Mapelli, M., Matsuzaki, F. and Morin, X. (2017). Loss of the canonical spindle orientation function in the Pins/LGN homolog AGS3. EMBO Rep. PubMed ID: 28684399

    Yoshiura, S., Ohta, N. and Matsuzaki, F. (2012). Tre1 GPCR signaling orients stem cell divisions in the Drosophila central nervous system. Dev Cell 22: 79-91. PubMed ID: 22178499

    Wang, C., Li, S., Januschke, J., Rossi, F., Izumi, Y., Garcia-Alvarez, G., Gwee, S. S., Soon, S. B., Sidhu, H. K., Yu, F., Matsuzaki, F., Gonzalez, C. and Wang, H. (2011). An ana2/ctp/mud complex regulates spindle orientation in Drosophila neuroblasts. Dev Cell 21: 520-533. PubMed ID: 21920316

    Kitajima, A., Fuse, N., Isshiki, T. and Matsuzaki, F. (2010). Progenitor properties of symmetrically dividing Drosophila neuroblasts during embryonic and larval development. Dev Biol 347: 9-23. PubMed ID: 20599889

  • Erika Matunis Department of Cell Biology, Johns Hopkins University School of Medicine
    Mahadevaraju, S., Fear, J. M., Akeju, M., Galletta, B. J., Pinheiro, M., Avelino, C. C., Cabral-de-Mello, D. C., Conlon, K., Dell'Orso, S., Demere, Z., Mansuria, K., Mendonça, C. A., Palacios-Gimenez, O. M., Ross, E., Savery, M., Yu, K., Smith, H. E., Sartorelli, V., Yang, H., Rusan, N. M., Vibranovski, M. D., Matunis, E. and Oliver, B. (2021). Dynamic sex chromosome expression in Drosophila male germ cells. Nat Commun 12(1): 892. PubMed ID: 33563972

    Greenspan, L. J. and Matunis, E. L. (2018). Retinoblastoma intrinsically regulates niche cell quiescence, identity, and niche number in the adult Drosophila testis. Cell Rep 24(13): 3466-3476.e3468. PubMed ID: 30257208

    Ma, Q., de Cuevas, M. and Matunis, E.L. (2016). Chinmo is sufficient to induce male fate in somatic cells of the adult Drosophila ovary. Development [Epub ahead of print]. PubMed ID: 26811385

    Greenspan, L. J., de Cuevas, M. and Matunis, E. (2015). Genetics of Gonadal Stem Cell Renewal. Annu Rev Cell Dev Biol. PubMed ID: 26355592

    Hasan, S., Hétié, P. and Matunis, E.L. (2015). Niche signaling promotes stem cell survival in the Drosophila testis via the Jak-STAT target DIAP1. Dev Biol [Epub ahead of print]. PubMed ID: 25941003

    Ma, Q., Wawersik, M. and Matunis, E. L. (2014). The Jak-STAT Target Chinmo Prevents Sex Transformation of Adult Stem Cells in the Drosophila Testis Niche. Dev Cell 31: 474-486. PubMed ID: 25453558

    Stine, R. R., Greenspan, L. J., Ramachandran, K. V. and Matunis, E. L. (2014). Coordinate regulation of stem cell competition by Slit-Robo and JAK-STAT signaling in the Drosophila testis. PLoS Genet 10: e1004713. PubMed ID: 25375180

    Li, Y., Ma, Q., Cherry, C. M. and Matunis, E. L. (2014). Steroid signaling promotes stem cell maintenance in the Drosophila testis. Dev Biol. PubMed ID: 25093968

    Hetie, P., de Cuevas, M. and Matunis, E. (2014). Conversion of Quiescent Niche Cells to Somatic Stem Cells Causes Ectopic Niche Formation in the Drosophila Testis. Cell Rep. PubMed ID: 24746819

    Stine, R. R., Matunis, E. L. (2013) Stem cell competition: finding balance in the niche. Trends Cell Biol. PubMed ID: 23597843

  • Cédric Maurange Developmental Biology Unit Marseille
    Genovese, S., Clement, R., Gaultier, C., Besse, F., Narbonne-Reveau, K., Daian, F., Foppolo, S., Luis, N. M. and Maurange, C. (2019). Coopted temporal patterning governs cellular hierarchy, heterogeneity and metabolism in Drosophila neuroblast tumors. Elife 8. PubMed ID: 31566561

    Narbonne-Reveau, K. and Maurange, C. (2019). Developmental regulation of regenerative potential in Drosophila by ecdysone through a bistable loop of ZBTB transcription factors. PLoS Biol 17(2): e3000149. PubMed ID: 30742616

    Dillard, C., Narbonne-Reveau, K., Foppolo, S., Lanet, E. and Maurange, C. (2018). Two distinct mechanisms silence chinmo in Drosophila neuroblasts and neuroepithelial cells to limit their self-renewal. Development 145(2). PubMed ID: 29361557

    Narbonne-Reveau, K., Lanet, E., Dillard, C., Foppolo, S., Chen, C. H., Parrinello, H., Rialle, S., Sokol, N. S. and Maurange, C. (2016). Neural stem cell-encoded temporal patterning delineates an early window of malignant susceptibility in Drosophila. Elife 5 [Epub ahead of print]. PubMed ID: 27296804

    Lanet, E. and Maurange, C. (2014). Building a brain under nutritional restriction: insights on sparing and plasticity from studies. Front Physiol 5: 117. PubMed ID: 24723892

    Lanet, E., Gould, A. P., Maurange, C. (2013) Protection of Neuronal Diversity at the Expense of Neuronal Numbers during Nutrient Restriction in the Drosophila Visual System. Cell Rep. PubMed ID: 23478023

    Maurange, C. (2012). Temporal specification of neural stem cells: insights from Drosophila neuroblasts. Curr Top Dev Biol 98: 199-228. PubMed ID: 22305164

  • Daniel Matute Biology Department, University of North Carolina, Chapel Hill
    Serrato-Capuchina, A., Wang, J., Earley, E., Peede, D., Isbell, K. and Matute, D. R. (2020). Paternally inherited P-element copy number affects the magnitude of hybrid dysgenesis in Drosophila simulans and D. melanogaster. Genome Biol Evol. PubMed ID: 32339225

    Comeault, A. A. and Matute, D. R. (2018). Genetic divergence and the number of hybridizing species affect the path to homoploid hybrid speciation. Proc Natl Acad Sci U S A 115(39): 9761-9766. PubMed ID: 30209213

    Cooper, B. S., Sedghifar, A., Nash, W. T., Comeault, A. A. and Matute, D. R. (2018). A Maladaptive Combination of Traits Contributes to the Maintenance of a Drosophila Hybrid Zone. Curr Biol. PubMed ID: 30174184

    Turissini, D. A., McGirr, J. A., Patel, S. S., David, J. R. and Matute, D. R. (2017). The rate of evolution of postmating-prezygotic reproductive isolation in Drosophila. Mol Biol Evol 35(2):312-334. PubMed ID: 29048573

    Turissini, D. A. and Matute, D. R. (2017). Fine scale mapping of genomic introgressions within the Drosophila yakuba clade. PLoS Genet 13(9): e1006971. PubMed ID: 28873409

    Turissini, D. A., Comeault, A. A., Liu, G., Lee, Y. C. and Matute, D. R. (2017). The ability of Drosophila hybrids to locate food declines with parental divergence. Evolution. PubMed ID: 28085186

    Cooper, B. S., Ginsberg, P. S., Turelli, M. and Matute, D. R. (2017). Wolbachia in the Drosophila yakuba Complex: Pervasive Frequency Variation and Weak Cytoplasmic Incompatibility, but No Apparent Effect on Reproductive Isolation. Genetics 205(1): 333-351. PubMed ID: 27821433

    Miller, C. J. and Matute, D. R. (2016). The Effect of Temperature on Drosophila Hybrid Fitness. G3 (Bethesda). PubMed ID: 27913636

    Comeault, A. A., Venkat, A. and Matute, D. R. (2016). Correlated evolution of male and female reproductive traits drive a cascading effect of reinforcement in Drosophila yakuba. Proc Biol Sci 283(1835). PubMed ID: 27440664

  • Ugo Mayor Ikerbasque, Basque Foundation for Science, Bizkaia, Spain
    Martinez, A., Lectez, B., Ramirez, J., Popp, O., Sutherland, J. D., Urbe, S., Dittmar, G., Clague, M. J. and Mayor, U. (2017). Quantitative proteomic analysis of Parkin substrates in Drosophila neurons. Mol Neurodegener 12(1): 29. PubMed ID: 28399880

    Ramirez, J., Martinez, A., Lectez, B., Lee, S. Y., Franco, M., Barrio, R., Dittmar, G. and Mayor, U. (2015). Proteomic Analysis of the Ubiquitin Landscape in the Drosophila Embryonic Nervous System and the Adult Photoreceptor Cells. PLoS One 10(10): e0139083. PubMed ID: 26460970

    Talamillo, A., Herboso, L., Pirone, L., Perez, C., Gonzalez, M., Sanchez, J., Mayor, U., Lopitz-Otsoa, F., Rodriguez, M. S., Sutherland, J. D. and Barrio, R. (2013). Scavenger receptors mediate the role of SUMO and Ftz-f1 in Drosophila steroidogenesis. PLoS Genet 9(4): e1003473. PubMed ID: 23637637

  • Alexander Mazo Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia
    Kohzaki, H., Asano, M., Murakami, Y. and Mazo, A. (2020). Epigenetic regulation affects gene amplification in Drosophila development. Front Biosci (Landmark Ed) 25: 632-645. PubMed ID: 31585908

    Li, T., Hodgson, J. W., Petruk, S., Mazo, A. and Brock, H. W. (2017). Additional sex combs interacts with enhancer of zeste and trithorax and modulates levels of trimethylation on histone H3K4 and H3K27 during transcription of hsp70. Epigenetics Chromatin 10(1): 43. PubMed ID: 28927461

    Black, K, L., Petruk, S., Fenstermaker, T. K., Hodgson, J. W., Caplan, J. L., Brock, H. W. and Mazo, A. (2016). Chromatin proteins and RNA are associated with DNA during all phases of mitosis. Cell Discov 2: 16038. PubMed ID: 27807477

    Petruk, S., Black, K. L., Kovermann, S. K., Brock, H. W. and Mazo, A. (2013). Stepwise histone modifications are mediated by multiple enzymes that rapidly associate with nascent DNA during replication. Nat Commun 4: 2841. PubMed ID: 24276476

    Carbonell, A., Mazo, A., Serras, F. and Corominas, M. (2013). Ash2 acts as an ecdysone receptor coactivator by stabilizing the histone methyltransferase Trr. Mol Biol Cell 24: 361-372. PubMed ID: 23197473

    Petruk, S., Sedkov, Y., Johnston, D. M., Hodgson, J. W., Black, K. L., Kovermann, S. K., Beck, S., Canaani, E., Brock, H. W. and Mazo, A. (2012). TrxG and PcG proteins but not methylated histones remain associated with DNA through replication. Cell 150: 922-933. PubMed ID: 22921915

    Johnston, D. M., Sedkov, Y., Petruk, S., Riley, K. M., Fujioka, M., Jaynes, J. B. and Mazo, A. (2011). Ecdysone- and NO-mediated gene regulation by competing EcR/Usp and E75A nuclear receptors during Drosophila development. Mol Cell 44: 51-61. PubMed ID: 21981918

    Brock, H. W., Hodgson, J. W., Petruk, S. and Mazo, A. (2009). Regulatory noncoding RNAs at Hox loci. Biochem Cell Biol 87: 27-34. PubMed ID: 19234521

  • Rachid Mazroui Faculty of Medicine, Laval University, Quebec
    Gareau, C., Houssin, E., Martel, D., Coudert, L., Mellaoui, S., Huot, M. E., Laprise, P. and Mazroui, R. (2013). Characterization of fragile x mental retardation protein recruitment and dynamics in Drosophila stress granules. PLoS One 8: e55342. PubMed ID: 23408971

    Gareau, C., Martel, D., Coudert, L., Mellaoui, S. and Mazroui, R. (2013). Characterization of Fragile X Mental Retardation Protein granules formation and dynamics in Drosophila. Biol Open 2: 68-81. PubMed ID: 23336078

  • Brian McCabeÉcole Polytechnique Fédérale De Lausanne
    Banerjee, S., Vernon, S., Jiao, W., Choi, B. J., Ruchti, E., Asadzadeh, J., Burri, O., Stowers, R. S. and McCabe, B. D. (2021). Miniature neurotransmission is required to maintain Drosophila synaptic structures during ageing. Nat Commun 12(1): 4399. PubMed ID: 34285221

    Choi, B. J., Imlach, W. L., Jiao, W., Wolfram, V., Wu, Y., Grbic, M., Cela, C., Baines, R. A., Nitabach, M. N. and McCabe, B. D. (2014). Miniature neurotransmission regulates Drosophila synaptic structural maturation. Neuron 82: 618-634. PubMed ID: 24811381

    Lotti, F., Imlach, W. L., Saieva, L., Beck, E. S., Hao le, T., Li, D. K., Jiao, W., Mentis, G. Z., Beattie, C. E., McCabe, B. D. and Pellizzoni, L. (2012). An SMN-dependent U12 splicing event essential for motor circuit function. Cell 151: 440-454. PubMed ID: 23063131

    Imlach, W. L., Beck, E. S., Choi, B. J., Lotti, F., Pellizzoni, L. and McCabe, B. D. (2012). SMN is required for sensory-motor circuit function in Drosophila. Cell 151: 427-439. PubMed ID: 23063130

    Beck, E. S., et al. (2012). Regulation of Fasciclin II and synaptic terminal development by the splicing factor Beag. J. Neurosci. 32(20): 7058-73. PubMed Citation: 22593074

  • Kim McCall Department of Biology, Boston University
    Lebo, D. P. V., Chirn, A., Taylor, J. D., Levan, A., Doerre Torres, V., Agreda, E., Serizier, S. B., Lord, A. K., Jenkins, V. K. and McCall, K. (2021). An RNAi screen of the kinome in epithelial follicle cells of the Drosophila melanogaster ovary reveals genes required for proper germline death and clearance. G3 (Bethesda) 11(2). PubMed ID: 33693600

    Yalonetskaya, A., Mondragon, A. A., Hintze, Z. J., Holmes, S. and McCall, K. (2019). Nuclear degradation dynamics in a nonapoptotic programmed cell death. Cell Death Differ. PubMed ID: 31285547

    Mondragon, A. A., Yalonetskaya, A., Ortega, A. J., Zhang, Y., Naranjo, O., Elguero, J., Chung, W. S. and McCall, K. (2019). Lysosomal machinery drives extracellular acidification to direct non-apoptotic cell death. Cell Rep 27(1): 11-19.e13. PubMed ID: 30943394

    Santoso, C. S., Meehan, T. L., Peterson, J. S., Cedano, T. M., Turlo, C. V. and McCall, K. (2018). The ABC transporter Eato promotes cell clearance in the Drosophila melanogaster ovary. G3 (Bethesda). PubMed ID: 29295819

    Meehan, T.L., Joudi, T.F., Timmons, A.K., Taylor, J.D., Habib, C.S., Peterson, J.S., Emmanuel, S., Franc, N.C. and McCall, K. (2016). Components of the engulfment machinery have distinct roles in corpse processing. PLoS One 11: e0158217. PubMed ID: 27347682

    Etchegaray, J. I., Elguero, E. J., Tran, J. A., Sinatra, V., Feany, M. B. and McCall, K. (2016). Defective phagocytic corpse processing results in neurodegeneration and can be rescued by TORC1 activation. J Neurosci 36: 3170-3183. PubMed ID: 26985028

    Timmons, A. K., Mondragon, A. A., Schenkel, C. E., Yalonetskaya, A., Taylor, J. D., Moynihan, K. E., Etchegaray, J. I., Meehan, T. L. and McCall, K. (2016). Phagocytosis genes nonautonomously promote developmental cell death in the Drosophila ovary. Proc Natl Acad Sci U S A. PubMed ID: 26884181

    Meehan, T. L., Kleinsorge, S. E., Timmons, A. K., Taylor, J. D. and McCall, K. (2015). Polarization of the epithelial layer and apical localization of integrins are required for engulfment of apoptotic cells. Dis Model Mech. PubMed ID: 26398951

    Peterson, J. S. and McCall, K. (2013). Combined Inhibition of Autophagy and Caspases Fails to Prevent Developmental Nurse Cell Death in the Ovary. PLoS One 8: e76046. PubMed ID: 24098761

    Jenkins, V. K., Timmons, A. K. and McCall, K. (2013). Diversity of cell death pathways: insight from the fly ovary. Trends Cell Biol. PubMed ID: 23968895

    Timmons, A. K., Meehan, T. L., Gartmond, T. D. and McCall, K. (2013). Use of necrotic markers in the Drosophila ovary. Methods Mol Biol 1004: 215-228. PubMed ID: 23733580

  • Brooke McCartney Department of Biological Sciences Carnegie Mellon University, Pittsburgh
    Logan, G., Chou, W. C. and McCartney, B. M. (2022). A Diaphanous and Enabled-dependent asymmetric actin cable array repositions nuclei during Drosophila oogenesis. Development 149(13). PubMed ID: 35686626

    Keith, S. A., Bishop, C., Fallacaro, S. and McCartney, B. M. (2021). Arc1 and the microbiota together modulate growth and metabolic traits in Drosophila. Development. PubMed ID: 34240138

    Molinar-Inglis, O., Oliver, S. L., Davison, P., Kunttas, E. and McCartney, B. M. (2018). APC2 associates with the actin cortex through a multi-part mechanism to regulate cortical actin organization and dynamics in the Drosophila ovary. Cytoskeleton (Hoboken). PubMed ID: 30019417

    Kunttas-Tatli, E., Roberts, D. M. and McCartney, B. M. (2014). Self-association of the APC tumor suppressor is required for the assembly, stability, and activity of the Wnt signaling destruction complex. Mol Biol Cell. PubMed ID: 25208568

    Komori, H., Xiao, Q., McCartney, B. M. and Lee, C. Y. (2013). Brain tumor specifies intermediate progenitor cell identity by attenuating beta-catenin/Armadillo activity. Development. PubMed ID: 24257623

    Jaiswal, R., Stepanik, V., Rankova, A., Molinar, O., Goode, B. L., McCartney, B. (2013) Drosophila homologues of Adenomatous polyposis coli (APC) and the formin Diaphanous collaborate by a conserved mechanism to stimulate actin filament assembly. J Biol Chem. PubMed ID: 23558679

    Kunttas-Tatli, E., Zhou, M. N., Zimmerman, S., Molinar, O., Zhouzheng, F., Carter, K., Kapur, M., Cheatle, A., Decal, R. and McCartney, B. M. (2012). Destruction complex function in the Wnt signaling pathway of Drosophila requires multiple interactions between Adenomatous polyposis coli 2 and Armadillo. Genetics 190: 1059-1075. PubMed ID: 22174073

    Zhou, M. N., Kunttas-Tatli, E., Zimmerman, S., Zhouzheng, F. and McCartney, B. M. (2011). Cortical localization of APC2 plays a role in actin organization but not in Wnt signaling in Drosophila. J Cell Sci 124: 1589-1600. PubMed ID: 21486956

  • John McDermott York University, Toronto
    Belozerov, V. E., Ratkovic, S., McNeill, H., Hilliker, A. J. and McDermott, J. C. (2013). In vivo interaction proteomics reveal a novel p38MAPK/Rack1 pathway regulating proteostasis in Drosophila muscle. Mol Cell Biol. PubMed ID: 24277934

    Belozerov, V. E., Lin, Z. Y., Gingras, A. C., McDermott, J. C. and Michael Siu, K. W. (2012). High-resolution protein interaction map of the Drosophila melanogaster p38 mitogen-activated protein kinases reveals limited functional redundancy. Mol Cell Biol 32: 3695-3706. PubMed ID: 22801366

  • Jocelyn McDonald Department of Molecular Genetics, Lerner Research Institute, Cleveland
    Kotian, N., Troike, K. M., Curran, K. N., Lathia, J. D. and McDonald, J. A. (2021). A Drosophila RNAi screen reveals conserved glioblastoma-related adhesion genes that regulate collective cell migration. G3 (Bethesda) 12(1). PubMed ID: 34849760

    Chen, Y., Kotian, N., Aranjuez, G., Chen, L., Messer, C. L., Burtscher, A., Sawant, K., Ramel, D., Wang, X. and McDonald, J. A. (2020). Protein phosphatase 1 activity controls a balance between collective and single cell modes of migration. Elife 9. PubMed ID: 32369438

    Sawant, K., Chen, Y., Kotian, N., Preuss, K. M. and McDonald, J. A. (2018). Rap1 GTPase promotes coordinated collective cell migration in vivo. Mol Biol Cell: mbcE17120752. PubMed ID: 30156466

    Klebanow, L. R., Peshel, E. C., Schuster, A. T., De, K., Sarvepalli, K., Lemieux, M. E., Lenoir, J. J., Moore, A. W., McDonald, J. A. and Longworth, M. S. (2016). Drosophila Condensin II subunit, Chromosome Associated Protein-D3, regulates cell fate determination through non-cell autonomous signaling. Development. PubMed ID: 27317808

    Aranjuez, G., Burtscher, A., Sawant, K., Majumder, P. and McDonald, J. A. (2016). Dynamic myosin activation promotes collective morphology and migration by locally balancing oppositional forces from surrounding tissue. Mol Biol Cell. PubMed ID: 27122602

    Geisbrecht, E. R., Sawant, K., Su, Y., Liu, Z. C., Silver, D. L., Burtscher, A., Wang, X., Zhu, A. J. and McDonald, J. A. (2013). Genetic interaction screens identify a role for hedgehog signaling in Drosophila border cell migration. Dev Dyn. PubMed ID: 23335293

    Aranjuez, G., Kudlaty, E., Longworth, M. S. and McDonald, J. A. (2012). On the role of PDZ domain-encoding genes in Drosophila border cell migration. G3 (Bethesda) 2: 1379-1391. PubMed ID: 23173089

    Majumder, P., Aranjuez, G., Amick, J. and McDonald, J. A. (2012). Par-1 controls myosin-II activity through myosin phosphatase to regulate border cell migration. Curr Biol 22: 363-372. PubMed ID: 22326025

  • William McGinnis Division of Biological Sciences, U. C. San Diego
    Capilla, A., Karachentsev, D., Patterson, R. A., Hermann, A., Juarez, M. T. and McGinnis, W. (2017). Toll pathway is required for wound-induced expression of barrier repair genes in the Drosophila epidermis. Proc Natl Acad Sci U S A. PubMed ID: 28289197

    Juarez, M. T., Patterson, R. A., Li, W. and McGinnis, W. (2013). Microinjection Wound Assay and In vivo Localization of Epidermal Wound Response Reporters in Drosophila Embryos. J Vis Exp. PubMed ID: 24300796

    Patterson, R. A., Juarez, M. T., Hermann, A., Sasik, R., Hardiman, G., McGinnis, W. (2013) Serine proteolytic pathway activation reveals an expanded ensemble of wound response genes in Drosophila. PLoS One 8: e61773. PubMed ID: 23637905

    Lemons, D., Pare, A. and McGinnis, W. (2012). Three Drosophila Hox complex microRNAs do not have major effects on expression of evolutionarily conserved Hox gene targets during embryogenesis. PLoS One 7: e31365. PubMed ID: 22393361

    Juarez, M. T., Patterson, R. A., Sandoval-Guillen, E. and McGinnis, W. (2011). Duox, Flotillin-2, and Src42A are required to activate or delimit the spread of the transcriptional response to epidermal wounds in Drosophila. PLoS Genet 7: e1002424. PubMed ID: 22242003

    McHale, P., Mizutani, C. M., Kosman, D., MacKay, D. L., Belu, M., Hermann, A., McGinnis, W., Bier, E. and Hwa, T. (2011). Gene length may contribute to graded transcriptional responses in the Drosophila embryo. Dev Biol 360: 230-240. PubMed ID: 21920356

  • Elizabeth McGraw School of Biological Sciences, Monash University, Clayton, Victoria, Australia
    Ye, Y. H., Seleznev, A., Flores, H. A., Woolfit, M. and McGraw, E. A. (2016). Gut microbiota in Drosophila melanogaster interacts with Wolbachia but does not contribute to Wolbachia-mediated antiviral protection. J Invertebr Pathol 143: 18-25. PubMed ID: 27871813

    McGuigan, K., Collet, J. M., McGraw, E. A., Ye, Y. H., Allen, S. L., Chenoweth, S. F. and Blows, M. W. (2014). The nature and extent of mutational pleiotropy in gene expression of male Drosophila serrata. Genetics 196(3): 911-921. PubMed ID: 21859495

  • Alistair McGregor Department of Biological and Medical Sciences, Oxford Brookes University
    Hagen, J. F. D., Mendes, C. C., Booth, S. R., Jimenez, J. F., Tanaka, K. M., Franke, F. A., Baudouin-Gonzalez, L., Ridgway, A. M., Arif, S., Nunes, M. D. S. and McGregor, A. P. (2020). Unravelling the genetic basis for the rapid diversification of male genitalia between Drosophila species. Mol Biol Evol. PubMed ID: 32931587

    Holzem, M., Franke, F. A., Mendes, C. C. and McGregor, A. P. (2020). Wnt gene regulation and function during maxillary palp development in Drosophila melanogaster. Dev Biol. PubMed ID: 32229133

    Gaspar, P., Arif, S., Sumner-Rooney, L., Kittelmann, M., Bodey, A. J., Stern, D. L., Nunes, M. D. S. and McGregor, A. P. (2020). Characterization of the Genetic Architecture Underlying Eye Size Variation Within Drosophila melanogaster and Drosophila simulans. G3 (Bethesda). PubMed ID: 31919111

    Kittelmann, S., Buffry, A. D., Franke, F. A., Almudi, I., Yoth, M., Sabaris, G., Couso, J. P., Nunes, M. D. S., Frankel, N., Gomez-Skarmeta, J. L., Pueyo-Marques, J., Arif, S. and McGregor, A. P. (2018). Gene regulatory network architecture in different developmental contexts influences the genetic basis of morphological evolution. PLoS Genet 14(5): e1007375. PubMed ID: 29723190

    Kittelmann, S., Buffry, A. D., Franke, F. A., Almudi, I., Yoth, M., Sabaris, G., Couso, J. P., Nunes, M. D. S., Frankel, N., Gomez-Skarmeta, J. L., Pueyo-Marques, J., Arif, S. and McGregor, A. P. (2018). Gene regulatory network architecture in different developmental contexts influences the genetic basis of morphological evolution. PLoS Genet 14(5): e1007375. PubMed ID: 29723190

    Tanaka, K. M., Hopfen, C., Herbert, M. R., Schlotterer, C., Stern, D. L., Masly, J. P., McGregor, A. P. and Nunes, M. D. (2015). Genetic architecture and functional characterization of genes underlying the rapid diversification of male external genitalia between Drosophila simulans and Drosophila mauritiana. Genetics [Epub ahead of print]. PubMed ID: 25783699

    Arif, S., Kittelmann, S. and McGregor, A. P. (2015). From shavenbaby to the naked valley: trichome formation as a model for evolutionary developmental biology. Evol Dev 17: 120-126. PubMed ID: 25627718

    Hilbrant, M., Almudi, I., Leite, D. J., Kuncheria, L., Posnien, N., Nunes, M. and McGregor, A. P. (2014). Sexual dimorphism and natural variation within and among species in the Drosophila retinal mosaic. BMC Evol Biol 14: 240. PubMed ID: 25424626

    Arif, S., Murat, S., Almudi, I., Nunes, M. D., Bortolamiol-Becet, D., McGregor, N. S., Currie, J. M., Hughes, H., Ronshaugen, M., Sucena, E., Lai, E. C., Schlotterer, C., McGregor, A. P. (2013) Evolution of mir-92a Underlies Natural Morphological Variation in Drosophila melanogaster. Curr Biol. PubMed ID: 23453955

    Hilbrant, M., Damen, W. G. and McGregor, A. P. (2012). Evolutionary crossroads in developmental biology: the spider Parasteatoda tepidariorum. Development 139: 2655-2662. PubMed ID: 22782720

    Posnien, N., Hopfen, C., Hilbrant, M., Ramos-Womack, M., Murat, S., Schonauer, A., Herbert, S. L., Nunes, M. D., Arif, S., Breuker, C. J., Schlotterer, C., Mitteroecker, P. and McGregor, A. P. (2012). Evolution of eye morphology and rhodopsin expression in the Drosophila melanogaster species subgroup. PLoS One 7: e37346. PubMed ID: 22662147 >

  • Daniel McKay University of North Carolina at Chapel Hill
    Nystrom, S. L., Niederhuber, M. J. and McKay, D. J. (2020). Expression of E93 provides an instructive cue to control dynamic enhancer activity and chromatin accessibility during development. Development 147(6). PubMed ID: 32094114

    Uyehara, C. M. and McKay, D. J. (2019). Direct and widespread role for the nuclear receptor EcR in mediating the response to ecdysone in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 31019084

    Meers, M. P., Adelman, K., Duronio, R. J., Strahl, B. D., McKay, D. J. and Matera, A. G. (2018). Transcription start site profiling uncovers divergent transcription and enhancer-associated RNAs in Drosophila melanogaster. BMC Genomics 19(1): 157. PubMed ID: 29466941

    Uyehara, C. M., Nystrom, S. L., Niederhuber, M. J., Leatham-Jensen, M., Ma, Y., Buttitta, L. A. and McKay, D. J. (2017). Hormone-dependent control of developmental timing through regulation of chromatin accessibility. Genes Dev [Epub ahead of print]. PubMed ID: 28536147

    Pearson, J. C., McKay, D. J., Lieb, J. D. and Crews, S. T. (2016). Chromatin profiling of Drosophila CNS subpopulations identifies active transcriptional enhancers. Development 143(20): 3723-3732. PubMed ID: 27802137

    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 5(11): 1648-1661. PubMed ID: 27737823

  • Stephen McKechnie Faculty of Science, Monash University, Melbourne
    Cockerell, F. E., Sgro, C. M. and McKechnie, S. W. (2013). Latitudinal clines in heat tolerance, protein synthesis rate and transcript level of a candidate gene in Drosophila melanogaster. J Insect Physiol. PubMed ID: 24333150

    Hoffmann, A. A., Blacket, M. J., McKechnie, S. W., Rako, L., Schiffer, M., Rane, R. V., Good, R. T., Robin, C. and Lee, S. F. (2012). A proline repeat polymorphism of the Frost gene of Drosophila melanogaster showing clinal variation but not associated with cold resistance. Insect Mol Biol 21: 437-445. PubMed ID: 22708613

    Johnson, T. K., Cockerell, F. E. and McKechnie, S. W. (2011). Transcripts from the Drosophila heat-shock gene hsr-omega influence rates of protein synthesis but hardly affect resistance to heat knockdown. Mol Genet Genomics 285: 313-323. PubMed ID: 21399957

  • Bruce McKee Genome Science and Technology, University of Tennessee, Knoxville
    Krishnan, B., Thomas, S. E., Yan, R., Yamada, H., Zhulin, I. B. and McKee, B. D. (2014). Sisters Unbound Is Required for Meiotic Centromeric Cohesion in Drosophila melanogaster. Genetics. PubMed ID: 25194162

    Yan, R. and McKee, B. D. (2013). The cohesion protein SOLO associates with SMC1 and is required for synapsis, recombination, homolog bias and cohesion and pairing of centromeres in Drosophila Meiosis. PLoS Genet 9: e1003637. PubMed ID: 23874232

    McKee, B. D., Yan, R. and Tsai, J. H. (2012). Meiosis in male Drosophila. Spermatogenesis 2: 167-184. PubMed ID: 23087836

  • Kim McKim Waksman Institute of Microbiology, Rutgers
    Fellmeth, J. E., Sturm, H., Jang, J., Changela, N., Parikh, A., Persaud, M. and McKim, K. S. (2023). A Dynamic population of prophase CENP-C is required for meiotic chromosome segregation. bioRxiv. PubMed ID: 36993339

    Jang, J. K., Gladstein, A. C., Das, A., Shapiro, J. G., Sisco, Z. L. and McKim, K. S. (2021). Multiple pools of PP2A regulate spindle assembly, kinetochore attachments, and cohesion in Drosophila oocytes. J Cell Sci. PubMed ID: 34160620

    Wang, L. I., Das, A. and McKim, K. S. (2019). Sister centromere fusion during meiosis I depends on maintaining cohesins and destabilizing microtubule attachments. PLoS Genet 15(5): e1008072. PubMed ID: 31150390

    Das, A., Cesario, J., Hinman, A. M., Jang, J. K. and McKim, K. S. (2018). Kinesin 6 regulation in Drosophila female meiosis by the non-conserved N- and C-terminal domains. G3 (Bethesda). PubMed ID: 29514846

    Radford, S. J., Go, A. M. and McKim, K. S. (2016). Cooperation between kinesin motors promotes spindle symmetry and chromosome organization in Oocytes. Genetics [Epub ahead of print]. PubMed ID: 27932541

    Gyuricza, M. R., Manheimer, K. B., Apte, V., Krishnan, B., Joyce, E. F., McKee, B. D. and McKim, K. S. (2016). Dynamic and stable cohesins regulate synaptonemal complex assembly and chromosome segregation. Curr Biol [Epub ahead of print]. PubMed ID: 27291057

    Das, A., Shah, S. J., Fan, B., Paik, D., DiSanto, D. J., Hinman, A. M., Cesario, J. M., Battaglia, R. A., Demos, N. and McKim, K. S. (2015). Spindle assembly and chromosome segregation requires central spindle proteins in Drosophila oocytes. Genetics. PubMed ID: 26564158

    Radford, S. J., Hoang, T. L., Gluszek, A. A., Ohkura, H. and McKim, K. S. (2015). Lateral and end-on kinetochore attachments are coordinated to achieve bi-orientation in Drosophila oocytes. PLoS Genet 11: e1005605. PubMed ID: 26473960

    Radford, S. J., Harrison, A. M. and McKim, K. S. (2012). Microtubule-depolymerizing kinesin KLP10A restricts the length of the acentrosomal meiotic spindle in Drosophila females. Genetics 192: 431-440. PubMed ID: 22865737

    Radford, S. J., Harrison, A. M. and McKim, K. S. (2012). Microtubule-depolymerizing kinesin KLP10A restricts the length of the acentrosomal meiotic spindle in Drosophila females. Genetics 192: 431-440. PubMed ID: 22865737

  • Helen McNeill Mount Sinai Hospital
    Webb, J. L., Moe, S. M., Bolstad, A. K. and McNeill, E. M. (2021). Identification of conserved transcriptome features between humans and Drosophila in the aging brain utilizing machine learning on combined data from the NIH Sequence Read Archive. PLoS One 16(8): e0255085. PubMed ID: 34379632

    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

    Yeung, K., Boija, A., Karlsson, E., Holmqvist, P. H., Tsatskis, Y., Nisoli, I., Yap, D. B., Lorzadeh, A., Moksa, M., Hirst, M., Aparicio, S., Fanto, M., Stenberg, P., Mannervik, M. and McNeill, H. (2017). Atrophin controls developmental signaling pathways via interactions with Trithorax-like. Elife 6. PubMed ID: 28327288

    Kwan, J., Sczaniecka, A., Arash, E. H., Nguyen, L., Chen, C. C., Ratkovic, S., Klezovitch, O., Attisano, L., McNeill, H., Emili, A. and Vasioukhin, V. (2016). DLG5 connects cell polarity and Hippo signaling protein networks by linking PAR-1 with MST1/2. Genes Dev 30(24): 2696-2709. PubMed ID: 28087714

    Delanoue, R., Meschi, E., Agrawal, N., Mauri, A., Tsatskis, Y., McNeill, H. and Láopold, P. (2016). Drosophila insulin release is triggered by adipose Stunted ligand to brain Methuselah receptor. Science 353: 1553-1556. PubMed ID: 27708106

    Chang, Y. J., Zhou, L., Binari, R., Manoukian, A., Mak, T., McNeill, H. and Stambolic, V. (2016). The Rho guanine nucleotide exchange factor DRhoGEF2 is a genetic modifier of the PI3K pathway in Drosophila. PLoS One 11: e0152259. PubMed ID: 27015411

    Payankaulam, S., Yeung, K., McNeill, H., Henry, R.W. and Arnosti, D.N. (2016). Regulation of cell polarity determinants by the Retinoblastoma tumor suppressor protein. Sci Rep 6: 22879. PubMed ID: 26971715

    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

    Sing, A., Tsatskis, Y., Fabian, L., Hester, I., Rosenfeld, R., Serricchio, M., Yau, N., Bietenhader, M., Shanbhag, R., Jurisicova, A., Brill, J. A., McQuibban, G. A. and McNeill, H. (2014). The atypical cadherin fat directly regulates mitochondrial function and metabolic state. Cell 158: 1293-1308. PubMed ID: 25215488

    Enderle, L. and McNeill, H. (2013). Hippo gains weight: added insights and complexity to pathway control. Sci Signal 6: re7. PubMed ID: 24106343

  • James McNew Department of Biochemistry & Cell Biology, Rice University, Houston
    Betancourt-Solis, M. A. and McNew, J. A. (2019). Detergent-assisted Reconstitution of Recombinant Drosophila Atlastin into Liposomes for Lipid-mixing Assays. J Vis Exp(149). PubMed ID: 31329173

    Xu, S., Stern, M. and McNew, J. A. (2016). Beneficial effects of rapamycin in a Drosophila model for hereditary spastic paraplegia. J Cell Sci [Epub ahead of print]. PubMed ID: 27909242

    Summerville, J., Faust, J., Fan, E., Pendin, D., Daga, A., Formella, J., Stern, M. and McNew, J. A. (2016). The effects of ER morphology on synaptic structure and function in Drosophila melanogaster. J Cell Sci. PubMed ID: 26906425

    Faust, J. E., Manisundaram, A., Ivanova, P. T., Milne, S. B., Summerville, J. B., Brown, H. A., Wangler, M., Stern, M. and McNew, J. A. (2014). Peroxisomes Are Required for Lipid Metabolism and Muscle Function in Drosophila melanogaster. PLoS One 9: e100213. PubMed ID: 24945818

    Faust, J. E., Verma, A., Peng, C. and McNew, J. A. (2012). An inventory of peroxisomal proteins and pathways in Drosophila melanogaster. Traffic 13: 1378-1392. PubMed ID: 22758915

  • Mitch McVey Department of Biology, Tufts University, Medford, MA
    Jajoo, A., Donlon, C., Shnayder, S., Levin, M. and McVey, M. (2020). Sertraline induces DNA damage and cellular toxicity in Drosophila that can be ameliorated by antioxidants. Sci Rep 10(1): 4512. PubMed ID: 32161356

    Kocak, E., Dykstra, S., Nemeth, A., Coughlin, C. G., Rodgers, K. and McVey, M. (2019). The Drosophila melanogaster PIF1 helicase promotes survival during replication stress and processive DNA synthesis during double-strand gap repair. Genetics. PubMed ID: 31537623

    Cassidy, D., Epiney, D., Salameh, C., Zhou, L. T., Salomon, R. N., Schirmer, A. E., McVey, M. and Bolterstein, E. (2019). Evidence for premature aging in a Drosophila model of Werner syndrome. Exp Gerontol: 110733. PubMed ID: 31518666

    Khodaverdian, V. Y., Hanscom, T., Yu, A. M., Yu, T. L., Mak, V., Brown, A. J., Roberts, S. A. and McVey, M. (2017). Secondary structure forming sequences drive SD-MMEJ repair of DNA double-strand breaks. Nucleic Acids Res. PubMed ID: 29121353

    Alexander, J. L., Beagan, K., Orr-Weaver, T. L. and McVey, M. (2016). Multiple mechanisms contribute to double-strand break repair at rereplication forks in Drosophila follicle cells. Proc Natl Acad Sci U S A. [Epub ahead of print] PubMed ID: 27849606

    Bolterstein, E., Rivero, R., Marquez, M. and McVey, M. (2014). The Drosophila Werner Exonuclease Participates in an Exonuclease-Independent Response to Replication Stress. Genetics. PubMed ID: 24709634

    Thomas, A. M., Hui, C., South, A. and McVey, M. (2013). Common variants of Drosophila melanogaster Cyp6d2 cause camptothecin sensitivity and synergize with loss of Brca2. G3 (Bethesda) 3: 91-99. PubMed ID: 23316441

    Kane, D. P., Shusterman, M., Rong, Y. and McVey, M. (2012). Competition between replicative and translesion polymerases during homologous recombination repair in Drosophila. PLoS Genet 8: e1002659. PubMed ID: 22532806

  • Bernard M Mechler Institute of Cellular Biology, Charles University in Prague
    Farkas, R., Benova-Liszekova, D., Mentelova, L., Mahmood, S., Datkova, Z., Beno, M., Pecenova, L., Raska, O., Smigova, J., Chase, B. A., Raska, I. and Mechler, B. M. (2015). Vacuole dynamics in the salivary glands of Drosophila melanogaster during prepupal development. Dev Growth Differ 57: 74-96. PubMed ID: 25611296

    Farkas, R., Datkova, Z., Mentelova, L., Low, P., Benova-Liszekova, D., Beno, M., Sass, M., Rehulka, P., Rehulkova, H., Raska, O., Kovacik, L., Smigova, J., Raska, I. and Mechler, B. M. (2014). Apocrine secretion in Drosophila salivary glands: subcellular origin, dynamics, and identification of secretory proteins. PLoS One 9: e94383. PubMed ID: 24732043

    Benbahouche Nel, H., Iliopoulos, I., Torok, I., Marhold, J., Henri, J., Kajava, A. V., Farkas, R., Kempf, T., Schnolzer, M., Meyer, P., Kiss, I., Bertrand, E., Mechler, B. M. and Pradet-Balade, B. (2014). Drosophila Spag is the homolog of RNA polymerase II-associated protein 3 (RPAP3) and recruits the heat shock proteins 70 and 90 (Hsp70 and Hsp90) during the assembly of cellular machineries. J Biol Chem 289: 6236-6247. PubMed ID: 24394412

  • Adam Megighian Department of Biomedical Science, University of Padua
    Frighetto, G., Zordan, M. A., Castiello, U. and Megighian, A. (2019). Action-based attention in Drosophila melanogaster. J Neurophysiol. PubMed ID: 31042449

    Zornetta, I., Scorzeto, M., Mendes Dos Reis, P. V., De Lima, M. E., Montecucco, C., Megighian, A. and Rossetto, O. (2017). Electrophysiological Characterization of the Antarease Metalloprotease from Tityus serrulatus Venom. Toxins (Basel) 9(3). PubMed ID: 28264432

    Romano, G., Appocher, C., Scorzeto, M., Klima, R., Baralle, F. E., Megighian, A. and Feiguin, F. (2015). Glial TDP-43 regulates axon wrapping, GluRIIA clustering and fly motility by autonomous and non-autonomous mechanisms. Hum Mol Genet 24(21): 6134-6145. PubMed ID: 26276811

  • Timothy Megraw Florida State University College of Medicine, Tallahassee
    Debec, A., Loppin, B., Zheng, C., Liu, X. and Megraw, T. L. (2020). The Enigma of Centriole Loss in the 1182-4 Cell Line. Cells 9(5). PubMed ID: 32456186

    Zheng, Y., Buchwalter, R. A., Zheng, C., Wight, E. M., Chen, J. V. and Megraw, T. L. (2020). A perinuclear microtubule-organizing centre controls nuclear positioning and basement membrane secretion. Nat Cell Biol 22(3): 297-309. PubMed ID: 32066907

    Tillery, M. M. L., Blake-Hedges, C., Zheng, Y., Buchwalter, R. A. and Megraw, T. L. (2018). Centrosomal and Non-Centrosomal Microtubule-Organizing Centers (MTOCs) in Drosophila melanogaster. Cells 7(9). PubMed ID: 30154378

    Chen, J. V., Buchwalter, R. A., Kao, L. R. and Megraw, T. L. (2017). A Splice Variant of Centrosomin Converts Mitochondria to Microtubule-Organizing Centers. Curr Biol 27(13): 1928-1940 e1926. PubMed ID: 28669756

    Zheng, Y., Mennella, V., Marks, S., Wildonger, J., Elnagdi, E., Agard, D. and Megraw, T. L. (2016). The Seckel syndrome and centrosomal protein Ninein localizes asymmetrically to stem cell centrosomes, but is not required for normal development, behavior, or DNA damage response in Drosophila. Mol Biol Cell. PubMed ID: 27053665

    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

    Chen, J. V. and Megraw, T. L. (2014). Spermitin: a novel mitochondrial protein in Drosophila spermatids. PLoS One 9: e108802. PubMed ID: 25265054

    Riparbelli, M. G., Cabrera, O. A., Callaini, G. and Megraw, T. L. (2013). Unique properties of Drosophila spermatocyte primary cilia. Biol Open 2: 1137-1147. PubMed ID: 24244850

    Riparbelli, M. G., Callaini, G. and Megraw, T. L. (2012). Assembly and persistence of primary cilia in dividing Drosophila spermatocytes. Dev Cell 23: 425-432. PubMed ID: 22898783

    Kao, L. R. and Megraw, T. L. (2009). Centrocortin cooperates with centrosomin to organize Drosophila embryonic cleavage furrows. Curr Biol 19: 937-942. PubMed ID: 19427213

  • Carine Meignin Institut de Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France
    Donelick, H. M., Talide, L., Bellet, M., Aruscavage, J., Lauret, E., Aguiar, E., Marques, J. T., Meignin, C. and Bass, B. L. (2020). In vitro studies provide insight into effects of Dicer-2 helicase mutations in Drosophila melanogaster. RNA. PubMed ID: 32843367

    Kuhn, L., Majzoub, K., Einhorn, E., Chicher, J., Pompon, J., Imler, J. L., Hammann, P. and Meignin, C. (2017). Definition of a RACK1 Interaction Network in Drosophila melanogaster Using SWATH-MS. G3 (Bethesda). PubMed ID: 28522639

    Lamiable, O., Meignin, C. and Imler, J. L. (2016). WntD and Diedel: Two immunomodulatory cytokines in Drosophila immunity. Fly (Austin) 10(4): 187-194. PubMed ID: 27314646

    Lamiable, O., Arnold, J., de Faria, I. J., Olmo, R. P., Bergami, F., Meignin, C., Hoffmann, J. A., Marques, J. T. and Imler, J. L. (2016). Analysis of the Contribution of Hemocytes and Autophagy to Drosophila Antiviral Immunity. J Virol 90(11): 5415-5426. PubMed ID: 27009948

  • Ian Meinertzhagen Laboratory of Invertebrate Biology, Dahlhouse University, Halifax
    Justs, K. A., Lu, Z., Chouhan, A. K., Borycz, J. A., Lu, Z., Meinertzhagen, I. A. and Macleod, G. T. (2021). Presynaptic Mitochondrial Volume and Packing Density Scale with Presynaptic Power Demand. J Neurosci. PubMed ID: 34907026

    Stawarski, M., Hernandez, R. X., Feghhi, T., Borycz, J. A., Lu, Z., Agarwal, A., Reihl, K., Tavora, R., Lau, A. W. C., Meinertzhagen, I. A., Renden, R. and Macleod, G. T. (2020). Neuronal glutamatergic synaptic clefts alkalinize rather than acidify during neurotransmission. J Neurosci. PubMed ID: 31964719

    Shinomiya, K., Horne, J. A., McLin, S., Wiederman, M., Nern, A., Plaza, S. M. and Meinertzhagen, I. A. (2019). The organization of the second optic chiasm of the Drosophila optic lobe. Front Neural Circuits 13: 65. PubMed ID: 31680879

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

    Tsai, J. W., Kostyleva, R., Chen, P. L., Rivas-Serna, I. M., Clandinin, M. T., Meinertzhagen, I. A. and Clandinin, T. R. (2019). Transcriptional feedback links lipid synthesis to synaptic vesicle pools in Drosophila photoreceptors. Neuron. PubMed ID: 30737130

    Shinomiya, K., Huang, G., Lu, Z., Parag, T., Xu, C. S., Aniceto, R., Ansari, N., Cheatham, N., Lauchie, S., Neace, E., Ogundeyi, O., Ordish, C., Peel, D., Shinomiya, A., Smith, C., Takemura, S., Talebi, I., Rivlin, P. K., Nern, A., Scheffer, L. K., Plaza, S. M. and Meinertzhagen, I. A. (2019). Comparisons between the ON- and OFF-edge motion pathways in the Drosophila brain. Elife 8. PubMed ID: 30624205

    Horne, J. A., Langille, C., McLin, S., Wiederman, M., Lu, Z., Xu, C. S., Plaza, S. M., Scheffer, L. K., Hess, H. F. and Meinertzhagen, I. A. (2018). A resource for the Drosophila antennal lobe provided by the connectome of glomerulus VA1v. Elife 7. PubMed ID: 30382940

    Borycz, J., Ziegler, A., Borycz, J. A., Uhlenbrock, G., Tapken, D., Caceres, L., Hollmann, M., Hovemann, B. T. and Meinertzhagen, I. A. (2018). Location and functions of Inebriated in the Drosophila eye. Biol Open 7(7). PubMed ID: 30037884

    Astigarraga, S., Douthit, J., Tarnogorska, D., Creamer, M. S., Mano, O., Clark, D. A., Meinertzhagen, I. A. and Treisman, J. E. (2018). Drosophila Sidekick is required in developing photoreceptors to enable visual motion detection. Development. PubMed ID: 29361567

    Takemura, S. Y., Nern, A., Chklovskii, D. B., Scheffer, L. K., Rubin, G. M. and Meinertzhagen, I. A. (2017). The comprehensive connectome of a neural substrate for 'ON' motion detection in Drosophila. Elife 6 [Epub ahead of print]. PubMed ID: 28432786

    Lu, Z., Chouhan, A. K., Borycz, J. A., Lu, Z., Rossano, A. J., Brain, K. L., Zhou, Y., Meinertzhagen, I. A. and Macleod, G. T. (2016). High-probability neurotransmitter release sites represent an energy-efficient design. Curr Biol 26: 2562-2571. PubMed ID: 27593375

    Lin, T. Y., Luo, J., Shinomiya, K., Ting, C. Y., Lu, Z., Meinertzhagen, I. A. and Lee, C. H. (2016). Mapping chromatic pathways in the Drosophila visual system. J Comp Neurol 524: Spc1. PubMed ID: 26648045

  • Pascal Meier Institute of Cancer Research: Royal Cancer Hospital, London
    Morris, O., Liu, X., Domingues, C., Runchel, C., Chai, A., Basith, S., Tenev, T., Chen, H., Choi, S., Pennetta, G., Buchon, N. and Meier, P. (2016). Signal integration by the IkappaB protein Pickle shapes Drosophila innate host defense. Cell Host Microbe 20: 283-295. PubMed ID: 27631699

    Orme, M. H., et al. (2016). The unconventional myosin CRINKLED and its mammalian orthologue MYO7A regulate caspases in their signalling roles. Nat Commun 7: 10972. PubMed ID: 26960254

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

    Saez, L., Derasmo, M., Meyer, P., Stieglitz, J. and Young, M. W. (2011). A key temporal delay in the circadian cycle of Drosophila is mediated by a nuclear localization signal in the timeless protein. Genetics 188: 591-600. PubMed ID: 21515571

  • Colin Meiklejohn School of Biological Sciences, University of Nebraska, Lincoln
    Lemos, B., Brano, A. T., Jiang, P. P., Hartl, D. L. and Meiklejohn, C. D. (2013). Genome-Wide Gene Expression Effects of Sex Chromosome Imprinting in Drosophila. G3 (Bethesda). PubMed ID: 24318925

    Meiklejohn, C. D., Holmbeck, M. A., Siddiq, M. A., Abt, D. N., Rand, D. M. and Montooth, K. L. (2013). An Incompatibility between a mitochondrial tRNA and its nuclear-encoded tRNA synthetase compromises development and fitness in Drosophila. PLoS Genet 9: e1003238. PubMed ID: 23382693

    Meiklejohn, C. D. and Presgraves, D. C. (2012). Little evidence for demasculinization of the Drosophila X chromosome among genes expressed in the male germline. Genome Biol Evol 4: 1007-1016. PubMed ID: 22975718

    Meiklejohn, C. D., Landeen, E. L., Cook, J. M., Kingan, S. B. and Presgraves, D. C. (2011). Sex chromosome-specific regulation in the Drosophila male germline but little evidence for chromosomal dosage compensation or meiotic inactivation. PLoS Biol 9: e1001126. PubMed ID: 21857805

  • Victoria Meller Biological Sciences, Wayne State University, Detroit
    Deshpande, N. and Meller, V. H. (2018). Chromatin that guides dosage compensation is modulated by the siRNA pathway in Drosophila melanogaster. Genetics. PubMed ID: 29921620

    Joshi, S. S. and Meller, V. H. (2017). Satellite repeats identify X chromatin for dosage compensation in Drosophila melanogaster males. Curr Biol 27(10): 1393-1402.e1392. PubMed ID: 28457869

    Koya, S. K. and Meller, V. H. (2015). Modulation of heterochromatin by male specific lethal proteins and roX RNA in Drosophila melanogaster males. PLoS One 10: e0140259. PubMed ID: 26468879

    Menon, D. U. and Meller, V. H. (2015). Identification of the Drosophila X chromosome: The long and short of it. RNA Biol: 1-6. PubMed ID: 26367502

    Apte, M.S. and Meller, V.H. (2015). Sex differences in Drosophila melanogaster heterochromatin are regulated by non-sex specific factors. PLoS One 10: e0128114. PubMed ID: 26053165

    Menon, D. U., Coarfa, C., Xiao, W., Gunaratne, P. H. and Meller, V. H. (2014). siRNAs from an X-linked satellite repeat promote X-chromosome recognition in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 25368194

    Joshi, S. S., Cheong, H. and Meller, V. H. (2014). A strategy for generation and balancing of autosome: Y chromosome translocations. Fly (Austin) 8: 58-62. PubMed ID: 24646827

    Apte, M. S., Moran, V. A., Menon, D. U., Rattner, B. P., Barry, K. H., Zunder, R. M., Kelley, R. and Meller, V. H. (2013). Generation of a Useful roX1 Allele by Targeted Gene Conversion. G3 (Bethesda). PubMed ID: 24281425

    Menon, D. U. and Meller, V. H. (2012). A role for siRNA in X-chromosome dosage compensation in Drosophila melanogaster. Genetics 191: 1023-1028. PubMed ID: 22554892

    Koya, S. K. and Meller, V. H. (2011). roX RNAs and Genome Regulation in Drosophila Melanogaster. Prog Mol Subcell Biol 51: 147-160. PubMed ID: 21287137

  • Barbara Mellone Molecular & Cell Biology Department, University of Connecticut, Storrs
    Palladino, J., Chavan, A., Sposato, A., Mason, T. D. and Mellone, B. G. (2020). Targeted De Novo Centromere Formation in Drosophila Reveals Plasticity and Maintenance Potential of CENP-A Chromatin. Dev Cell 52(3): 379-394. PubMed ID: 32049040

    Chang, C. H., Chavan, A., Palladino, J., Wei, X., Martins, N. M. C., Santinello, B., Chen, C. C., Erceg, J., Beliveau, B. J., Wu, C. T., Larracuente, A. M. and Mellone, B. G. (2019). Islands of retroelements are major components of Drosophila centromeres. PLoS Biol 17(5): e3000241. PubMed ID: 31086362

    Rosin, L. and Mellone, B.G. (2016). Co-evolving CENP-A and CAL1 domains mediate centromeric CENP-A deposition across Drosophila species. Dev Cell 37: 136-147. PubMed ID: 27093083

    Chen, C. C., Dechassa, M. L., Bettini, E., Ledoux, M. B., Belisario, C., Heun, P., Luger, K. and Mellone, B. G. (2014). CAL1 is the Drosophila CENP-A assembly factor. J Cell Biol. PubMed ID: 24469636

    Ross, B. D., Rosin, L., Thomae, A. W., Hiatt, M. A., Vermaak, D., de la Cruz, A. F., Imhof, A., Mellone, B. G. and Malik, H. S. (2013). Stepwise evolution of essential centromere function in a Drosophila neogene. Science 340: 1211-1214. PubMed ID: 23744945

    Chen, C. C., Greene, E., Bowers, S. R. and Mellone, B. G. (2012). A role for the CAL1-partner Modulo in centromere integrity and accurate chromosome segregation in Drosophila. PLoS One 7: e45094. PubMed ID: 23028777

  • Samir Merabet Institut de Génomique Fonctionnelle de Lyon
    Naville, M. and Merabet, S. (2021). In-Depth Annotation of the Drosophila Bithorax-Complex Reveals the Presence of Several Alternative ORFs That Could Encode for Motif-Rich Peptides. Cells 10(11). PubMed ID: 34831206

    Duffraisse, M., Paul, R., Carnesecchi, J., Hudry, B., Banreti, A., Reboulet, J., Ajuria, L., Lohmann, I. and Merabet, S. (2020). Role of a versatile peptide motif controlling Hox nuclear export and autophagy in the Drosophila fat body. J Cell Sci 133(18). PubMed ID: 32878938

    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

    Baeza, M., Viala, S., Heim, M., Dard, A., Hudry, B., Duffraisse, M., Rogulja-Ortmann, A., Brun, C. and Merabet, S. (2015). Inhibitory activities of short linear motifs underlie Hox interactome specificity in vivo. Elife 4. PubMed ID: 25869471

    Boube, M., Hudry, B., Immarigeon, C., Carrier, Y., Bernat-Fabre, S., Merabet, S., Graba, Y., Bourbon, H. M. and Cribbs, D. L. (2014). Drosophila melanogaster Hox transcription factors access the RNA polymerase II machinery through direct homeodomain binding to a conserved motif of mediator subunit Med19. PLoS Genet 10: e1004303. PubMed ID: 24786462

    Sambrani, N., Hudry, B., Maurel-Zaffran, C., Zouaz, A., Mishra, R., Merabet, S. and Graba, Y. (2013). Distinct molecular strategies for Hox-mediated limb suppression in Drosophila: from cooperativity to dispensability/antagonism in TALE partnership. PLoS Genet 9: e1003307. PubMed ID: 23505377

  • Thomas Merritt Genomics and Bioinformatics, Laurentian University, Sudbury, ON
    Doran, M. L., Knee, J. M., Wang, N., Rzezniczak, T. Z., Parkes, T. L., Li, L. and Merritt, T. J. S. (2017). Metabolomic analysis of oxidative stress: Superoxide dismutase mutation and paraquat induced stress in Drosophila melanogaster. Free Radic Biol Med 113: 323-334. PubMed ID: 29031835

    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). PubMed ID: 25213691

    Rzezniczak, T. Z., Lum, T. E., Harniman, R. and Merritt, T. J. (2012). A combination of structural and cis-regulatory factors drives biochemical differences in Drosophila melanogaster malic enzyme. Biochem Genet 50: 823-837. PubMed ID: 22733181

    Rzezniczak, T. Z., Douglas, L. A., Watterson, J. H. and Merritt, T. J. (2011). Paraquat administration in Drosophila for use in metabolic studies of oxidative stress. Anal Biochem 419: 345-347. PubMed ID: 21910964

    Bernard, K. E., Parkes, T. L. and Merritt, T. J. (2011). A model of oxidative stress management: moderation of carbohydrate metabolizing enzymes in SOD1-null Drosophila melanogaster. PLoS One 6: e24518. PubMed ID: 21909438

    Lum, T. E. and Merritt, T. J. (2011). Nonclassical regulation of transcription: interchromosomal interactions at the malic enzyme locus of Drosophila melanogaster. Genetics 189: 837-849. PubMed ID: 21900270

  • Frederic Mery CNRS, Gif sur Yvette
    Dawson, E. H., Bailly, T. P. M., Dos Santos, J., Moreno, C., Devilliers, M., Maroni, B., Sueur, C., Casali, A., Ujvari, B., Thomas, F., Montagne, J. and Mery, F. (2018). Social environment mediates cancer progression in Drosophila. Nat Commun 9(1): 3574. PubMed ID: 30177703

    Teseo, S., Veerus, L. and Mery, F. (2016). Fighting experience affects fruit fly behavior in a mating context. Naturwissenschaften 103: 38. PubMed ID: 27108453

    Philippe, A.S., Jeanson, R., Pasquaretta, C., Rebaudo, F., Sueur, C. and Mery, F. (2016). Genetic variation in aggregation behaviour and interacting phenotypes in Drosophila. Proc Biol Sci 283 (1827). PubMed ID: 27009219

    Pasquaretta, C., Battesti, M., Klenschi, E., Bousquet, C.A., Sueur, C. and Mery, F. (2016). How social network structure affects decision-making in Drosophila melanogaster. Proc Biol Sci 283(1826). PubMed ID: 26936247

    Teseo, S., Veerus, L., Moreno, C. and Mery, F. (2016). Sexual harassment induces a temporary fitness cost but does not constrain the acquisition of environmental information in fruit flies. Biol Lett 12. PubMed ID: 26763219

    Foucaud, J., Philippe, A. S., Moreno, C. and Mery, F. (2013). A genetic polymorphism affecting reliance on personal versus public information in a spatial learning task in Drosophila melanogaster. Proc Biol Sci 280: 20130588. PubMed ID: 23576793

    Burns, J. G., Svetec, N., Rowe, L., Mery, F., Dolan, M. J., Boyce, W. T. and Sokolowski, M. B. (2012). Gene-environment interplay in Drosophila melanogaster: chronic food deprivation in early life affects adult exploratory and fitness traits. Proc Natl Acad Sci U S A 109 Suppl 2: 17239-17244. PubMed ID: 23045644

  • Mark Metzstein Department of Human Genetics, University of Utah
    Nelson, J. O., Forster, D., Frizzell, K. A., Luschnig, S. and Metzstein, M. M. (2018). Multiple nonsense-mediated mRNA processes require Smg5 in Drosophila. Genetics. PubMed ID: 29903866

    Nikolova, L. S. and Metzstein, M. M. (2015). Intracellular lumen formation in Drosophila proceeds via a novel subcellular compartment. Development [Epub ahead of print]. PubMed ID: 26428009

    Jones, T. A., Nikolova, L. S., Schjelderup, A. and Metzstein, M. M. (2014). Exocyst-mediated membrane trafficking is required for branch outgrowth in Drosophila tracheal terminal cells. Dev Biol. PubMed ID: 24607370

    Chapin, A., Hu, H., Rynearson, S. G., Hollien, J., Yandell, M. and Metzstein, M. M. (2014). In Vivo Determination of Direct Targets of the Nonsense Mediated Decay Pathway in Drosophila. G3 (Bethesda). PubMed ID: 24429422

    Jones, T. A. and Metzstein, M. M. (2013). Examination of Drosophila larval tracheal terminal cells by light microscopy. J Vis Exp. PubMed ID: 23892488

    Ruiz, O. E., Nikolova, L. S. and Metzstein, M. M. (2012). Drosophila Zpr1 (Zinc finger protein 1) is required downstream of both EGFR and FGFR signaling in tracheal subcellular lumen formation. PLoS One 7: e45649. PubMed ID: 23029159

    Frizzell, K. A., Rynearson, S. G. and Metzstein, M. M. (2012). Drosophila mutants show NMD pathway activity is reduced, but not eliminated, in the absence of Smg6. RNA 18: 1475-1486. PubMed ID: 22740637

    Jones, T. A. and Metzstein, M. M. (2011). A novel function for the PAR complex in subcellular morphogenesis of tracheal terminal cells in Drosophila melanogaster. Genetics 189: 153-164. PubMed ID: 21750259

  • Craig Micchelli Department of Developmental Biology, Washington University School of Medicine, St. Louis
    Beebe, K., Park, D., Taghert, P. H. and Micchelli, C. A. (2015). The Drosophila pro-secretory transcription factor dimmed is dynamically regulated in adult enteroendocrine cells and protects against gram-negative infection. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25999585

    Beehler-Evans, R. and Micchelli, C. A. (2015). Generation of enteroendocrine cell diversity in midgut stem cell lineages. Development 142: 654-664. PubMed ID: 25670792

    Micchelli, C. A. (2014). Whole-mount immunostaining of the adult Drosophila gastrointestinal tract. Methods. PubMed ID: 24680702

    Strand, M. and Micchelli, C. A. (2013). Regional Control of Drosophila Gut Stem Cell Proliferation: EGF Establishes GSSC Proliferative Set Point & Controls Emergence from Quiescence. PLoS One 8: e80608. PubMed ID: 24236188

    Lee, W. C. and Micchelli, C. A. (2013). Development and characterization of a chemically defined food for Drosophila. PLoS One 8: e67308. PubMed ID: 23844001

    Micchelli, C. A. (2012). The origin of intestinal stem cells in Drosophila. Dev Dyn 241: 85-91. PubMed ID: 21972080

    Strand, M. and Micchelli, C. A. (2011). Quiescent gastric stem cells maintain the adult Drosophila stomach. Proc Natl Acad Sci U S A 108: 17696-17701. PubMed ID: 21984734

    Micchelli, C. A., Sudmeier, L., Perrimon, N., Tang, S. and Beehler-Evans, R. (2011). Identification of adult midgut precursors in Drosophila. Gene Expr Patterns 11: 12-21. PubMed ID: 20804858

  • Pawel Michalak Biocomplexity Institute, Virginia Tech, Blacksburg, VA
    Kang, L., Garner, H. R., Price, D. K. and Michalak, P. (2017). A test for gene flow among sympatric and allopatric Hawaiian picture-winged Drosophila. J Mol Evol. PubMed ID: 28492967

    Kang, L., Aggarwal, D. D., Rashkovetsky, E., Korol, A. B. and Michalak, P. (2016). Rapid genomic changes in Drosophila melanogaster adapting to desiccation stress in an experimental evolution system. BMC Genomics 17: 233. PubMed ID: 26979755

    Kim, Y. B., Oh, J. H., McIver, L. J., Rashkovetsky, E., Michalak, K., Garner, H. R., Kang, L., Nevo, E., Korol, A. B. and Michalak, P. (2014). Divergence of Drosophila melanogaster repeatomes in response to a sharp microclimate contrast in Evolution Canyon, Israel. Proc Natl Acad Sci U S A. PubMed ID: 25006263

    Hubner, S., Rashkovetsky, E., Kim, Y. B., Oh, J. H., Michalak, K., Weiner, D., Korol, A. B., Nevo, E. and Michalak, P. (2013). Genome differentiation of Drosophila melanogaster from a microclimate contrast in Evolution Canyon, Israel. Proc Natl Acad Sci U S A 110: 21059-21064. PubMed ID: 24324170

    Ma, D. and Michalak, P. (2011). Ephemeral association between gene CG5762 and hybrid male sterility in Drosophila sibling species. J Mol Evol 73: 181-187. PubMed ID: 22052252

  • Gero Miesenbock Department of Physiology, Anatomy & Genetics, University of Oxford
    Vrontou, E., Groschner, L. N., Szydlowski, S., Brain, R., Krebbers, A. and Miesenbock, G. (2021). Response competition between neurons and antineurons in the mushroom body. Curr Biol. PubMed ID: 34610272

    Harrell, E. R., Pimentel, D. and Miesenbock, G. (2021). Changes in presynaptic gene expression during homeostatic compensation at a central synapse. J Neurosci. PubMed ID: 33608385

    Groschner, L. N., Chan Wah Hak, L., Bogacz, R., DasGupta, S. and Miesenbock, G. (2018). Dendritic Integration of Sensory Evidence in Perceptual Decision-Making. Cell 173(4): 894-905.e813. PubMed ID: 29706545

    Donlea, J. M., Pimentel, D., Talbot, C. B., Kempf, A., Omoto, J. J., Hartenstein, V. and Miesenbock, G. (2018). Recurrent circuitry for balancing sleep need and sleep. Neuron 97(2): 378-389.e374. PubMed ID:
    29307711

    Pimentel, D., Donlea, J.M., Talbot, C.B., Song, S.M., Thurston, A.J. and Miesenböck, G. (2016). Operation of a homeostatic sleep switch. Nature [Epub ahead of print]. PubMed ID: 27487216

    DasGupta, S., Ferreira, C. H. and Miesenbock, G. (2014). FoxP influences the speed and accuracy of a perceptual decision in Drosophila. Science 344: 901-904. PubMed ID: 24855268

    DasGupta, S., Ferreira, C. H. and Miesenbock, G. (2014). FoxP influences the speed and accuracy of a perceptual decision in Drosophila. Science 344: 901-904. PubMed ID: 24855268

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

    Lin, A. C., Bygrave, A. M., de Calignon, A., Lee, T. and Miesenbock, G. (2014). Sparse, decorrelated odor coding in the mushroom body enhances learned odor discrimination. Nat Neurosci. PubMed ID: 24561998

    Parnas, M., Lin, A. C., Huetteroth, W. and Miesenbock, G. (2013). Odor discrimination in Drosophila: from neural population codes to behavior. Neuron 79: 932-944. PubMed ID: 24012006

  • Irene Miguel-Aliaga Gut Signalling and Metabolism Group, Imperial College London
    Millington, J. W., Brownrigg, G. P., Chao, C., Sun, Z., Basner-Collins, P. J., Wat, L. W., Hudry, B., Miguel-Aliaga, I. and Rideout, E. J. (2021). Female-biased upregulation of insulin pathway activity mediates the sex difference in Drosophila body size plasticity. Elife 10. PubMed ID: 33448263

    Messal, H. A., Almagro, J., Zaw Thin, M., Tedeschi, A., Ciccarelli, A., Blackie, L., Anderson, K. I., Miguel-Aliaga, I., van Rheenen, J. and Behrens, A. (2021). Antigen retrieval and clearing for whole-organ immunofluorescence by FLASH. Nat Protoc 16(1): 239-262. PubMed ID: 33247285

    Hadjieconomou, D., King, G., Gaspar, P., Mineo, A., Blackie, L., Ameku, T., Studd, C., de Mendoza, A., Diao, F., White, B. H., Brown, A. E. X., Placais, P. Y., Preat, T. and Miguel-Aliaga, I. (2020). Enteric neurons increase maternal food intake during reproduction. Nature 587(7834): 455-459. PubMed ID: 33116314

    Hudry, B., de Goeij, E., Mineo, A., Gaspar, P., Hadjieconomou, D., Studd, C., Mokochinski, J. B., Kramer, H. B., Placais, P. Y., Preat, T. and Miguel-Aliaga, I. (2019). Sex Differences in intestinal carbohydrate metabolism promote food intake and sperm maturation. Cell 178(4): 901-918. PubMed ID: 31398343

    Perea, D., Guiu, J., Hudry, B., Konstantinidou, C., Milona, A., Hadjieconomou, D., Carroll, T., Hoyer, N., Natarajan, D., Kallijarvi, J., Walker, J. A., Soba, P., Thapar, N., Burns, A. J., Jensen, K. B. and Miguel-Aliaga, I. (2017). Ret receptor tyrosine kinase sustains proliferation and tissue maturation in intestinal epithelia. Embo J. PubMed ID: 28899900

    Hudry, B., Khadayate, S. and Miguel-Aliaga, I. (2016). The sexual identity of adult intestinal stem cells controls organ size and plasticity. Nature 530: 344-348. PubMed ID: 26887495

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

    Linneweber, G. A., Jacobson, J., Busch, K. E., Hudry, B., Christov, C. P., Dormann, D., Yuan, M., Otani, T., Knust, E., de Bono, M. and Miguel-Aliaga, I. (2014). Neuronal Control of Metabolism through Nutrient-Dependent Modulation of Tracheal Branching. Cell 156: 69-83. PubMed ID: 24439370

    Lemaitre, B. and Miguel-Aliaga, I. (2013). The Digestive Tract of Drosophila melanogaster. Annu Rev Genet. PubMed ID: 24016187

    Sarraf-Zadeh, L., Christen, S., Sauer, U., Cognigni, P., Miguel-Aliaga, I., Stocker, H., Kohler, K. and Hafen, E. (2013). Local requirement of the Drosophila insulin binding protein imp-L2 in coordinating developmental progression with nutritional conditions. Dev Biol 381: 97-106. PubMed ID: 23773803

  • József Mihály Biological Research Centre, Szeged
    Das, R., Bhattacharjee, S., Letcher, J. M., Harris, J. M., Nanda, S., Foldi, I., Lottes, E. N., Bobo, H. M., Grantier, B. D., Mihaly, J., Ascoli, G. A. and Cox, D. N. (2021). Formin 3 directs dendritic architecture via microtubule regulation and is required for somatosensory nociceptive behavior. Development 148(16). PubMed ID: 34322714

    Kiss, V., Jipa, A., Varga, K., Takats, S., Maruzs, T., Lorincz, P., Simon-Vecsei, Z., Szikora, S., Foldi, I., Bajusz, C., Toth, D., Vilmos, P., Gaspar, I., Ronchi, P., Mihaly, J. and Juhasz, G. (2019). Drosophila Atg9 regulates the actin cytoskeleton via interactions with profilin and Ena. Cell Death Differ. PubMed ID: 31740789

    Szikora, S., Gajdos, T., Novak, T., Farkas, D., Foldi, I., Lenart, P., Erdelyi, M. and Mihaly, J. (2020). Nanoscopy reveals the layered organization of the sarcomeric H-zone and I-band complexes. J Cell Biol 219(1). PubMed ID: 31816054

    Migh, E., Gotz, T., Foldi, I., Szikora, S., Gombos, R., Darula, Z., Medzihradszky, K. F., Maleth, J., Hegyi, P., Sigrist, S. and Mihaly, J. (2018). Microtubule organization in presynaptic boutons relies on the formin DAAM. Development 145(6). PubMed ID: 29487108

    Vig, A. T., Foldi, I., Szikora, S., Migh, E., Gombos, R., Toth, M. A., Huber, T., Pinter, R., Talian, G. C., Mihaly, J. and Bugyi, B. (2017). The activities of the c-terminal regions of the formin protein Disheveled-associated activator of morphogenesis (daam) in actin dynamics. J Biol Chem [Epub ahead of print]. PubMed ID: 28642367

    Szikora, S., Foldi, I., Toth, K., Migh, E., Vig, A., Bugyi, B., Maleth, J., Hegyi, P., Kaltenecker, P., Sanchez-Soriano, N. and Mihaly, J. (2017). The formin DAAM is required for coordination of the actin and microtubule cytoskeleton in axonal growth cones. J Cell Sci. PubMed ID: 28606990

    Dollar, G., Gombos, R., Barnett, A. A., Sanchez Hernandez, D., Maung, S. M., Mihaly, J. and Jenny, A. (2016). Unique and overlapping functions of formins Frl and DAAM during ommatidial rotation and neuronal development in Drosophila. Genetics. PubMed ID: 26801180

    Gombos, R., Migh, E., Antal, O., Mukherjee, A., Jenny, A. and Mihaly, J. (2015). The Formin DAAM Functions as Molecular Effector of the Planar Cell Polarity Pathway during Axonal Development in Drosophila. J Neurosci 35: 10154-10167. PubMed ID: 26180192

    Vogler, G., Liu, J., Iafe, T. W., Migh, E., Mihaly, J. and Bodmer, R. (2014). Cdc42 and formin activity control non-muscle myosin dynamics during Drosophila heart morphogenesis. J Cell Biol 206: 909-922. PubMed ID: 25267295

    Molnar, I., Migh, E., Szikora, S., Kalmar, T., Vegh, A. G., Deak, F., Barko, S., Bugyi, B., Orfanos, Z., Kovacs, J., Juhasz, G., Varo, G., Nyitrai, M., Sparrow, J. and Mihaly, J. (2014). DAAM is required for thin filament formation and Sarcomerogenesis during muscle development in Drosophila. PLoS Genet 10: e1004166. PubMed ID: 24586196

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

    Wayland, M. T., Defaye, A., Rocha, J., Jayaram, S. A., Royet, J., Miguel-Aliaga, I., Leulier, F. and Cognigni, P. (2014). Spotting the differences: probing host/microbiota interactions with a dedicated software tool for the analysis of faecal outputs in Drosophila. J Insect Physiol 69: 126-135. PubMed ID: 24907675

    Martorell, O., Merlos-Suarez, A., Campbell, K., Barriga, F. M., Christov, C. P., Miguel-Aliaga, I., Batlle, E., Casanova, J. and Casali, A. (2014). Conserved mechanisms of tumorigenesis in the Drosophila adult midgut. PLoS One 9: e88413. PubMed ID: 24516653

    Sarraf-Zadeh, L., Christen, S., Sauer, U., Cognigni, P., Miguel-Aliaga, I., Stocker, H., Kohler, K. and Hafen, E. (2013). Local requirement of the Drosophila insulin binding protein imp-L2 in coordinating developmental progression with nutritional conditions. Dev Biol 381: 97-106. PubMed ID: 23773803

  • Marco Milán Development and Growth Control Laboratory, IRB Barcelona
    Romao, D., Muzzopappa, M., Barrio, L. and Milan, M. (2021). The Upd3 cytokine couples inflammation to maturation defects in Drosophila. Curr Biol. PubMed ID: 33609452

    Barrio, L. and Milan, M. (2020). Regulation of Anisotropic Tissue Growth by Two Orthogonal Signaling Centers. Dev Cell 52(5): 659-672.e653. PubMed ID: 32084357

    Murcia, L., Clemente-Ruiz, M., Pierre-Elies, P., Royou, A. and Milan, M. (2019). Selective Killing of RAS-Malignant Tissues by Exploiting Oncogene-Induced DNA Damage. Cell Rep 28(1): 119-131.e114. PubMed ID: 31269434

    Benhra, N., Barrio, L., Muzzopappa, M. and Milan, M. (2018). Chromosomal instability induces cellular invasion in epithelial tissues. Dev Cell. PubMed ID: 30245154

    Muzzopappa, M., Murcia, L. and Milan, M. (2017). Feedback amplification loop drives malignant growth in epithelial tissues. Proc Natl Acad Sci U S A. PubMed ID: 28808034

    Barrio, L. and Milan, M. (2017). Boundary Dpp promotes growth of medial and lateral regions of the Drosophila wing. Elife 6. PubMed ID: 28675372

    Terriente-Félix, A., Pérez, L., Bray, S.J., Nebreda, A.R. and Milán, M. (2017). Drosophila model of myeloproliferative neoplasm reveals a feed-forward loop in the JAK pathway mediated by p38 MAPK signalling. Dis Model Mech [Epub ahead of print]. PubMed ID: 28237966

    Recasens-Alvarez, C., Ferreira, A. and Milán, M. (2017). JAK/STAT controls organ size and fate specification by regulating morphogen production and signalling. Nat Commun 8: 13815. PubMed ID: 28045022

    Clemente-Ruiz, M., Murillo-Maldonado, J.M., Benhra, N., Barrio, L., Pérez, L., Quiroga, G., Nebreda, A.R. and Milán, M. (2016). Gene dosage imbalance contributes to chromosomal instability-induced tumorigenesis. Dev Cell 36: 290-302. PubMed ID: 26859353

    Sinadinos, C., Valles-Ortega, J., Boulan, L., Solsona, E., Tevy, M. F., Marquez, M., Duran, J., Lopez-Iglesias, C., Calbo, J., Blasco, E., Pumarola, M., Milan, M. and Guinovart, J. J. (2014). Neuronal glycogen synthesis contributes to physiological aging. Aging Cell. PubMed ID: 25059425

  • Sean Millard School of Biomedical Sciences, University of Queensland, St Lucia, Australia
    Odierna, G. L., Kerwin, S. K., Harris, L. E., Shin, G. J., Lavidis, N. A., Noakes, P. G. and Millard, S. S. (2020). Dscam2 suppresses synaptic strength through a PI3K-dependent endosomal pathway. J Cell Biol 219(6). PubMed ID: 32259198

    Millard, S. S. and Pecot, M. Y. (2018). Strategies for assembling columns and layers in the Drosophila visual system. Neural Dev 13(1): 11. PubMed ID: 29875010

    Kerwin, S. K., Li, J. S. S., Noakes, P. G., Shin, G. J. and Millard, S. S. (2018). Regulated Alternative Splicing of Drosophila Dscam2 Is Necessary for Attaining the Appropriate Number of Photoreceptor Synapses. Genetics 208(2): 717-728. PubMed ID: 29208630

    Lim, N. R., Shohayeb, B., Zaytseva, O., Mitchell, N., Millard, S. S., Ng, D. C. H. and Quinn, L. M. (2017). Glial-Specific Functions of Microcephaly Protein WDR62 and Interaction with the Mitotic Kinase AURKA Are Essential for Drosophila Brain Growth. Stem Cell Reports 9(1): 32-41. PubMed ID: 28625535

  • Tom Millard Faculty of Life Sciences, University of Manchester
    Matsubayashi, Y., Coulson-Gilmer, C. and Millard, T. H. (2015). Endocytosis-dependent coordination of multiple actin regulators is required for wound healing. J Cell Biol 210: 419-433. PubMed ID: 26216900

    Pickering, K., Alves-Silva, J., Goberdhan, D., Millard, T. H. (2013) Par3/Bazooka and phosphoinositides regulate actin protrusion formation during Drosophila dorsal closure and wound healing. Development. PubMed ID: 23318638">

    Stramer, B., Moreira, S., Millard, T., Evans, I., Huang, C. Y., Sabet, O., Milner, M., Dunn, G., Martin, P. and Wood, W. (2010). Clasp-mediated microtubule bundling regulates persistent motility and contact repulsion in Drosophila macrophages in vivo. J Cell Biol 189: 681-689. PubMed ID: 20457764

  • Kathryn Miller
    Isaji, M., Lenartowska, M., Noguchi, T., Frank, D. J. and Miller, K. G. (2011). Myosin VI regulates actin structure specialization through conserved cargo-binding domain sites. PLoS One 6: e22755. PubMed ID: 21853045

    Lenartowska, M., Isaji, M. and Miller, K. G. (2012). A pre-embedding immunogold approach reveals localization of myosin VI at the ultrastructural level in the actin cones that mediate Drosophila spermatid individualization. Protoplasma 249: 337-346. PubMed ID: 21573935

  • Jonathan Minden Department of Biological Sciences, Mellon Institute, Pittsburgh
    Krajcovic, M. M. and Minden, J. S. (2012). Assessing the critical period for Rho kinase activity during Drosophila ventral furrow formation. Dev Dyn 241: 1729-1743. PubMed ID: 22972587

    Monzo, K., Dowd, S. R., Minden, J. S. and Sisson, J. C. (2010). Proteomic analysis reveals CCT is a target of Fragile X mental retardation protein regulation in Drosophila. Dev Biol 340: 408-418. PubMed ID: 20122915

    Fishilevich, E., Fitzpatrick, J. A. and Minden, J. S. (2010). pHMA, a pH-sensitive GFP reporter for cell engulfment, in Drosophila embryos, tissues, and cells. Dev Dyn 239: 559-573. PubMed ID: 20014104

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

    Gutorov, R., Peters, M., Katz, B., Brandwine, T., Barbera, N. A., Levitan, I. and Minke, B. (2019). Modulation of Transient Receptor Potential C Channel Activity by Cholesterol. Front Pharmacol 10: 1487. PubMed ID: 31920669

    Katz, B., Voolstra, O., Tzadok, H., Yasin, B., Rhodes-Modrov, E., Bartels, J. P., Strauch, L., Huber, A. and Minke, B. (2017). The latency of the light response is modulated by the phosphorylation state of Drosophila TRP at a specific site. Channels (Austin): 0. PubMed ID: 28762890

    Voolstra, O., Rhodes-Mordov, E., Katz, B., Bartels, J. P., Oberegelsbacher, C., Schotthofer, S. K., Yasin, B., Tzadok, H., Huber, A. and Minke, B. (2017). The phosphorylation state of the Drosophila TRP channel modulates the frequency response to oscillating light in vivo. J Neurosci 37(15): 4213-4224. PubMed ID: 28314815

    Weiss, S. and Minke, B. (2015). A new genetic model for calcium induced autophagy and ER-stress in Drosophila photoreceptor cells. Channels (Austin) 9: 14-20. PubMed ID: 25664921

    Kohn, E., Katz, B., Yasin, B., Peters, M., Rhodes, E., Zaguri, R., Weiss, S. and Minke, B. (2015). Functional Cooperation between the IP3 Receptor and Phospholipase C Secures the High Sensitivity to Light of Drosophila Photoreceptors In Vivo. J Neurosci 35: 2530-2546. PubMed ID: 25673847

    Weiss, S. and Minke, B. (2015). A new genetic model for calcium induced autophagy and ER-stress in Drosophila photoreceptor cells. Channels (Austin) 9: 14-20. PubMed ID: 25664921

    Katz, B., Oberacker, T., Richter, D., Tzadok, H., Peters, M., Minke, B. and Huber, A. (2013). Drosophila TRP and TRPL are assembled as homomultimeric channels in vivo. J Cell Sci 126: 3121-3133. PubMed ID: 23687378

  • Vincent Mirouse GReD Laboratory, Clermont Universitém Clermont-Ferrand, France
    Venugopal, P., Veyssière, H., Couderc, J. L., Richard, G., Vachias, C. and Mirouse, V. (2020). Multiple functions of the scaffold protein Discs large 5 in the control of growth, cell polarity and cell adhesion in Drosophila melanogaster. BMC Dev Biol 20(1): 10. PubMed ID: 32552730

    Cerqueira Campos, F., Dennis, C., Alegot, H., Fritsch, C., Isabella, A., Pouchin, P., Bardot, O., Horne-Badovinac, S. and Mirouse, V. (2020). Oriented basement membrane fibrils provide a memory for F-actin planar polarization via the Dystrophin-Dystroglycan complex during tissue elongation. Development. PubMed ID: 32156755
    Alegot, H., Pouchin, P., Bardot, O. and Mirouse, V. (2018). Jak-Stat pathway induces Drosophila follicle elongation by a gradient of apical contractility. Elife 7. PubMed ID: 29420170

    Couderc, J. L., Richard, G., Vachias, C. and Mirouse, V. (2017). Drosophila LKB1 is required for the assembly of the polarized actin structure that allows spermatid individualization. PLoS One 12(8): e0182279. PubMed ID: 28767695

    Vachias, C., Fritsch, C., Pouchin, P., Bardot, O. and Mirouse, V. (2014). Tight coordination of growth and differentiation between germline and soma provides robustness for Drosophila egg development. Cell Rep 9: 531-541. PubMed ID: 25373901

    Renaud, Y., Baillif, A., Perez, J. B., Agier, M., Mephu Nguifo, E. and Mirouse, V. (2012). DroPNet: a web portal for integrated analysis of Drosophila protein-protein interaction networks. Nucleic Acids Res 40: W134-139. PubMed ID: 22641854

    Morais-de-Sa, E., Mirouse, V. and St Johnston, D. (2010). aPKC phosphorylation of Bazooka defines the apical/lateral border in Drosophila epithelial cells. Cell 141: 509-523. PubMed ID: 20434988

  • Christen Mirth Instituto Gulbenkian de Ciência, Portugal
    Nogueira Alves, A., Oliveira, M. M., Koyama, T., Shingleton, A. and Mirth, C. K. (2022). Ecdysone coordinates plastic growth with robust pattern in the developing wing. Elife 11. PubMed ID: 35261337

    Alves, A. N., Sgro, C. M., Piper, M. D. W. and Mirth, C. K. (2022). Target of Rapamycin Drives Unequal Responses to Essential Amino Acid Depletion for Egg Laying in Drosophila Melanogaster. Front Cell Dev Biol 10: 822685. PubMed ID: 35252188

    Chakraborty, A., Sgro, C. M. and Mirth, C. K. (2021). The proximate sources of genetic variation in body size plasticity: the relative contributions of feeding behaviour and development in Drosophila melanogaster. J Insect Physiol: 104321. PubMed ID: 34653505

    Chakraborty, A., Sgro, C. M. and Mirth, C. K. (2020). Does local adaptation along a latitudinal cline shape plastic responses to combined thermal and nutritional stress? Evolution 74(9): 2073-2087. PubMed ID: 33616935

    Pocas, G. M., Crosbie, A. E. and Mirth, C. K. (2020). When does diet matter? The roles of larval and adult nutrition in regulating adult size traits in Drosophila melanogaster. J Insect Physiol: 104051. PubMed ID: 32229143

    Henstridge, M. A., Aulsebrook, L., Koyama, T., Johnson, T. K., Whisstock, J. C., Tiganis, T., Mirth, C. K. and Warr, C. G. (2018). Torso-like is a component of the hemolymph and regulates the insulin signalling pathway in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 29440191

    Shingleton, A. W., Masandika, J. R., Thorsen, L. S., Zhu, Y. and Mirth, C. K. (2017). The sex-specific effects of diet quality versus quantity on morphology in Drosophila melanogaster. R Soc Open Sci 4(9): 170375. PubMed ID: 28989746

    Koyama, T. and Mirth, C. K. (2016). Growth-blocking peptides as nutrition-sensitive signals for insulin secretion and body size regulation. PLoS Biol 14: e1002392. PubMed ID: 26928023

    Mendes, C. C. and Mirth, C. K. (2015). Stage-Specific Plasticity in Ovary Size Is Regulated by Insulin/Insulin-Like Growth Factor and Ecdysone Signalling in Drosophila. Genetics. PubMed ID: 26715667

    Matavelli, C., Carvalho, M. J., Martins, N. E. and Mirth, C. K. (2015). Differences in larval nutritional requirements and female oviposition preference reflect the order of fruit colonization of Zaprionus indianus and Drosophila simulans. J Insect Physiol 82: 66-74. PubMed ID: 26358399

  • Rakesh Mishra Centre for Cellular and Molecular Biology, Hyderabad
    Pathak, R. U., Bihani, A., Sureka, R., Varma, P. and Mishra, R. K. (2022). In situ nuclear matrix preparation in Drosophila melanogaster embryos/tissues and its use in studying the components of nuclear architecture. Nucleus 13(1): 116-128. PubMed ID: 35239464Verma, S., Pathak, R. U. and Mishra, R. K. (2021). Genomic organization of the autonomous regulatory domain of eyeless locus in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 34570231

    Ramanujam, P. L., Mehrotra, S., Kumar, R. P., Verma, S., Deshpande, G., Mishra, R. K. and Galande, S. (2021). Global chromatin organizer SATB1 acts as a context-dependent regulator of the Wnt/Wg target genes. Sci Rep 11(1): 3385. PubMed ID: 33564000

    Ponrathnam, T., Saini, R., Banu, S. and Mishra, R. K. (2021). Drosophila Hox genes induce melanized pseudo-tumors when misexpressed in hemocytes. Sci Rep 11(1): 1838. PubMed ID: 33469139

    Sureka, R. and Mishra, R. (2021). Identification of Evolutionarily Conserved Nuclear Matrix Proteins and Their Prokaryotic Origins. J Proteome Res 20(1): 518-530. PubMed ID: 33289389

    Saha, P., Sowpati, D. T., Soujanya, M., Srivastava, I. and Mishra, R. K. (2020). Interplay of pericentromeric genome organization and chromatin landscape regulates the expression of Drosophila melanogaster heterochromatic genes. Epigenetics Chromatin 13(1): 41. PubMed ID: 33028366

    Paddibhatla, I., Gautam, D. K. and Mishra, R. K. (2019). SETDB1 modulates the differentiation of both the crystal cells and the lamellocytes in Drosophila. Dev Biol. PubMed ID: 31422102

    Srivastava, S., Avvaru, A. K., Sowpati, D. T. and Mishra, R. K. (2019). Patterns of microsatellite distribution across eukaryotic genomes. BMC Genomics 20(1): 153. PubMed ID: 30795733

    Saha, P., Sowpati, D. T. and Mishra, R. K. (2019). Epigenomic and genomic landscape of Drosophila melanogaster heterochromatic genes. Genomics 111(2): 177-185. PubMed ID: 29432976

    Hamid, R., Hajirnis, N., Kushwaha, S., Saleem, S., Kumar, V. and Mishra, R. K. (2018). Drosophila Choline transporter non-canonically regulates pupal eclosion and NMJ integrity through a neuronal subset of mushroom body. Dev Biol. PubMed ID: 30529058

    Varma, P. and Mishra, R. K. (2018). Little imaginal discs, a Trithorax group member, is a constituent of nuclear matrix of Drosophila melanogaster embryos. J Biosci 43(4): 621-633. PubMed ID: 30207309

  • Fanis Missirlis Center for Research and Advanced Studies of the National Polytechnic Institute, Departamento de Fisiología, Biofísica y Neurociencias, Mexico City
    Garay, E., Schuth, N., Barbanente, A., Tejeda-GuzmAn, C., Vitone, D., Osorio, B., Clark, A. H., Nachtegaal, M., Haumann, M., Dau, H., Vela, A., Arnesano, F., Quintanar, L. and Missirlis, F. (2022). Tryptophan regulates Drosophila zinc stores. Proc Natl Acad Sci U S A 119(16): e2117807119. PubMed ID: 35412912

    Vasquez-Procopio, J., Rajpurohit, S. and Missirlis, F. (2020). Cuticle darkening correlates with increased body copper content in Drosophila melanogaster. Biometals 33(6): 293-303. PubMed ID: 33026606

    Hernandez-Gallardo, A. K. and Missirlis, F. (2020). Cellular iron sensing and regulation: Nuclear IRP1 extends a classic paradigm. Biochim Biophys Acta Mol Cell Res: 118705. PubMed ID: 32199885

    Vasquez-Procopio, J., Osorio, B., Cortes-Martinez, L., Hernandez-Hernandez, F., Medina-Contreras, O., Rios-Castro, E., Comjean, A., Li, F., Hu, Y., Mohr, S., Perrimon, N. and Missirlis, F. (2019). Intestinal response to dietary manganese depletion in Drosophila. Metallomics. PubMed ID: 31799578

    Marelja, Z., Leimkuhler, S. and Missirlis, F. (2018). Iron sulfur and molybdenum cofactor enzymes regulate the Drosophila life cycle by controlling cell metabolism. Front Physiol 9: 50. PubMed ID: 29491838

    Rosas-Arellano, A., Vasquez-Procopio, J., Gambis, A., Blowes, L. M., Steller, H., Mollereau, B. and Missirlis, F. (2016). Ferritin Assembly in Enterocytes of Drosophila melanogaster. Int J Mol Sci 17 [Epub ahead of print]. PubMed ID: 26861293

    Llorens, J. V., Metzendorf, C., Missirlis, F. and Lind, M. I. (2015). Mitochondrial iron supply is required for the developmental pulse of ecdysone biosynthesis that initiates metamorphosis in Drosophila melanogaster. J Biol Inorg Chem. PubMed ID: 26468126

    Rempoulakis, P., Afshar, N., Osorio, B., Barajas-Aceves, M., Szular, J., Ahmad, S., Dammalage, T., Tomas, U. S., Nemny-Lavy, E., Salomon, M., Vreysen, M. J., Nestel, D. and Missirlis, F. (2014). Conserved metallomics in two insect families evolving separately for a hundred million years. Biometals 27: 1323-1335. PubMed ID: 25298233

    González-Morales, N., Mendoza-Ortíz, M.Á., Blowes, L.M., Missirlis, F. and Riesgo-Escovar, J.R. (2015). Ferritin is required in multiple tissues during Drosophila melanogaster development. PLoS One 10: e0133499. PubMed ID: 26192321

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

  • Masayuki Miura Center for Medical System Innovation, University of Tokyo
    Kashio, S. and Miura, M. (2020). Kynurenine Metabolism in the Fat Body Non-autonomously Regulates Imaginal Disc Repair in Drosophila. iScience 23(12): 101738. PubMed ID: 33376969

    Yamauchi, T., Oi, A., Kosakamoto, H., Akuzawa-Tokita, Y., Murakami, T., Mori, H., Miura, M. and Obata, F. (2020). Gut Bacterial Species Distinctively Impact Host Purine Metabolites during Aging in Drosophila. iScience 23(9): 101477. PubMed ID: 32916085

    Fujisawa, Y., Shinoda, N., Chihara, T. and Miura, M. (2020). ROS Regulate Caspase-Dependent Cell Delamination without Apoptosis in the Drosophila Pupal Notum. iScience 23(8): 101413. PubMed ID: 32791328

    Kosakamoto, H., Yamauchi, T., Akuzawa-Tokita, Y., Nishimura, K., Soga, T., Murakami, T., Mori, H., Yamamoto, K., Miyazaki, R., Koto, A., Miura, M. and Obata, F. (2020). Local Necrotic Cells Trigger Systemic Immune Activation via Gut Microbiome Dysbiosis in Drosophila. Cell Rep 32(3): 107938. PubMed ID: 32698005

    Tsuda-Sakurai, K., Kimura, M. and Miura, M. (2019). Diphthamide modification of eEF2 is required for gut tumor-like hyperplasia induced by oncogenic Ras. Genes Cells. PubMed ID: 31828897

    Shinoda, N., Hanawa, N., Chihara, T., Koto, A. and Miura, M. (2019). Dronc-independent basal executioner caspase activity sustains Drosophila imaginal tissue growth. Proc Natl Acad Sci U S A 116(41): 20539-20544. PubMed ID: 31548372

    Fujisawa, Y., Kosakamoto, H., Chihara, T. and Miura, M. (2019). Non-apoptotic function of Drosophila caspase activation in epithelial thorax closure and wound healing. Development 146(4). PubMed ID: 30770378

    Obata, F., Tsuda-Sakurai, K., Yamazaki, T., Nishio, R., Nishimura, K., Kimura, M., Funakoshi, M. and Miura, M. (2018). Nutritional control of stem cell division through S-Adenosylmethionine in Drosophila intestine. Dev Cell 44(6): 741-751.e743. PubMed ID: 29587144

    Kosakamoto, H., Fujisawa, Y., Obata, F. and Miura, M. (2018). High expression of A-type lamin in the leading front is required for Drosophila thorax closure. Biochem Biophys Res Commun 499(2): 209-214. PubMed ID: 29559239

    Matsukawa, K., Hashimoto, T., Matsumoto, T., Ihara, R., Chihara, T., Miura, M., Wakabayashi, T. and Iwatsubo, T. (2016). Familial ALS-linked mutations in Profilin 1 exacerbate TDP-43-induced degeneration in the retina of Drosophila melanogaster through an increase in the cytoplasmic localization of TDP-43. J Biol Chem [Epub ahead of print]. PubMed ID: 27634045

    Sakuma, C., Saito, Y., Umehara, T., Kamimura, K., Maeda, N., Mosca, T. J., Miura, M. and Chihara, T. (2016). The Strip-Hippo pathway regulates synaptic terminal formation by modulating actin organization at the Drosophila neuromuscular synapses. Cell Rep 16: 2289-2297. PubMed ID: 27545887

  • Marek Mlodzik Mount Sinai Hospital
    Founounou, N., Farhadifar, R., Collu, G. M., Weber, U., Shelley, M. J. and Mlodzik, M. (2021). Tissue fluidity mediated by adherens junction dynamics promotes planar cell polarity-driven ommatidial rotation. Nat Commun 12(1): 6974. PubMed ID: 34848713

    Medina-Yanez, I., Olivares, G. H., Vega-Macaya, F., Mlodzik, M. and Olguin, P. (2020). Phosphatidic acid increases Notch signalling by affecting Sanpodo trafficking during Drosophila sensory organ development. Sci Rep 10(1): 21731. PubMed ID: 33303974

    Humphries, A. C., Narang, S. and Mlodzik, M. (2020). Mutations associated with human neural tube defects display disrupted planar cell polarity in Drosophila. Elife 9. PubMed ID: 32234212

    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

    Domingos, P. M., Jenny, A., Combie, K. F., Del Alamo, D., Mlodzik, M., Steller, H. and Mollereau, B. (2019). Regulation of Numb during planar cell polarity establishment in the Drosophila eye. Mech Dev: 103583. PubMed ID: 31678471

    Thuveson, M., Gaengel, K., Collu, G. M., Chin, M. L., Singh, J. and Mlodzik, M. (2019). Integrins are required for synchronous ommatidial rotation in the Drosophila eye linking planar cell polarity signalling to the extracellular matrix. Open Biol 9(8): 190148. PubMed ID: 31409231

    Vuong, L. T., Iomini, C., Balmer, S., Esposito, D., Aaronson, S. A. and Mlodzik, M. (2018). Kinesin-2 and IFT-A act as a complex promoting nuclear localization of beta-catenin during Wnt signalling. Nat Commun 9(1): 5304. PubMed ID: 30546012

    Bigenzahn, J. W., Collu, G. M., Kartnig, F., Pieraks, M., Vladimer, G. I., Heinz, L. X., Sedlyarov, V., Schischlik, F., Fauster, A., Rebsamen, M., Parapatics, K., Blomen, V. A., Muller, A. C., Winter, G. E., Kralovics, R., Brummelkamp, T. R., Mlodzik, M. and Superti-Furga, G. (2018). LZTR1 is a regulator of RAS ubiquitination and signaling. Science 362(6419): 1171-1177. PubMed ID: 30442766

    Bala Tannan, N., Collu, G., Humphries, A. C., Serysheva, E., Weber, U. and Mlodzik, M. (2018). AKAP200 promotes Notch stability by protecting it from Cbl/lysosome-mediated degradation in Drosophila melanogaster. PLoS Genet 14(1): e1007153. PubMed ID: 29309414

    Weber, U. and Mlodzik, M. (2017). APC/CFzr/Cdh1-dependent regulation of planar cell polarity establishment via Nek2 kinase acting on Dishevelled. Dev Cell 40(1): 53-66. PubMed ID: 28041906

    Vega-Macaya, F., Manieu, C., Valdivia, M., Mlodzik, M. and Olguin, P. (2016). Establishment of the muscle-tendon junction during thorax morphogenesis in Drosophila requires the Rho-kinase. Genetics. PubMed ID: 27585845

  • Ken Moberg Dept. of Cell Biology, Emory University, Atlanta
    Rounds, J. C., Corgiat, E. B., Ye, C., Behnke, J. A., Kelly, S. M., Corbett, A. H. and Moberg, K. H. (2021). The Disease-Associated Proteins Drosophila Nab2 and Ataxin-2 Interact with Shared RNAs and Coregulate Neuronal Morphology. Genetics. PubMed ID: 34791182

    Corgiat, E. B., List, S. M., Rounds, J. C., Corbett, A. H. and Moberg, K. H. (2021). The RNA-binding protein Nab2 regulates the proteome of the developing Drosophila brain. J Biol Chem 297(1): 100877. PubMed ID: 34139237

    Lee, W. H., Corgiat, E. B., Rounds, J. C., Shepherd, Z., Corbett, A. H. and Moberg, K. H. (2020). A Genetic Screen Links the Disease-Associated Nab2 RNA-Binding Protein to the Planar Cell Polarity Pathway in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32817074

    Morton, D. J., Jalloh, B., Kim, L., Kremsky, I., Nair, R. J., Nguyen, K. B., Rounds, J. C., Sterrett, M. C., Brown, B., Le, T., Karkare, M. C., McGaughey, K. D., Sheng, S., Leung, S. W., Fasken, M. B., Moberg, K. H. and Corbett, A. H. (2020). A Drosophila model of Pontocerebellar Hypoplasia reveals a critical role for the RNA exosome in neurons. PLoS Genet 16(7): e1008901. PubMed ID: 32645003

    Bienkowski, R. S., Banerjee, A., Rounds, J. C., Rha, J., Omotade, O. F., Gross, C., Morris, K. J., Leung, S. W., Pak, C., Jones, S. K., Santoro, M. R., Warren, S. T., Zheng, J. Q., Bassell, G. J., Corbett, A. H. and Moberg, K. H. (2017). The conserved, disease-associated RNA binding protein dNab2 interacts with the Fragile X Protein ortholog in Drosophila neurons. Cell Rep 20(6): 1372-1384. PubMed ID: 28793261

    Barron, D.A. and Moberg, K. (2016). Inverse regulation of two classic Hippo pathway target genes in Drosophila by the dimerization hub protein Ctp. Sci Rep 6: 22726. PubMed ID: 26972460

    Zhang, C., Robinson, B. S., Xu, W., Yang, L., Yao, B., Zhao, H., Byun, P. K., Jin, P., Veraksa, A. and Moberg, K. H. (2015). The ecdysone receptor coactivator Taiman links Yorkie to transcriptional control of germline stem cell factors in somatic tissue. Dev Cell 34: 168-180. PubMed ID: 26143992

    Kelly, S.M., Bienkowski, R., Banerjee, A., Melicharek, D.J., Brewer, Z.A., Marenda, D.R., Corbett, A.H. and Moberg, K.H. (2015). The Drosophila ortholog of the ZC3H14 RNA binding protein acts within neurons to pattern axon projection in the developing brain. Dev Neurobiol [Epub ahead of print]. PubMed ID: 25980665

    Gross, C., Chang, C.W., Kelly, S.M., Bhattacharya, A., McBride, S.M., Danielson, S.W., Jiang, M.Q., Chan, C.B., Ye, K., Gibson, J.R., Klann, E., Jongens, T.A., Moberg, K.H., Huber, K.M. and Bassell, G.J. (2015). Increased expression of the PI3K enhancer PIKE mediates deficits in synaptic plasticity and behavior in Fragile X syndrome. Cell Rep [Epub ahead of print]. PubMed ID: 25921541

    Xu, P., Tan, H., Duong, D. M., Yang, Y., Kupsco, J., Moberg, K. H., Li, H., Jin, P. and Peng, J. (2012). Stable isotope labeling with amino acids in Drosophila for quantifying proteins and modifications. J Proteome Res 11: 4403-4412. PubMed ID: 22830426

  • Amanda Moehring Department of Biology, University of Western Ontario
    Isaacson, J. R., Berg, M. D., Charles, B., Jagiello, J., Villen, J., Brandl, C. J. and Moehring, A. J. (2022). A Novel Mistranslating tRNA Model in Drosophila melanogaster has Diverse, Sexually Dimorphic Effects. G3 (Bethesda). PubMed ID: 35143655

    Dhillon, A., Chowdhury, T., Morbey, Y. E. and Moehring, A. J. (2020). Reproductive consequences of an extra long-term sperm storage organ. BMC Evol Biol 20(1): 159. PubMed ID: 33256600

    Ward, H. and Moehring, A. J. (2020). Genes underlying species differences in CHC production between Drosophila melanogaster and D. simulans. Genome. PubMed ID: 33211537

    Chowdhury, T., Calhoun, R. M., Bruch, K. and Moehring, A. J. (2020). The fruitless gene affects female receptivity and species isolation. Proc Biol Sci 287(1923): 20192765. PubMed ID: 32208837

    Kanippayoor, R. L., Alpern, J. H. M. and Moehring, A. J. (2020). A common suite of cellular abnormalities and spermatogenetic errors in sterile hybrid males in Drosophila. Proc Biol Sci 287(1919): 20192291. PubMed ID: 31964309

    Austin, C. J. and Moehring, A. J. (2019). Local thermal adaptation detected during multiple life stages across populations of Drosophila melanogaster. J Evol Biol. PubMed ID: 31454449

    Nguyen, T. T. X. and Moehring, A. J. (2019). Males from populations with higher competitive mating success produce sons with lower fitness. J Evol Biol. PubMed ID: 30811733
    Alpern, J. H. M., Asselin, M. M. and Moehring, A. J. (2019). Identification of a novel sperm class and its role in fertilization in Drosophila. J Evol Biol 32(3): 259-266. PubMed ID: 30484924

    Nguyen, T. T. X. and Moehring, A. J. (2018). A male's seminal fluid increases later competitors' productivity. J Evol Biol. PubMed ID: 30007107

    Pardy, J. A., Rundle, H. D., Bernards, M. A. and Moehring, A. J. (2018). The genetic basis of female pheromone differences between Drosophila melanogaster and D. simulans. Heredity (Edinb). Pubmed ID: 29777168

    Nguyen, T. T. and Moehring, A. J. (2017). Cross-generational comparison of reproductive success in recently caught strains of Drosophila melanogaster. BMC Evol Biol 17(1): 41. PubMed ID: 28166714

    Nguyen, T. T. and Moehring, A. J. (2015). Accurate Alternative Measurements for Female Lifetime Reproductive Success in Drosophila melanogaster. PLoS One 10: e0116679. PubMed ID: 26125633

  • Stephanie Mohr Genetics, Harvard Medical School
    Kenny, A., Morgan, M. B., Mohr, S. and Macdonald, P. M. (2021). Knock down analysis reveals critical phases for specific oskar noncoding RNA functions during Drosophila oogenesis. G3 (Bethesda). PubMed ID: 34586387

    Ding, G., Xiang, X., Hu, Y., Xiao, G., Chen, Y., Binari, R., Comjean, A., Li, J., Rushworth, E., Fu, Z., Mohr, S. E., Perrimon, N. and Song, W. (2021). Coordination of tumor growth and host wasting by tumor-derived Upd3. Cell Rep 36(7): 109553. PubMed ID: 34407411

    Xia, B., Amador, G., Viswanatha, R., Zirin, J., Mohr, S. E. and Perrimon, N. (2020). CRISPR-based engineering of gene knockout cells by homology-directed insertion in polyploid Drosophila S2R+ cells. Nat Protoc 15(10): 3478-3498. PubMed ID: 32958931

    Kanca, O., Zirin, J., Garcia-Marques, J., Knight, S. M., Yang-Zhou, D., Amador, G., Chung, H., Zuo, Z., Ma, L., He, Y., Lin, W. W., Fang, Y., Ge, M., Yamamoto, S., Schulze, K. L., Hu, Y., Spradling, A. C., Mohr, S. E., Perrimon, N. and Bellen, H. J. (2019). An efficient CRISPR-based strategy to insert small and large fragments of DNA using short homology arms. Elife 8. PubMed ID: 31674908

    Hu, Y., Comjean, A., Perrimon, N. and Mohr, S. E. (2017). The Drosophila Gene Expression Tool (DGET) for expression analyses. BMC Bioinformatics 18(1): 98. PubMed ID: 28187709

    Hu, Y., Comjean, A., Perkins, L. A., Perrimon, N. and Mohr, S. E. (2015). GLAD: an online database of gene list annotation for Drosophila. J Genomics 3: 75-81. PubMed ID: 26157507

    Yilmazel, B., Hu, Y., Sigoillot, F., Smith, J. A., Shamu, C. E., Perrimon, N. and Mohr, S. E. (2014). Online GESS: prediction of miRNA-like off-target effects in large-scale RNAi screen data by seed region analysis. BMC Bioinformatics 15: 192. PubMed ID: 24934636

    Mohr, S. E. (2014). RNAi screening in Drosophila cells and in vivo. Methods. PubMed ID: 24576618

    Vinayagam, A., Zirin, J., Roesel, C., Hu, Y., Yilmazel, B., Samsonova, A. A., Neumuller, R. A., Mohr, S. E. and Perrimon, N. (2013). Integrating protein-protein interaction networks with phenotypes reveals signs of interactions. Nat Methods. PubMed ID: 24240319

    Hu, Y., Sopko, R., Foos, M., Kelley, C., Flockhart, I., Ammeux, N., Wang, X., Perkins, L., Perrimon, N. and Mohr, S. E. (2013). FlyPrimerBank: An Online Database for Drosophila melanogaster Gene Expression Analysis and Knockdown Evaluation of RNAi Reagents. G3 (Bethesda). PubMed ID: 23893746

    Bergwitz, C., Wee, M. J., Sinha, S., Huang, J., DeRobertis, C., Mensah, L. B., Cohen, J., Friedman, A., Kulkarni, M., Hu, Y., Vinayagam, A., Schnall-Levin, M., Berger, B., Perkins, L. A., Mohr, S. E. and Perrimon, N. (2013). Genetic determinants of phosphate response in Drosophila. PLoS One 8: e56753. PubMed ID: 23520455

  • Bertrand Mollereau Laboratory of Molecular Biology of the Cell, Ecole Normale Superieure of Lyon
    Arsac, J. N., Sedru, M., Dartiguelongue, M., Vulin, J., Davoust, N., Baron, T. and Mollereau, B. (2021). Chronic Exposure to Paraquat Induces Alpha-Synuclein Pathogenic Modifications in Drosophila. Int J Mol Sci 22(21). PubMed ID: 34769043

    Girard, V., Jollivet, F., Knittelfelder, O., Celle, M., Arsac, J. N., Chatelain, G., Van den Brink, D. M., Baron, T., Shevchenko, A., Kuhnlein, R. P., Davoust, N. and Mollereau, B. (2021). Abnormal accumulation of lipid droplets in neurons induces the conversion of alpha-Synuclein to proteolytic resistant forms in a Drosophila model of Parkinson's disease. PLoS Genet 17(11): e1009921. PubMed ID: 34788284

    Girard, V., Goubard, V., Querenet, M., Seugnet, L., Pays, L., Nataf, S., Dufourd, E., Cluet, D., Mollereau, B. and Davoust, N. (2020). Spen modulates lipid droplet content in adult Drosophila glial cells and protects against paraquat toxicity. Sci Rep 10(1): 20023. PubMed ID: 33208773

    Domingos, P. M., Jenny, A., Combie, K. F., Del Alamo, D., Mlodzik, M., Steller, H. and Mollereau, B. (2019). Regulation of Numb during planar cell polarity establishment in the Drosophila eye. Mech Dev: 103583. PubMed ID: 31678471

    Robin, M., Issa, A. R., Santos, C. C., Napoletano, F., Petitgas, C., Chatelain, G., Ruby, M., Walter, L., Birman, S., Domingos, P. M., Calvi, B. R. and Mollereau, B. (2019). Drosophila p53 integrates the antagonism between autophagy and apoptosis in response to stress. Autophagy 15(5): 771-784. PubMed ID: 30563404

    Betsch, L., Boltz, V., Brioudes, F., Pontier, G., Girard, V., Savarin, J., Wipperman, B., Chambrier, P., Tissot, N., Benhamed, M., Mollereau, B., Raynaud, C., Bendahmane, M. and Szecsi, J. (2019). TCTP and CSN4 control cell cycle progression and development by regulating CULLIN1 neddylation in plants and animals. PLoS Genet 15(1): e1007899. PubMed ID: 30695029

    Robin, M., Issa, A. R., Santos, C. C., Napoletano, F., Petitgas, C., Chatelain, G., Ruby, M., Walter, L., Birman, S., Domingos, P. M., Calvi, B. R. and Mollereau, B. (2018). Drosophila p53 integrates the antagonism between autophagy and apoptosis in response to stress. Autophagy. PubMed ID: 30563404

    Rosas-Arellano, A., Vasquez-Procopio, J., Gambis, A., Blowes, L. M., Steller, H., Mollereau, B. and Missirlis, F. (2016). Ferritin Assembly in Enterocytes of Drosophila melanogaster. Int J Mol Sci 17 [Epub ahead of print]. PubMed ID: 26861293

    Querenet, M., Danjoy, M. L., Mollereau, B. and Davoust, N. (2015). Expression of dengue virus NS3 protein in Drosophila alters its susceptibility to infection. Fly (Austin) 9: 1-6. PubMed ID: 26267447

    Querenet, M., Goubard, V., Chatelain, G., Davoust, N. and Mollereau, B. (2015). Spen is required for pigment cell survival during pupal development in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 25872184

    Ducuing, A., Keeley, C., Mollereau, B. and Vincent, S. (2015).A DPP-mediated feed-forward loop canalizes morphogenesis during Drosophila dorsal closure. J Cell Biol 208: 239-248. PubMed ID: 25601405

  • Veronique Monnier Paris Diderot University
    Russi, M., Martin, E., D'Autreaux, B., Tixier, L., Tricoire, H. and Monnier, V. (2020). A Drosophila model of Friedreich ataxia with CRISPR/Cas9 insertion of GAA repeats in the frataxin gene reveals in vivo protection by N-acetyl cysteine. Hum Mol Genet 29(17): 2831-2844. PubMed ID: 32744307

    Monnier, V., Llorens, J. V. and Navarro, J. A. (2018). Impact of Drosophila Models in the Study and Treatment of Friedreich's Ataxia. Int J Mol Sci 19(7). PubMed ID: 29986523

    Palandri, A., Martin, E., Russi, M., Rera, M., Tricoire, H. and Monnier, V. (2018). Identification of cardioprotective drugs by medium-scale in vivo pharmacological screening on a Drosophila cardiac model of Friedreich's ataxia. Dis Model Mech 11(7). PubMed ID: 29898895

  • Jacques Montagne Institut de Biologie Intégrative de la Cellule, CNRS, Gif-sur-Yvette
    Devilliers, M., Garrido, D., Poidevin, M., Rubin, T., Le Rouzic, A. and Montagne, J. (2021). Differential metabolic sensitivity of insulin-like-response- and TORC1-dependent overgrowth in Drosophila fat cells. Genetics 217(1): 1-12. PubMed ID: 33683355

    Zahoor, M. K., Poidevin, M., Lecerf, C., Garrido, D. and Montagne, J. (2019). A Drosophila genetic screen for suppressors of S6kinase-dependent growth identifies the F-box subunit Archipelago/FBXW7. Mol Genet Genomics. PubMed ID: 30656413

    Garrido, D., Rubin, T., Poidevin, M., Maroni, B., Le Rouzic, A., Parvy, J. P. and Montagne, J. (2015). Fatty Acid Synthase Cooperates with Glyoxalase 1 to Protect against Sugar Toxicity. PLoS Genet 11: e1004995. PubMed ID: 25692475

    Parvy, J. P., Wang, P., Garrido, D., Maria, A., Blais, C., Poidevin, M. and Montagne, J. (2014). Forward and feedback regulation of cyclic steroid production in Drosophila melanogaster. Development 141: 3955-3965. PubMed ID: 25252945

    Parvy, J. P., Napal, L., Rubin, T., Poidevin, M., Perrin, L., Wicker-Thomas, C. and Montagne, J. (2012). Drosophila melanogaster Acetyl-CoA-carboxylase sustains a fatty acid-dependent remote signal to waterproof the respiratory system. PLoS Genet 8: e1002925. PubMed ID: 22956916

  • Eurico Morais de Så
    Ventura, G., Moreira, S., Barros-Carvalho, A., Osswald, M. and Morais-de-Sa, E. (2020). Lgl cortical dynamics are independent of binding to the Scrib-Dlg complex but require Dlg-dependent restriction of aPKC. Development 147(15). PubMed ID: 32665243

    Afonso, O., Castellani, C. M., Cheeseman, L. P., Ferreira, J. G., Orr, B., Ferreira, L. T., Chambers, J. J., Morais-de-Sa, E., Maresca, T. J. and Maiato, H. (2019). Spatiotemporal control of mitotic exit during anaphase by an aurora B-Cdk1 crosstalk. Elife 8. PubMed ID: 31424385

    Osswald, M., Santos, A. F. and Morais-de-Sa, E. (2019). Light-Induced Protein Clustering for Optogenetic Interference and Protein Interaction Analysis in Drosophila S2 Cells. Biomolecules 9(2). PubMed ID: 30759894

    Moreira, S., Osswald, M., Ventura, G., Goncalves, M., Sunkel, C. E. and Morais-de-Sa, E. (2019). PP1-Mediated Dephosphorylation of Lgl Controls Apical-basal Polarity. Cell Rep 26(2): 293-301 e297. PubMed ID: 30625311

  • Catherine Montchamp Moreau Laboratoire Evolution, Genomes et Speciation, CNRS, Paris
    Helleu, Q., Gerard, P. R., Dubruille, R., Ogereau, D., Prud'homme, B., Loppin, B. and Montchamp-Moreau, C. (2016). Rapid evolution of a Y-chromosome heterochromatin protein underlies sex chromosome meiotic drive. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26979956

    Wurmser, F., Mary-Huard, T., Daudin, J. J., Joly, D. and Montchamp-Moreau, C. (2013). Variation of Gene Expression Associated with Colonisation of an Anthropized Environment: Comparison between African and European Populations of. PLoS One 8: e79750. PubMed ID: 24260296

    Bastide, H., Gerard, P. R., Ogereau, D., Cazemajor, M. and Montchamp-Moreau, C. (2013). Local dynamics of a fast-evolving sex-ratio system in Drosophila simulans. Mol Ecol 22: 5352-5367. PubMed ID: 24118375

    Fouvry, L., Ogereau, D., Berger, A., Gavory, F. and Montchamp-Moreau, C. (2011). Sequence Analysis of the Segmental Duplication Responsible for Paris Sex-Ratio Drive in Drosophila simulans. G3 (Bethesda) 1: 401-410. PubMed ID: 22384350

    Bastide, H., Cazemajor, M., Ogereau, D., Derome, N., Hospital, F. and Montchamp-Moreau, C. (2011). Rapid rise and fall of selfish sex-ratio X chromosomes in Drosophila simulans: spatiotemporal analysis of phenotypic and molecular data. Mol Biol Evol 28: 2461-2470. PubMed ID: 21498605

    Wurmser, F., Ogereau, D., Mary-Huard, T., Loriod, B., Joly, D. and Montchamp-Moreau, C. (2011). Population transcriptomics: insights from Drosophila simulans, Drosophila sechellia and their hybrids. Genetica 139: 465-477. PubMed ID: 21424276

  • Craig Montell Neuroscience Research Institute and the Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara
    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

    Ganguly, A., Chandel, A., Turner, H., Wang, S., Liman, E. R. and Montell, C. (2021). Requirement for an Otopetrin-like protein for acid taste in Drosophila. Proc Natl Acad Sci U S A 118(51). PubMed ID: 34911758

    Li, Q. and Montell, C. (2021). Mechanism for food texture preference based on grittiness. Curr Biol. PubMed ID: 33657409

    Sun, G., Ding, X. A., Argaw, Y., Guo, X. and Montell, D. J. (2020). Akt1 and dCIZ1 promote cell survival from apoptotic caspase activation during regeneration and oncogenic overgrowth. Nat Commun 11(1): 5726. PubMed ID: 33184261

    Miao, G., Godt, D. and Montell, D. J. (2020). Integration of Migratory Cells into a New Site In Vivo Requires Channel-Independent Functions of Innexins on Microtubules. Dev Cell. PubMed ID: 32668209

    Zhang, J., Luo, J., Chen, J., Dai, J. and Montell, C. (2020). The Role of Y Chromosome Genes in Male Fertility in Drosophila melanogaster. Genetics. PubMed ID: 32404399

    Chen, Z. and Montell, C. (2020). A Family of Auxiliary Subunits of the TRP Cation Channel Encoded by the Complex inaF Locus. Genetics. PubMed ID: 32434796

    Li, Q., DeBeaubien, N. A., Sokabe, T. and Montell, C. (2020). Temperature and Sweet Taste Integration in Drosophila. Curr Biol. PubMed ID: 32330421

    Liu, W., Ganguly, A., Huang, J., Wang, Y., Ni, J. D., Gurav, A. S., Aguilar, M. A. and Montell, C. (2019). Neuropeptide F regulates courtship in Drosophila through a male-specific neuronal circuit. Elife 8. PubMed ID: 31403399

    Rimal, S., Sang, J., Poudel, S., Thakur, D., Montell, C. and Lee, Y. (2019). Mechanism of acetic acid gustatory repulsion in Drosophila. Cell Rep 26(6): 1432-1442.e1434. PubMed ID: 30726729

    Ni, J. D., Gurav, A. S., Liu, W., Ogunmowo, T. H., Hackbart, H., Elsheikh, A., Verdegaal, A. A. and Montell, C. (2019). Differential regulation of the Drosophila sleep homeostat by circadian and arousal inputs. Elife 8. PubMed ID: 30719975

    Liu, J., Sokabe, T. and Montell, C. (2018). A temperature gradient assay to determine thermal preferences of Drosophila larvae. J Vis Exp(136). PubMed ID: 29985331

  • Denise Montell Molecular Cellular and Developmental Biology, University of California Santa Barbara
    Miao, G., Guo, L. and Montell, D. J. (2022). Border cell polarity and collective migration require the spliceosome component Cactin. J Cell Biol 221(7). PubMed ID: 35612426

    Easwaran, S., Van Ligten, M., Kui, M. and Montell, D. J. (2022). Enhanced germline stem cell longevity in Drosophila diapause. Nat Commun 13(1): 711. PubMed ID: 35132083

    Lowe, D. D. and Montell, D. J. (2021). Unconventional translation initiation factor EIF2A is required for Drosophila spermatogenesis. Dev Dyn. PubMed ID: 34278643

    Dai, W., Guo, X., Cao, Y., Mondo, J. A., Campanale, J. P., Montell, B. J., Burrous, H., Streichan, S., Gov, N., Rappel, W. J. and Montell, D. J. (2020). Tissue topography steers migrating Drosophila border cells. Science 370(6519): 987-990. PubMed ID: 33214282

    Mishra, A. K., Mondo, J. A., Campanale, J. P. and Montell, D. J. (2019). Coordination of protrusion dynamics within and between collectively migrating border cells by myosin II. Mol Biol Cell 30(19): 2490-2502. PubMed ID: 31390285

    Chang, Y. C., Wu, J. W., Hsieh, Y. C., Huang, T. H., Liao, Z. M., Huang, Y. S., Mondo, J. A., Montell, D. and Jang, A. C. (2018). Rap1 Negatively Regulates the Hippo Pathway to Polarize Directional Protrusions in Collective Cell Migration. Cell Rep 22(8): 2160-2175. PubMed ID: 29466741

    Manning, L., Sheth, J., Bridges, S., Saadin, A., Odinammadu, K., Andrew, D., Spencer, S., Montell, D. and Starz-Gaiano, M. (2017). A hormonal cue promotes timely follicle cell migration by modulating transcription profiles. Mech Dev. PubMed ID: 28610887

    Cho, A., Kato, M., Whitwam, T., Kim, J.H. and Montell, D.J. (2016). An atypical tropomyosin in Drosophila with intermediate filament-like properties. Cell Rep [Epub ahead of print]. PubMed ID: 27396338

    Ding, A. X., Sun, G., Argaw, Y. G., Wong, J. O., Easwaran, S. and Montell, D. J. (2016). CasExpress reveals widespread and diverse patterns of cell survival of caspase-3 activation during development. Elife 5. PubMed ID: 27058168

    Xiang, W., Zhang, D. and Montell, D. J. (2015). Tousled-like kinase regulates cytokine-mediated communication between cooperating cell types during collective border cell migration. Mol Biol Cell. PubMed ID: 26510500

  • Kristi Montooth Department of Biology, University of Indiana, Bloomington
    Matoo, O. B., Julick, C. R. and Montooth, K. L. (2019). Genetic Variation for Ontogenetic Shifts in Metabolism Underlies Physiological Homeostasis in Drosophila. Genetics. PubMed ID: 30975764

    Hoekstra, L. A., Julick, C. R., Mika, K. M. and Montooth, K. L. (2018). Energy demand and the context-dependent effects of genetic interactions underlying metabolism. Evol Lett 2(2): 102-113. PubMed ID: 30283668

    Lockwood, B. L., Julick, C. R. and Montooth, K. L. (2017). Maternal loading of a small heat shock protein increases embryo thermal tolerance in Drosophila melanogaster. J Exp Biol 220(Pt 23): 4492-4501. PubMed ID: 29097593

    Cooper, B.S., Burrus, C.R., Ji, C., Hahn, M.W. and Montooth, K.L. (2015). Similar efficacies of selection shape mitochondrial and nuclear genes in both Drosophila melanogaster and Homo sapiens. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26297726

    Cooper, B. S., Hammad, L. A. and Montooth, K. L. (2014). Thermal adaptation of cellular membranes in natural populations of Drosophila melanogaster. Funct Ecol 28: 886-894. PubMed ID: 25382893

    Cooper, B. S., Hammad, L. A. and Montooth, K. L. (2014). Thermal adaptation of cellular membranes in natural populations of. Funct Ecol 28: 886-894. PubMed ID: 25382893

    Hoekstra, L. A., Siddiq, M. A. and Montooth, K. L. (2013). Pleiotropic Effects of a Mitochondrial-Nuclear Incompatibility Depend Upon the Accelerating Effect of Temperature in Drosophila. Genetics. PubMed ID: 24026098

    Hoekstra, L. A., Montooth, K. L. (2013) Inducing extra copies of the Hsp70 gene in Drosophila melanogaster increases energetic demand. BMC Evol Biol 13: 68. PubMed ID: 23510136

    Meiklejohn, C. D., Holmbeck, M. A., Siddiq, M. A., Abt, D. N., Rand, D. M., Montooth, K. L. (2013) An Incompatibility between a Mitochondrial tRNA and Its Nuclear-Encoded tRNA Synthetase Compromises Development and Fitness in Drosophila. PLoS Genet 9: e1003238. PubMed ID: 23382693

    Meiklejohn, C. D., Holmbeck, M. A., Siddiq, M. A., Abt, D. N., Rand, D. M. and Montooth, K. L. (2013). An Incompatibility between a Mitochondrial tRNA and Its Nuclear-Encoded tRNA Synthetase Compromises Development and Fitness in Drosophila. PLoS Genet 9: e1003238. PubMed ID: 23382693

  • Eduardo Moreno Institute of Cell Biology, Bern University, Switzerland
    Coelho, D. S. and Moreno, E. (2020). Neuronal Selection Based on Relative Fitness Comparison Detects and Eliminates Amyloid-β-Induced Hyperactive Neurons in Drosophila. iScience 23(9): 101468. PubMed ID: 32866827

    Madan, E., Pelham, C. J., Nagane, M., Parker, T. M., Canas-Marques, R., Fazio, K., Shaik, K., Yuan, Y., Henriques, V., Galzerano, A., Yamashita, T., Pinto, M. A. F., Palma, A. M., Camacho, D., Vieira, A., Soldini, D., Nakshatri, H., Post, S. R., Rhiner, C., Yamashita, H., Accardi, D., Hansen, L. A., Carvalho, C., Beltran, A. L., Kuppusamy, P., Gogna, R. and Moreno, E. (2019). Flower isoforms promote competitive growth in cancer. Nature. PubMed ID: 31341286

    Coelho, D. S., Schwartz, S., Merino, M. M., Hauert, B., Topfel, B., Tieche, C., Rhiner, C. and Moreno, E. (2018). Culling less fit neurons protects against Amyloid-beta-induced brain damage and cognitive and motor decline. Cell Rep 25(13): 3661-3673. PubMed ID: 30590040

    Levayer, R., Dupont, C. and Moreno, E. (2016). Tissue Crowding Induces Caspase-Dependent Competition for Space. Curr Biol [Epub ahead of print]. PubMed ID: 26898471

    Casas-Tinto, S., Lolo, F. N. and Moreno, E. (2015). Active JNK-dependent secretion of Drosophila Tyrosyl-tRNA synthetase by loser cells recruits haemocytes during cell competition. Nat Commun 6: 10022. PubMed ID: 26658841

    Moreno, E., Fernandez-Marrero, Y., Meyer, P. and Rhiner, C. (2015). Brain regeneration in Drosophila involves comparison of neuronal fitness. Curr Biol [Epub ahead of print]. PubMed ID: 25754635

    Merino, M. M., Rhiner, C., Lopez-Gay, J. M., Buechel, D., Hauert, B. and Moreno, E. (2015). Elimination of unfit cells maintains tissue health and prolongs lifespan. Cell. PubMed ID: 25601460

    Merino, M. M., Rhiner, C., Portela, M. and Moreno, E. (2013). "Fitness Fingerprints" Mediate Physiological Culling of Unwanted Neurons in Drosophila. Curr Biol. PubMed ID: 23810538

    Fernandez-Hernandez, I., Rhiner, C. and Moreno, E. (2013). Adult Neurogenesis in Drosophila. Cell Rep. PubMed ID: 23791523

    Portela, M., Casas-Tinto, S., Rhiner, C., Lopez-Gay, J. M., Dominguez, O., Soldini, D. and Moreno, E. (2010). Drosophila SPARC is a self-protective signal expressed by loser cells during cell competition. Dev Cell 19: 562-573. PubMed ID: 20951347

    Rhiner, C., Lopez-Gay, J. M., Soldini, D., Casas-Tinto, S., Martin, F. A., Lombardia, L. and Moreno, E. (2010). Flower forms an extracellular code that reveals the fitness of a cell to its neighbors in Drosophila. Dev Cell 18: 985-998. PubMed ID: 20627080

  • Marta Morey Developmental Biology and Genomics, Institut de Biomedicina de la Universitat de Barcelona
    Plazaola-Sasieta, H., Zhu, Q., Gaitan-Penas, H., Rios, M., Estevez, R. and Morey, M. (2019). Drosophila ClC-a is required in glia of the stem cell niche for proper neurogenesis and wiring of neural circuits. Glia. PubMed ID: 31479171

    Plazaola-Sasieta, H., Fernandez-Pineda, A., Zhu, Q. and Morey, M. (2017). Untangling the wiring of the Drosophila visual system: developmental principles and molecular strategies. J Neurogenet 31(4): 231-249. PubMed ID: 29078717

    Morey, M. (2017). Dpr-DIP matching expression in Drosophila synaptic pairs. Fly (Austin) 11(1): 19-26. PubMed ID: 27450981

    Tan, L., Zhang, K. X., Pecot, M. Y., Nagarkar-Jaiswal, S., Lee, P. T., Takemura, S. Y., McEwen, J. M., Nern, A., Xu, S., Tadros, W., Chen, Z., Zinn, K., Bellen, H. J., Morey, M. and Zipursky, S. L. (2015). Ig Superfamily Ligand and Receptor Pairs Expressed in Synaptic Partners in Drosophila. Cell 163(7): 1756-1769. PubMed ID: 26687360

  • Nam-Sung Moon McGill University
    Kim, M., Delos Santos, K. and Moon, N. S. (2021). Proper CycE-Cdk2 activity in endocycling tissues requires regulation of the cyclin-dependent kinase inhibitor Dacapo by dE2F1b in Drosophila. Genetics 217(1): 1-15. PubMed ID: 33683365

    Delos Santos, K., Kim, M., Yergeau, C., Jean, S. and Moon, N. S. (2019). Pleiotropic role of Drosophila phosphoribosyl pyrophosphate synthetase in autophagy and lysosome homeostasis. PLoS Genet 15(9): e1008376. PubMed ID: 31487280

    Kim, M., Tang, J. P. and Moon, N. S. (2018). An alternatively spliced form affecting the Marked Box domain of Drosophila E2F1 is required for proper cell cycle regulation. PLoS Genet 14(2): e1007204. PubMed ID: 29420631

    Bradley-Gill, M.R., Kim, M., Feingold, D., Yergeau, C., Houde, J. and Moon, N.S. (2016). Alternate transcripts of the Drosophila "activator" E2F are necessary for maintenance of cell cycle exit during development. Dev Biol [Epub ahead of print]. PubMed ID: 26859702

    Collins, H. and Moon, N. S. (2013). The Components of Drosophila Histone Chaperone dCAF-1 Are Required for the Cell Death Phenotype Associated with rbf1 Mutation. G3 (Bethesda). PubMed ID: 23893745

    Krivy, K., Bradley-Gill, M. R., Moon, N. S. (2013) Capicua regulates proliferation and survival of RB-deficient cells in Drosophila. Biol Open 2: 183-190. PubMed ID: 23429853

    Popova, M. K., He, W., Korenjak, M., Dyson, N. J. and Moon, N. S. (2011). Rb deficiency during Drosophila eye development deregulates EMC, causing defects in the development of photoreceptors and cone cells. J Cell Sci 124: 4203-4212. PubMed ID: 22193959

    Nicolay, B. N., Bayarmagnai, B., Moon, N. S., Benevolenskaya, E. V. and Frolov, M. V. (2010). Combined inactivation of pRB and hippo pathways induces dedifferentiation in the Drosophila retina. PLoS Genet 6: e1000918. PubMed ID: 20421993

  • Adrian Moore Riken Brain Science Institute, Hirosawa Wako City, Saitama
    Yoong, L. F., Lim, H. K., Tran, H., Lackner, S., Zheng, Z., Hong, P. and Moore, A. W. (2020). Atypical Myosin Tunes Dendrite Arbor Subdivision. Neuron. PubMed ID: 32155441

    Klebanow, L. R., Peshel, E. C., Schuster, A. T., De, K., Sarvepalli, K., Lemieux, M. E., Lenoir, J. J., Moore, A. W., McDonald, J. A. and Longworth, M. S. (2016). Drosophila Condensin II subunit, Chromosome Associated Protein-D3, regulates cell fate determination through non-cell autonomous signaling. Development. PubMed ID: 27317808

    Yalgin, C., Ebrahimi, S., Delandre, C., Yoong, L. F., Akimoto, S., Tran, H., Amikura, R., Spokony, R., Torben-Nielsen, B., White, K. P. and Moore, A. W. (2015). Centrosomin represses dendrite branching by orienting microtubule nucleation. Nat Neurosci. PubMed ID: 26322925

    Taniguchi, H. and Moore, A. W. (2014). Chromatin regulators in neurodevelopment and disease: Analysis of fly neural circuits provides insights: Networks of chromatin regulators and transcription factors underlie Drosophila neurogenesis and cognitive defects in intellectual disability and neuropsychiatric disorder models. Bioessays. PubMed ID: 25067789

    Nagel, J., Delandre, C., Zhang, Y., Forstner, F., Moore, A. W. and Tavosanis, G. (2012). Fascin controls neuronal class-specific dendrite arbor morphology. Development 139: 2999-3009. PubMed ID: 22764047

    Hohenauer, T. and Moore, A. W. (2012). The Prdm family: expanding roles in stem cells and development. Development 139: 2267-2282. PubMed ID: 22669819

    Endo, K., Karim, M. R., Taniguchi, H., Krejci, A., Kinameri, E., Siebert, M., Ito, K., Bray, S. J. and Moore, A. W. (2012). Chromatin modification of Notch targets in olfactory receptor neuron diversification. Nat Neurosci 15: 224-233. PubMed ID: 22197833

  • Ginés Morata Molecular Biology Institute of Barcelona
    Azpiazu, N. and Morata, G. (2022). Chromatin remodelling and retrotransposons activities during regeneration in Drosophila. Dev Biol 482: 7-16. PubMed ID: 34822846

    Medina, I., Calleja, M. and Morata, G. (2021). Tumorigenesis and cell competition in Drosophila in the absence of polyhomeotic function. Proc Natl Acad Sci U S A 118(45). PubMed ID: 34702735

    Lawrence, P. A., Casal, J., Celis, J. F. and Morata, G. (2019). A refutation to 'A new A-P compartment boundary and organizer in holometabolous insect wings'. Sci Rep 9(1): 7049. PubMed ID: 31065001

    Martin, R., Pinal, N. and Morata, G. (2017). Distinct regenerative potential of trunk and appendages of Drosophila mediated by JNK signalling. Development. PubMed ID: 28935711

    Montes, A. J. and Morata, G. (2017). Homeostatic response to blocking cell division in Drosophila imaginal discs: Role of the Fat/Dachsous (Ft/Ds) pathway. Dev Biol 424(2):113-123. PubMed ID: 28300568

    Calleja, M., Morata, G. and Casanova, J. (2016). The tumorigenic properties of Drosophila epithelial cells mutant for lethal giant larvae. Dev Dyn [Epub ahead of print]. PubMed ID: 27239786

    Herrera, S. C. and Morata, G. (2014). Transgressions of compartment boundaries and cell reprogramming during regeneration in Drosophila. Elife 3: e01831. PubMed ID: 24755288

    Morata, G. and Herrera, S. C. (2010). Differential division rates and size control in the wing disc. Fly (Austin) 4: 226-229. PubMed ID: 20224294

    Ballesteros-Arias, L., Saavedra, V. and Morata, G. (2013). Cell competition may function either as tumour-suppressing or as tumour-stimulating factor in Drosophila. Oncogene. PubMed ID: 24096487

    Shlevkov, E. and Morata, G. (2012). A dp53/JNK-dependant feedback amplification loop is essential for the apoptotic response to stress in Drosophila. Cell Death Differ 19: 451-460. PubMed ID: 21886179

    Morata, G., Shlevkov, E. and Perez-Garijo, A. (2011). Mitogenic signaling from apoptotic cells in Drosophila. Dev Growth Differ 53: 168-176. PubMed ID: 21338343

  • Ted Morgan Division of Biology, Kansas State University, Manhattan
    Freda, P. J., Ali, Z. M., Heter, N., Ragland, G. J. and Morgan, T. J. (2019). Stage-specific genotype-by-environment interactions for cold and heat hardiness in Drosophila melanogaster. Heredity (Edinb). PubMed ID: 31164731

    Gleason, J. M., Roy, P. R., Everman, E. R., Gleason, T. C. and Morgan, T. J. (2019). Phenology of Drosophila species across a temperate growing season and implications for behavior. PLoS One 14(5): e0216601. PubMed ID: 31095588

    Everman, E. R., Delzeit, J. L., Hunter, F. K., Gleason, J. M. and Morgan, T. J. (2018). Costs of cold acclimation on survival and reproductive behavior in Drosophila melanogaster. PLoS One 13(5): e0197822. PubMed ID: 29791517

    Noh, S., Everman, E. R., Berger, C. M. and Morgan, T. J. (2017). Seasonal variation in basal and plastic cold tolerance: Adaptation is influenced by both long- and short-term phenotypic plasticity. Ecol Evol 7(14): 5248-5257. PubMed ID: 28770063

    Gerken, A. R., Eller, O. C., Hahn, D. A. and Morgan, T. J. (2015). Constraints, independence, and evolution of thermal plasticity: Probing genetic architecture of long- and short-term thermal acclimation. Proc Natl Acad Sci U S A 112: 4399-4404. PubMed ID: 25805817

    Williams, C. M., Watanabe, M., Guarracino, M. R., Ferraro, M. B., Edison, A. S., Morgan, T. J., Boroujerdi, A. F. and Hahn, D. A. (2014). Cold adaptation shapes the robustness of metabolic networks in Drosophila melanogaster. Evolution 68: 3505-3523. PubMed ID: 25308124

    Fallis, L. C., Fanara, J. J. and Morgan, T. J. (2011). Genetic variation in heat-stress tolerance among South American Drosophila populations. Genetica 139: 1331-1337. PubMed ID: 22350564

  • Takako Morimoto Tokyo University of Pharmacy and Life Sciences
    Akiba, M., Sugimoto, K., Aoki, R., Murakami, R., Miyashita, T., Hashimoto, R., Hiranuma, A., Yamauchi, J., Ueno, T. and Morimoto, T. (2019). Dopamine modulates the optomotor response to unreliable visual stimuli in Drosophila melanogaster. Eur J Neurosci. PubMed ID: 31834948

    Matsumoto, Y., Shimizu, K., Arahata, K., Suzuki, M., Shimizu, A., Takei, K., Yamauchi, J., Hakeda-Suzuki, S., Suzuki, T. and Morimoto, T. (2018). Prepulse inhibition in Drosophila melanogaster larvae. Biol Open 7(9). PubMed ID: 30262549

    Koseki, N., Mori, S., Suzuki, S., Tonooka, Y., Kosugi, S., Miyakawa, H. and Morimoto, T. (2016). Individual differences in sensory responses influence decision making by Drosophila melanogaster larvae on exposure to contradictory cues. J Neurogenet: 1-39. PubMed ID: 27309770

    Suzuki, Y., Ikeda, H., Miyamoto, T., Miyakawa, H., Seki, Y., Aonishi, T. and Morimoto, T. (2015). Noise-robust recognition of wide-field motion direction and the underlying neural mechanisms in Drosophila melanogaster. Sci Rep 5: 10253. PubMed ID: 25974721

    Homma, M., Nagashima, S., Fukuda, T., Yanagi, S., Miyakawa, H., Suzuki, E. and Morimoto, T. (2014). Downregulation of Centaurin gamma1A increases synaptic transmission at Drosophila larval neuromuscular junctions. Eur J Neurosci. PubMed ID: 25074496

    Kikuchi, A., Ohashi, S., Fuse, N., Ohta, T., Suzuki, M., Suzuki, Y., Fujita, T., Miyamoto, T., Aonishi, T., Miyakawa, H. and Morimoto, T. (2013). Experience-dependent Plasticity of the Optomotor Response in Drosophila melanogaster. Dev Neurosci. PubMed ID: 23406844

    Fukui, A., Inaki, M., Tonoe, G., Hamatani, H., Homma, M., Morimoto, T., Aburatani, H. and Nose, A. (2012). Lola regulates glutamate receptor expression at the Drosophila neuromuscular junction. Biol Open 1: 362-375. PubMed ID: 23213426

    Miyashita, T., Oda, Y., Horiuchi, J., Yin, J. C., Morimoto, T. and Saitoe, M. (2012). Mg(2+) block of Drosophila NMDA receptors is required for long-term memory formation and CREB-dependent gene expression. Neuron 74: 887-898. PubMed ID: 22681692

  • Ted Morrow School of Life Sciences, University of Sussex
    Innocenti, P., Flis, I. and Morrow, E. H. (2014). Female responses to experimental removal of sexual selection components in Drosophila melanogaster. BMC Evol Biol 14: 239. PubMed ID: 25406540

    Abbott, J. K., Innocenti, P., Chippindale, A. K. and Morrow, E. H. (2013). Epigenetics and Sex-Specific Fitness: An Experimental Test Using Male-Limited Evolution in Drosophila melanogaster. PLoS One 8: e70493. PubMed ID: 23922998

    Bailey, R. I., Innocenti, P., Morrow, E. H., Friberg, U. and Qvarnstrom, A. (2011). Female Drosophila melanogaster gene expression and mate choice: the X chromosome harbours candidate genes underlying sexual isolation. PLoS One 6: e17358. PubMed ID: 21386982

    Innocenti, P. and Morrow, E. H. (2010). The sexually antagonistic genes of Drosophila melanogaster. PLoS Biol 8: e1000335. PubMed ID: 20305719

  • Nathan Mortimer School of Biological Sciences, Illinois State University, Normal
    Kr, P., Lee, J. and Mortimer, N. T. (2021). The S1A protease family members CG10764 and CG4793 regulate cellular immunity in Drosophila. MicroPubl Biol 2021. PubMed ID: 33644706

    Trainor, J. E., Kr, P. and Mortimer, N. T. (2021). Immune Cell Production Is Targeted by Parasitoid Wasp Virulence in a Drosophila-Parasitoid Wasp Interaction. Pathogens 10(1). PubMed ID: 33429864

    Ryan, S. M., Wildman, K., Oceguera-Perez, B., Barbee, S., Mortimer, N. T. and Vrailas-Mortimer, A. D. (2020). Evolutionarily conserved transcription factors drive the oxidative stress response in Drosophila. J Exp Biol 223(Pt 14). PubMed ID: 32532866

    Schachtner, L. T., Sola, I. E., Forand, D., Antonacci, S., Postovit, A. J., Mortimer, N. T., Killian, D. J. and Olesnicky, E. C. (2015). Drosophila Shep and C. elegans SUP-26 are RNA-binding proteins that play diverse roles in nervous system development. Dev Genes Evol 225(6): 319-330. PubMed ID: 26271810

  • David Morton Integrative Biosciences, Oregon Health & Science University
    Lembke, K. M., Law, A. D., Ahrar, J. and Morton, D. B. (2018). Deletion of a specific exon in the voltage-gated calcium channel, cacophony, causes disrupted locomotion in Drosophila larvae. J Exp Biol. PubMed ID: 30397173

    Lembke, K. M., Scudder, C. and Morton, D. B. (2017). Restoration of motor defects caused by loss of Drosophila TDP-43 by expression of the voltage-gated calcium channel, Cacophony, in central neurons. J Neurosci 37(39): 9486-9497. PubMed ID: 28847811

    Chang, J. C. and Morton, D. B. (2017). Drosophila lines with mutant and wild type human TDP-43 replacing the endogenous gene reveals phosphorylation and ubiquitination in mutant lines in the absence of viability or lifespan defects. PLoS One 12(7): e0180828. PubMed ID: 28686708

    Chang, J. C., Hazelett, D. J., Stewart, J. A. and Morton, D. B. (2013). Motor neuron expression of the voltage-gated calcium channel cacophony restores locomotion defects in a Drosophila, TDP-43 loss of function model of ALS. Brain Res. PubMed ID: 24275199

    Hazelett, D. J., Chang, J. C., Lakeland, D. L. and Morton, D. B. (2012). Comparison of parallel high-throughput RNA sequencing between knockout of TDP-43 and its overexpression reveals primarily nonreciprocal and nonoverlapping gene expression changes in the central nervous system of Drosophila. G3 (Bethesda) 2: 789-802. PubMed ID: 22870402

    Vermehren-Schmaedick, A., Scudder, C., Timmermans, W. and Morton, D. B. (2011). Drosophila gustatory preference behaviors require the atypical soluble guanylyl cyclases. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 197: 717-727. PubMed ID: 21350862

    Morton, D. B. (2011). Behavioral responses to hypoxia and hyperoxia in Drosophila larvae: molecular and neuronal sensors. Fly (Austin) 5: 119-125. PubMed ID: 21150317

  • Tim Mosca Department of Neuroscience, Jefferson University, Philadelphia, Pa
    Sapiro, A. L., Freund, E. C., Restrepo, L., Qiao, H. H., Bhate, A., Li, Q., Ni, J. Q., Mosca, T. J. and Li, J. B. (2020). Zinc Finger RNA-Binding Protein Zn72D Regulates ADAR-Mediated RNA Editing in Neurons. Cell Rep 31(7): 107654. PubMed ID: 32433963

    DePew, A. T., Aimino, M. A. and Mosca, T. J. (2019). The Tenets of Teneurin: Conserved Mechanisms Regulate Diverse Developmental Processes in the Drosophila Nervous System. Front Neurosci 13: 27. PubMed ID: 30760977

    Mosca, T. J., Luginbuhl, D. J., Wang, I. E. and Luo, L. (2017). Presynaptic LRP4 promotes synapse number and function of excitatory CNS neurons. Elife 6. PubMed ID: 28606304

    Sakuma, C., Saito, Y., Umehara, T., Kamimura, K., Maeda, N., Mosca, T. J., Miura, M. and Chihara, T. (2016). The Strip-Hippo Pathway Regulates Synaptic Terminal Formation by Modulating Actin Organization at the Drosophila Neuromuscular Synapses. Cell Rep 16(9): 2289-2297. PubMed ID: 27545887

  • Alexey Moskalev Syktyvkar State University Komi Republic, Russian Federation
    Shaposhnikov, M. V., Zakluta, A. S., Zemskaya, N. V., Guvatova, Z. G., Shilova, V. Y., Yakovleva, D. V., Gorbunova, A. A., Koval, L. A., Ulyasheva, N. S., Evgen'ev, M. B., Zatsepina, O. G. and Moskalev, A. A. (2022). Deletions of the cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE) genes, involved in the control of hydrogen sulfide biosynthesis, significantly affect lifespan and fitness components of Drosophila melanogaster. Mech Ageing Dev 203: 111656. PubMed ID: 35247392

    Proshkina, E., Yushkova, E., Koval, L., Zemskaya, N., Shchegoleva, E., Solovev, I., Yakovleva, D., Pakshina, N., Ulyasheva, N., Shaposhnikov, M. and Moskalev, A. (2021). Tissue-Specific Knockdown of Genes of the Argonaute Family Modulates Lifespan and Radioresistance in Drosophila Melanogaster. Int J Mol Sci 22(5). PubMed ID: 33673647

    Shaposhnikov, M. V., Zemskaya, N. V., Koval, L., Minnikhanova, N. R., Kechko, O. I., Mitkevich, V. A., Makarov, A. A. and Moskalev, A. (2020). Amyloid-beta peptides slightly affect lifespan or antimicrobial peptide gene expression in Drosophila melanogaster. BMC Genet 21(Suppl 1): 65. PubMed ID: 33092519

    Koval, L., Proshkina, E., Shaposhnikov, M. and Moskalev, A. (2019). The role of DNA repair genes in radiation-induced adaptive response in Drosophila melanogaster is differential and conditional. Biogerontology. PubMed ID: 31624983

    Solovev, I., Shegoleva, E., Fedintsev, A., Shaposhnikov, M. and Moskalev, A. (2019). Circadian clock genes' overexpression in Drosophila alters diet impact on lifespan. Biogerontology 20(2): 159-170. PubMed ID: 30470951

    Solovev, I., Dobrovolskaya, E., Shaposhnikov, M., Sheptyakov, M. and Moskalev, A. (2019). Neuron-specific overexpression of core clock genes improves stress-resistance and extends lifespan of Drosophila melanogaster. Exp Gerontol 117: 61-71. PubMed ID: 30415070

    Solovev, I., Shegoleva, E., Fedintsev, A., Shaposhnikov, M. and Moskalev, A. (2018). Circadian clock genes' overexpression in Drosophila alters diet impact on lifespan. Biogerontology. PubMed ID: 30470951

    Shaposhnikov, M., Proshkina, E., Shilova, L., Zhavoronkov, A. and Moskalev, A. (2015). Lifespan and stress resistance in Drosophila with overexpressed DNA repair genes. Sci Rep 5: 15299. PubMed ID: 26477511

    Zhikrevetskaya, S., Peregudova, D., Danilov, A., Plyusnina, E., Krasnov, G., Dmitriev, A., Kudryavtseva, A., Shaposhnikov, M. and Moskalev, A. (2015). Effect of Low Doses (5-40 cGy) of Gamma-irradiation on Lifespan and Stress-related Genes Expression Profile in Drosophila melanogaster. PLoS One 10: e0133840. PubMed ID: 26248317

    Omelyanchuk, L. V., Shaposhnikov, M. V. and Moskalev, A. A. (2015). Drosophila nervous system as a target of aging and anti-aging interventions. Front Genet 6: 89. PubMed ID: 25806047

  • Bernard Moussian University of Tubingen
    Zuber, R., Wang, Y., Gehring, N., Bartoszewski, S. and Moussian, B. (2020). Tweedle proteins form extracellular two-dimensional structures defining body and cell shape in Drosophila melanogaster. Open Biol 10(12): 200214. PubMed ID: 33292106

    Lerch, S., Zuber, R., Gehring, N., Wang, Y., Eckel, B., Klass, K. D., Lehmann, F. O. and Moussian, B. (2020). Resilin matrix distribution, variability and function in Drosophila. BMC Biol 18(1): 195. PubMed ID: 33317537

    Wang, Y., Farine, J. P., Yang, Y., Yang, J., Tang, W., Gehring, N., Ferveur, J. F. and Moussian, B. (2020). Transcriptional Control of Quality Differences in the Lipid-Based Cuticle Barrier in Drosophila suzukii and Drosophila melanogaster. Front Genet 11: 887. PubMed ID: 32849846

    Dong, W., Gao, Y. H., Zhang, X. B., Moussian, B. and Zhang, J. Z. (2020). Chitinase10 controls chitin amounts and organization in the wing cuticle of Drosophila. Insect Sci. PubMed ID: 32129536

    Wang, Y., Norum, M., Oehl, K., Yang, Y., Zuber, R., Yang, J., Farine, J. P., Gehring, N., Flotenmeyer, M., Ferveur, J. F. and Moussian, B. (2020). Dysfunction of Oskyddad causes Harlequin-type ichthyosis-like defects in Drosophila melanogaster. PLoS Genet 16(1): e1008363. PubMed ID: 31929524

    Dong, W., Dobler, R., Dowling, D. K. and Moussian, B. (2019). The cuticle inward barrier in Drosophila melanogaster is shaped by mitochondrial and nuclear genotypes and a sex-specific effect of diet. PeerJ 7: e7802. PubMed ID: 31592352

    Zhang, M., Ji, Y., Zhang, X., Ma, P., Wang, Y., Moussian, B. and Zhang, J. (2019). The putative chitin deacetylases serpentine and vermiform have non-redundant functions during Drosophila wing development. Insect Biochem Mol Biol. PubMed ID: 31108167

    Wang, Y., Maier, A., Gehring, N. and Moussian, B. (2019). Inhibition of fatty acid desaturation impairs cuticle differentiation in Drosophila melanogaster. Arch Insect Biochem Physiol: e21535. PubMed ID: 30672604

    Wang, Y., Berger, J. and Moussian, B. (2018). Trynity models a tube valve in the Drosophila larval airway system. Dev Biol [Epub ahead of print]. PubMed ID: 29518377

    Zuber, R., Norum, M., Wang, Y., Oehl, K., Gehring, N., Accardi, D., Bartozsewski, S., Berger, J., Flotenmeyer, M. and Moussian, B. (2017). The ABC transporter Snu and the extracellular protein Snsl cooperate in the formation of the lipid-based inward and outward barrier in the skin of Drosophila. Eur J Cell Biol. PubMed ID: 29306642

  • Amritpal Mudher Centre for Biological Sciences, University of Southampton
    Chatterjee, S., Ambegaokar, S. S., Jackson, G. R. and Mudher, A. (2019). Insulin-mediated changes in Tau hyperphosphorylation and autophagy in a Drosophila model of tauopathy and neuroblastoma cells. Front Neurosci 13: 801. PubMed ID: 31427921

    Quraishe, S., Cowan, C. M., Mudher, A. (2013) NAP (davunetide) rescues neuronal dysfunction in a Drosophila model of tauopathy. Mol Psychiatry. PubMed ID: 23587881

    Cowan, C. M., Quraishe, S. and Mudher, A. (2012). What is the pathological significance of tau oligomers? Biochem Soc Trans 40: 693-697. PubMed ID: 22817718

    Cowan, C. M., Sealey, M. A., Quraishe, S., Targett, M. T., Marcellus, K., Allan, D. and Mudher, A. (2011). Modelling tauopathies in Drosophila: insights from the fruit fly. Int J Alzheimers Dis 2011: 598157. PubMed ID: 22254145

    Sinadinos, C., Cowan, C. M., Wyttenbach, A. and Mudher, A. (2012). Increased throughput assays of locomotor dysfunction in Drosophila larvae. J Neurosci Methods 203: 325-334. PubMed ID: 21925540

  • Ashim Mukherjee Banaras Hindu University, Varanasi
    Maurya, B., Surabhi, S., Das, R., Pandey, P., Mukherjee, A. and Mutsuddi, M. (2021). Maheshvara regulates JAK/STAT signaling by interacting and stabilizing hopscotch transcripts which leads to apoptosis in Drosophila melanogaster. Cell Death Dis 12(4): 363. PubMed ID: 33824299

    Sharma, V., Mutsuddi, M. and Mukherjee, A. (2020). Deltex cooperates with TRAF6 to promote apoptosis and cell migration through Eiger-independent JNK activation in Drosophila. Cell Biol Int. PubMed ID: 33300258

    Singh, A., Paul, M. S., Dutta, D., Mutsuddi, M. and Mukherjee, A. (2019). Regulation of Notch signaling by a chromatin modeling protein Hat-trick. Development. PubMed ID: 31142544

    Dutta, D., Mutsuddi, M. and Mukherjee, A. (2019). Synergistic interaction of Deltex and Hrp48 leads to JNK activation. Cell Biol Int 43(3): 350-357. PubMed ID: 30597717

    Dutta, D., Mutsuddi, M. and Mukherjee, A. (2018). Synergistic interaction of Deltex and Hrp48 leads to JNK activation. Cell Biol Int. PubMed ID: 30597717

    Paul, M. S., Dutta, D., Singh, A., Mutsuddi, M. and Mukherjee, A. (2018). Regulation of Notch signaling in the developing Drosophila eye by a T-box containing transcription factor, Dorsocross. Genesis. PubMed ID: 30246928

    Dutta, D., Singh, A., Paul, M. S., Sharma, V., Mutsuddi, M. and Mukherjee, A. (2018). Deltex interacts with Eiger and consequently influences the cell death in Drosophila melanogaster. Cell Signal 49: 17-29. PubMed ID: 29775737

    Paul, M. S., Singh, A., Dutta, D., Mutsuddi, M. and Mukherjee, A. (2018). Notch signals modulate lgl mediated tumorigenesis by the activation of JNK signaling. BMC Res Notes 11(1): 247. PubMed ID: 29661224

    Singh, A., Dutta, D., Paul, M. S., Verma, D., Mutsuddi, M. and Mukherjee, A. (2018). Pleiotropic functions of the chromodomain-containing protein Hat-trick during oogenesis in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 29367451

    Tripathi, B. K., Das, R., Mukherjee, A. and Mutsuddi, M. (2017). Interaction of Spoonbill with Prospero in Drosophila: Implications in neuroblast development. Genesis. PubMed ID: 28722203

    Dutta, D., Paul, M. S., Singh, A., Mutsuddi, M. and Mukherjee, A. (2017). Regulation of Notch signaling by the Heterogeneous Nuclear Ribonucleoprotein Hrp48 and Deltex in Drosophila melanogaster. Genetics. PubMed ID: 28396507

  • Arno Muller Cell and Developmental Biology, College of Life Sciences, University of Dundee
    Bandarra, D., Biddlestone, J., Mudie, S., Muller, H. A. and Rocha, S. (2014). HIF-1alpha restricts NF-kappaB dependent gene expression to control innate immunity signals. Dis Model Mech. PubMed ID: 25510503

    Hain, D., Langlands, A., Sonnenberg, H. C., Bailey, C., Bullock, S. L. and Muller, H. A. (2014). The Drosophila MAST kinase Drop out is required to initiate membrane compartmentalisation during cellularisation and regulates dynein-based transport. Development 141: 2119-2130. PubMed ID: 24803657

    Muha, V., Muller, H. A. (2013) Functions and Mechanisms of Fibroblast Growth Factor (FGF) Signalling in Drosophila melanogaster. Int J Mol Sci 14: 5920-5937. PubMed ID: 23493057

    Mariappa, D., Sauert, K., Marino, K., Turnock, D., Webster, R., van Aalten, D. M., Ferguson, M. A. and Muller, H. A. (2011). Protein O-GlcNAcylation is required for fibroblast growth factor signaling in Drosophila. Sci Signal 4: ra89. PubMed ID: 22375049

    Winklbauer, R. and Muller, H. A. (2011). Mesoderm layer formation in Xenopus and Drosophila gastrulation. Phys Biol 8: 045001. PubMed ID: 21750361

  • Jürg Müller Max Planck Institute of Biochemistry, Martinsried
    Elizarev, P., Finkl, K. and Müller, J. (2021). Distinct requirements for Pho, Sfmbt, and Ino80 for cell survival in Drosophila. Genetics 219(1). PubMed ID: 34849913

    Finogenova, K., Bonnet, J., Poepsel, S., Schäfer, I. B., Finkl, K., Schmid, K., Litz, C., Strauss, M., Benda, C. and Muller, J. (2020). Structural basis for PRC2 decoding of active histone methylation marks H3K36me2/3. Elife 9. PubMed ID: 33211010

    Bonnet, J., Lindeboom, R. G. H., Pokrovsky, D., Stricker, G., Celik, M. H., Rupp, R. A. W., Gagneur, J., Vermeulen, M., Imhof, A. and Muller, J. (2019). Quantification of proteins and histone marks in Drosophila embryos reveals stoichiometric relationships impacting chromatin regulation. Dev Cell. PubMed ID: 31630981

    De, I., Chittock, E. C., Grotsch, H., Miller, T. C. R., McCarthy, A. A. and Muller, C. W. (2018). Structural basis for the activation of the deubiquitinase Calypso by the Polycomb protein ASX. Structure. PubMed ID: 30639226

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

    Schmahling, S., Meiler, A., Lee, Y., Mohammed, A., Finkl, K., Tauscher, K., Israel, L., Borath, M., Philippou-Massier, J., Blum, H., Habermann, B., Imhof, A., Song, J. J. and Muller, J. (2018). Regulation and function of H3K36 di-methylation by the trithorax-group protein complex AMC. Development. PubMed ID: 29540501

    Copur, O. and Muller, J. (2018). Histone demethylase activity of Utx is essential for viability and regulation of HOX gene expression in Drosophila. Genetics 208(2): 633-637. PubMed ID: 29247011

    Laprell, F., Finkl, K. and Muller, J. (2017). Propagation of Polycomb-repressed chromatin requires sequence-specific recruitment to DNA. Science [Epub ahead of print]. PubMed ID: 28302792

    Frey, F., Sheahan, T., Finkl, K., Stoehr, G., Mann, M., Benda, C. and Muller, J. (2016). Molecular basis of PRC1 targeting to Polycomb response elements by PhoRC. Genes Dev 30: 1116-1127. PubMed ID: 27151979

    Pengelly, A. R., Kalb, R., Finkl, K. and Muller, J. (2015). Transcriptional repression by PRC1 in the absence of H2A monoubiquitylation. Genes Dev 29: 1487-1492. PubMed ID: 26178786

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

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

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

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

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

    Murray, M. J., Southall, T. D., Liu, W., Fraval, H., Lorensuhewa, N., Brand, A. H. and Saint, R. (2012). Snail-dependent repression of the RhoGEF pebble is required for gastrulation consistency in Drosophila melanogaster. Dev Genes Evol 222: 361-368. PubMed ID: 22945369

    Murray, M. J. and Saint, R. (2011). Imaging pluripotent cell migration in Drosophila. Methods Mol Biol 750: 169-184. PubMed ID: 21618091

  • Mala Murthy Princeton Neuroscience Institute and Department of Molecular Biology
    Pacheco, D. A., Thiberge, S. Y., Pnevmatikakis, E. and Murthy, M. (2021). Auditory activity is diverse and widespread throughout the central brain of Drosophila. Nat Neurosci 24(1): 93-104. PubMed ID: 33230320

    Deutsch, D., Pacheco, D., Encarnacion-Rivera, L., Pereira, T., Fathy, R., Clemens, J., Girardin, C., Calhoun, A., Ireland, E., Burke, A., Dorkenwald, S., McKellar, C., Macrina, T., Lu, R., Lee, K., Kemnitz, N., Ih, D., Castro, M., Halageri, A., Jordan, C., Silversmith, W., Wu, J., Seung, H. S. and Murthy, M. (2020). The neural basis for a persistent internal state in Drosophila females. Elife 9. PubMed ID: 33225998

    Calhoun, A. J., Pillow, J. W. and Murthy, M. (2019). Unsupervised identification of the internal states that shape natural behavior. Nat Neurosci 22(12): 2040-2049. PubMed ID: 31768056

    Deutsch, D., Clemens, J., Thiberge, S. Y., Guan, G. and Murthy, M. (2019). Shared song detector neurons in Drosophila male and female brains drive sex-specific behaviors. Curr Biol 29(19): 3200-3215.e3205. PubMed ID: 31564492

    Clemens, J., Coen, P., Roemschied, F. A., Pereira, T. D., Mazumder, D., Aldarondo, D. E., Pacheco, D. A. and Murthy, M. (2018). Discovery of a new song mode in Drosophila reveals hidden structure in the sensory and neural drivers of behavior. Curr Biol 28(15): 2400-2412.e2406. PubMed ID: 30057309

    Clemens, J., Ozeri-Engelhard, N. and Murthy, M. (2018). Fast intensity adaptation enhances the encoding of sound in Drosophila. Nat Commun 9(1): 134. PubMed ID: 29317624

    Stern, D. L., Clemens, J., Coen, P., Calhoun, A. J., Hogenesch, J. B., Arthur, B. J. and Murthy, M. (2017). Experimental and statistical reevaluation provides no evidence for Drosophila courtship song rhythms. Proc Natl Acad Sci U S A 114(37): 9978-9983. PubMed ID: 28851830

    Crocker, A., Guan, X.J., Murphy, C.T. and Murthy, M. (2016). Cell-type-specific transcriptome analysis in the Drosophila mushroom body reveals memory-related changes in gene expression. Cell Rep [Epub ahead of print]. PubMed ID: 27160913

    Coen, P., Xie, M., Clemens, J. and Murthy, M. (2016). Sensorimotor transformations underlying variability in song intensity during Drosophila courtship. Neuron 89: 629-644. PubMed ID: 26844835

    Coen, P., Xie, M., Clemens, J. and Murthy, M. (2016). Sensorimotor transformations underlying variability in song intensity during Drosophila courtship. Neuron 89: 629-644. PubMed ID: 26844835

  • Marc Muskavitch Biology Department, Boston College
    Regna, K., Kurshan, P. T., Harwood, B. N., Jenkins, A. M., Lai, C. Q., Muskavitch, M. A., Kopin, A. S. and Draper, I. (2016). A critical role for the Drosophila dopamine D1-like receptor Dop1R2 at the onset of metamorphosis. BMC Dev Biol 16: 15. PubMed ID: 27184815

    Daskalaki, A., Shalaby, N. A., Kux, K., Tsoumpekos, G., Tsibidis, G. D., Muskavitch, M. A. and Delidakis, C. (2011). Distinct intracellular motifs of Delta mediate its ubiquitylation and activation by Mindbomb1 and Neuralized. J Cell Biol 195: 1017-1031. PubMed ID: 22162135

    Kang, K., Panzano, V. C., Chang, E. C., Ni, L., Dainis, A. M., Jenkins, A. M., Regna, K., Muskavitch, M. A. and Garrity, P. A. (2012). Modulation of TRPA1 thermal sensitivity enables sensory discrimination in Drosophila. Nature 481: 76-80. PubMed ID: 22139422

  • Mousumi Mutsuddi Molecular and Human Genetics, Banaras Hindu University, Varanasi
    Maurya, B., Surabhi, S., Das, R., Pandey, P., Mukherjee, A. and Mutsuddi, M. (2021). Maheshvara regulates JAK/STAT signaling by interacting and stabilizing hopscotch transcripts which leads to apoptosis in Drosophila melanogaster. Cell Death Dis 12(4): 363. PubMed ID: 33824299

    Tripathi, B. K., Surabhi, S., Bhaskar, P. K., Mukherjee, A. and Mutsuddi, M. (2016). The RNA binding KH domain of Spoonbill depletes pathogenic non-coding spinocerebellar ataxia 8 transcripts and suppresses neurodegeneration in Drosophila. Biochim Biophys Acta 1862: 1732-1741. PubMed ID: 27302466

    Surabhi, S., Tripathi, B. K., Maurya, B., Bhaskar, P. K., Mukherjee, A. and Mutsuddi, M. (2015). Regulation of Notch Signalling by an Evolutionary Conserved DEAD Box RNA Helicase, Maheshvara in Drosophila melanogaster. Genetics. PubMed ID: 26400611

    Sachan, N., Mishra, A. K., Mutsuddi, M. and Mukherjee, A. (2015). Chip physically interacts with Notch and their stoichiometry is critical for Notch function in wing development and cell proliferation in Drosophila. Biochim Biophys Acta 1850: 802-812. PubMed ID: 25597954

    Mishra, A. K., Sachan, N., Mutsuddi, M. and Mukherjee, A. (2014). TRAF6 is a novel regulator of Notch signaling in Drosophila melanogaster. Cell Signal 26: 3016-3026. PubMed ID: 25280943

  • Monn Monn Myat Department of Cell & Developmental Biology, Weill Cornell Medical College, New York
    Patel, U. and Myat, M. M. (2013). Receptor guanylyl cyclase Gyc76C is required for invagination, collective migration and lumen shape in the Drosophila embryonic salivary gland. Biol Open 2: 711-717. PubMed ID: 23862019

    Pirraglia, C., Walters, J., Ahn, N., Myat, M. M. (2013) Rac1 GTPase acts downstream of alphaPS1betaPS integrin to control collective migration and lumen size in the Drosophila salivary gland. Dev Biol. PubMed ID: 23500171

    Patel, U., Davies, S. A. and Myat, M. M. (2012). Receptor-type guanylyl cyclase Gyc76C is required for development of the Drosophila embryonic somatic muscle. Biol Open 1: 507-515. PubMed ID: 23213443

    Xu, N., Bagumian, G., Galiano, M. and Myat, M. M. (2011). Rho GTPase controls Drosophila salivary gland lumen size through regulation of the actin cytoskeleton and Moesin. Development 138: 5415-5427. PubMed ID: 22071107

    Zhan, Y., Maung, S. W., Shao, B. and Myat, M. M. (2010). The bHLH transcription factor, hairy, refines the terminal cell fate in the Drosophila embryonic trachea. PLoS One 5: e14134. PubMed ID: 21152432


  • Return to The Interactive Fly