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


  • Clemens Cabernard Neurobiology Biozentrum, University of Basel
    Sunchu, B., Lee, N. M., Taylor, J. A., Segura, R. C., Roubinet, C. and Cabernard, C. (2022). Asymmetric chromatin retention and nuclear envelopes separate chromosomes in fused cells in vivo. Commun Biol 5(1): 953. PubMed ID: 36123528

    Gallaud, E., Ramdas Nair, A., Horsley, N., Monnard, A., Singh, P., Pham, T. T., Salvador Garcia, D., Ferrand, A. and Cabernard, C. (2020). Dynamic centriolar localization of Polo and Centrobin in early mitosis primes centrosome asymmetry. PLoS Biol 18(8): e3000762. PubMed ID: 32760088

    Tsankova, A., Pham, T. T., Garcia, D. S., Otte, F. and Cabernard, C. (2017). Cell polarity regulates biased myosin activity and dynamics during asymmetric cell division via Drosophila Rho kincase and Protein kinase N. Dev Cell 42(2): 143-155.e145. PubMed ID: 28712722

    Ramdas Nair, A., Singh, P., Salvador Garcia, D., Rodriguez-Crespo, D., Egger, B. and Cabernard, C. (2016). The microcephaly-associated protein Wdr62/CG7337 is required to maintain centrosome asymmetry in Drosophila neuroblasts. Cell Rep. PubMed ID: 26804909

    Roth, M., Roubinet, C., Iffländer, N., Ferrand, A. and Cabernard, C. (2015). Asymmetrically dividing Drosophila neuroblasts utilize two spatially and temporally independent cytokinesis pathways. Nat Commun 6: 6551. PubMed ID: 25791062

    Singh, P., Ramdas Nair, A. and Cabernard, C. (2014). The Centriolar Protein Bld10/Cep135 Is Required to Establish Centrosome Asymmetry in Drosophila Neuroblasts. Curr Biol. PubMed ID: 24954048

    Cabernard, C. and Doe, C. Q. (2013). Live imaging of neuroblast lineages within intact larval brains in Drosophila. Cold Spring Harb Protoc 2013: 970-977. PubMed ID: 24086057

    Cabernard, C. (2012). Cytokinesis in Drosophila melanogaster. Cytoskeleton (Hoboken) 69: 791-809. PubMed ID: 22888045

    Connell, M., Cabernard, C., Ricketson, D., Doe, C. Q. and Prehoda, K. E. (2011). Asymmetric cortical extension shifts cleavage furrow position in Drosophila neuroblasts. Mol Biol Cell 22: 4220-4226. PubMed ID: 21937716

  • Kenneth Cadigan Molecular, Cellular, and Developmental Biology, University of Michigan
    Zhang, C. U. and Cadigan, K. M. (2017). The matrix protein Tiggrin regulates plasmatocyte maturation in Drosophila larva. Development. PubMed ID: 28526755

    Archbold, H. C., Broussard, C., Chang, M. V. and Cadigan, K. M. (2014). Bipartite Recognition of DNA by TCF/Pangolin Is Remarkably Flexible and Contributes to Transcriptional Responsiveness and Tissue Specificity of Wingless Signaling. PLoS Genet 10: e1004591. PubMed ID: 25188465

    Zhang, C. U., Blauwkamp, T. A., Burby, P. E. and Cadigan, K. M. (2014). Wnt-Mediated Repression via Bipartite DNA Recognition by TCF in the Drosophila Hematopoietic System. PLoS Genet 10: e1004509. PubMed ID: 25144371

    Bhambhani, C., Ravindranath, A. J., Mentink, R. A., Chang, M. V., Betist, M. C., Yang, Y. X., Koushika, S. P., Korswagen, H. C. and Cadigan, K. M. (2014). Distinct DNA Binding Sites Contribute to the TCF Transcriptional Switch in C. elegans and Drosophila. PLoS Genet 10: e1004133. PubMed ID: 24516405

    Ravindranath, A. and Cadigan, K. M. (2014). Structure-Function Analysis of the C-clamp of TCF/Pangolin in Wnt/ss-catenin Signaling. PLoS One 9: e86180. PubMed ID: 24465946

    Cadigan, K. M. (2012). TCFs and Wnt/beta-catenin signaling: more than one way to throw the switch. Curr Top Dev Biol 98: 1-34. PubMed ID: 22305157

    Kennell, J. A., Cadigan, K. M., Shakhmantsir, I. and Waldron, E. J. (2012). The microRNA miR-8 is a positive regulator of pigmentation and eclosion in Drosophila. Dev Dyn 241: 161-168. PubMed ID: 22174085

    Bhambhani, C., Chang, J. L., Akey, D. L. and Cadigan, K. M. (2011). The oligomeric state of CtBP determines its role as a transcriptional co-activator and co-repressor of Wingless targets. EMBO J 30: 2031-2043. PubMed ID: 21468031

  • Ross Cagan Mount Sinai Medical Center
    Das, T. K., Gatto, J., Mirmira, R., Hourizadeh, E., Kaufman, D., Gelb, B. D. and Cagan, R. (2021). Drosophila RASopathy models identify disease subtype differences and biomarkers of drug efficacy. iScience 24(4): 102306. PubMed ID: 33855281

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

    Padash Barmchi, M., Thomas, M., Thatte, J. V., Vats, A., Zhang, B., Cagan, R. L. and Banks, L. (2021). Inhibition of kinase IKKbeta suppresses cellular abnormalities induced by the human papillomavirus oncoprotein HPV 18E6. Sci Rep 11(1): 1111. PubMed ID: 33441820

    Ung, P. M. U., Sonoshita, M., Scopton, A. P., Dar, A. C., Cagan, R. L. and Schlessinger, A. (2019). Integrated computational and Drosophila cancer model platform captures previously unappreciated chemicals perturbing a kinase network. PLoS Comput Biol 15(4): e1006878. PubMed ID: 31026276

    Das, T. K., Esernio, J. and Cagan, R. (2018). Restraining network response to targeted cancer therapies improves efficacy and reduces cellular resistance. Cancer Res. PubMed ID: 29844121

    Das, T. K. and Cagan, R. L. (2017). KIF5B-RET oncoprotein signals through a multi-kinase signaling hub. Cell Rep 20(10): 2368-2383. PubMed ID: 28877471

    Bangi, E., Murgia, C., Teague, A.G., Sansom, O.J. and Cagan, R.L. (2016). Functional exploration of colorectal cancer genomes using Drosophila. Nat Commun 7: 13615. PubMed ID: 27897178 Levinson, S. and Cagan, R. L. (2016). Drosophila Cancer Models Identify Functional Differences between Ret Fusions. Cell Rep 16: 3052-3061. PubMed ID: 27626672

    Levine, B. D. and Cagan, R. L. (2016). Drosophila lung cancer models identify Trametinib plus Statin as candidate therapeutic. Cell Rep [Epub ahead of print]. PubMed ID: 26832408

    Hirabayashi, S. and Cagan, R. L. (2015). Salt-inducible kinases mediate nutrient-sensing to link dietary sugar and tumorigenesis in Drosophila. Elife 4 [Epub ahead of print]. PubMed ID: 26573956

    Na, J., Sweetwyne, M. T., Park, A. S., Susztak, K. and Cagan, R. L. (2015). Diet-Induced Podocyte Dysfunction in Drosophila and Mammals. Cell Rep 12: 636-647. PubMed ID: 26190114

  • Haini Cai Cellular Biology, University of Georgia
    Li, M., Zhao, Q., Belloli, R., Duffy, C. R. and Cai, H. N. (2021). Insulator foci distance correlates with cellular and nuclear morphology in early Drosophila embryos. Dev Biol 476: 189-199. PubMed ID: 33844976

    Li, M., Ma, Z., Liu, J. K., Roy, S., Patel, S. K., Lane, D. C. and Cai, H. N. (2015). An Organizational Hub of Developmentally Regulated Chromatin Loops in the Drosophila Antennapedia Complex. Mol Cell Biol. PubMed ID: 26391952

    Li, M., Belozerov, V. E. and Cai, H. N. (2010). Modulation of chromatin boundary activities by nucleosome-remodeling activities in Drosophila melanogaster. Mol Cell Biol 30: 1067-1076. PubMed ID: 19995906

  • Yu Cai Temasek Life Sciences Laboratory, Singapore
    Liu, S., Baeg, G. H., Yang, Y., Goh, F. G., Bao, H., Wagner, E. J., Yang, X. and Cai, Y. (2023). The Integrator complex desensitizes cellular response to TGF-β/BMP signaling. Cell Rep 42(1): 112007. PubMed ID: 36641752

    Yang, Y., Kong, R., Goh, F. G., Somers, W. G., Hime, G. R., Li, Z. and Cai, Y. (2021). dRTEL1 is essential for the maintenance of Drosophila male germline stem cells. PLoS Genet 17(10): e1009834. PubMed ID: 34644293

    Shan, Z., Tu, Y., Yang, Y., Liu, Z., Zeng, M., Xu, H., Long, J., Zhang, M., Cai, Y. and Wen, W. (2018). Basal condensation of Numb and Pon complex via phase transition during Drosophila neuroblast asymmetric division. Nat Commun 9(1): 737. PubMed ID: 29467404

    Luo, L., Siah, C. K. and Cai, Y. (2017). Engrailed acts with Nejire to control decapentaplegic expression in the Drosophila ovarian stem cell niche. Development 144(18): 3224-3231. PubMed ID: 28928281

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

    Luo, L., Wang, H., Fan, C., Liu, S. and Cai, Y. (2015). Wnt ligands regulate Tkv expression to constrain Dpp activity in the Drosophila ovarian stem cell niche. J Cell Biol 209: 595-608. PubMed ID: 26008746

    Li, Z., Liu, S. and Cai, Y. (2015). EGFR/MAPK signaling regulates the proliferation of Drosophila renal and nephric stem cells. J Genet Genomics 42: 9-20. PubMed ID: 25619598

    Luo, L., Chai, P. C. and Cai, Y. (2013). Immunostaining of germline stem cells and the niche in Drosophila ovaries. Methods Mol Biol 1035: 1-7. PubMed ID: 23959977

    Chai, P. C., Liu, Z., Chia, W. and Cai, Y. (2013). Hedgehog signaling acts with the temporal cascade to promote neuroblast cell cycle exit. PLoS Biol 11: e1001494. PubMed ID: 23468593

    Liu, M., Lim, T. M. and Cai, Y. (2010). The Drosophila female germline stem cell lineage acts to spatially restrict DPP function within the niche. Sci Signal 3: ra57. PubMed ID: 20664066

    Cai, Y. and Laughon, A. (2009). The Drosophila Smad cofactor Schnurri engages in redundant and synergistic interactions with multiple corepressors. Biochim Biophys Acta 1789: 232-245. PubMed ID: 19437622

  • Patrick Callaerts VIB, Flanders, Belgium
    Winant, M., Buhler, K., Clements, J., De Groef, S., Hens, K., Vulsteke, V. and Callaerts, P. (2022). Genome-wide analysis identifies Homothorax and Extradenticle as regulators of insulin in Drosophila Insulin-Producing cells. PLoS Genet 18(9): e1010380. PubMed ID: 36095003

    De Groef, S., Wilms, T., Balmand, S., Calevro, F. and Callaerts, P. (2021). Sexual Dimorphism in Metabolic Responses to Western Diet in Drosophila melanogaster. Biomolecules 12(1). PubMed ID: 35053181

    Boonen, B., Startek, J. B., Milici, A., Lopez-Requena, A., Beelen, M., Callaerts, P. and Talavera, K. (2021). Activation of Drosophila melanogaster TRPA1 Isoforms by Citronellal and Menthol. Int J Mol Sci 22(20). PubMed ID: 34681657

    Clements, J., Buhler, K., Winant, M., Vulsteke, V. and Callaerts, P. (2021). Glial and Neuronal Neuroglian, Semaphorin-1a and Plexin A Regulate Morphological and Functional Differentiation of Drosophila Insulin-Producing Cells. Front Endocrinol (Lausanne) 12: 600251. PubMed ID: 34276554

    Steyaert, J., Scheveneels, W., Vanneste, J., Van Damme, P., Robberecht, W., Callaerts, P., Bogaert, E. and Van Den Bosch, L. (2018). FUS-induced neurotoxicity in Drosophila is prevented by downregulating nucleocytoplasmic transport proteins. Hum Mol Genet. PubMed ID: 30379317

    Buhler, K., Clements, J., Winant, M., Bolckmans, L., Vulsteke, V. and Callaerts, P. (2018). Growth control through regulation of insulin-signaling by nutrition-activated steroid hormone in Drosophila. Development. PubMed ID: 30266830

    Snellings, Y., Herrera, B., Wildemann, B., Beelen, M., Zwarts, L., Wenseleers, T. and Callaerts, P. (2018). The role of cuticular hydrocarbons in mate recognition in Drosophila suzukii. Sci Rep 8(1): 4996. PubMed ID: 29567945

    Zwarts, L., Vulsteke, V., Buhl, E., Hodge, J. J. and Callaerts, P. (2017). SlgA, the homologue of the human schizophrenia associated PRODH gene, acts in clock neurons to regulate Drosophila aggression. Dis Model Mech. PubMed ID: 28331058

    Versteven, M., Vanden Broeck, L., Geurten, B., Zwarts, L., Decraecker, L., Beelen, M., Göpfert, M.C., Heinrich, R. and Callaerts, P. (2017). Hearing regulates Drosophila aggression. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 28115690

    Zwarts, L., Goossens, T., Clements, J., Kang, Y. Y. and Callaerts, P. (2016). Axon branch-specific Semaphorin-1a signaling in Drosophila mushroom body development. Front Cell Neurosci 10: 210. PubMed ID: 27656129

    Herteleer, L., Zwarts, L., Hens, K., Forero, D., Del-Favero, J. and Callaerts, P. (2016). Mood stabilizing drugs regulate transcription of immune, neuronal and metabolic pathway genes in Drosophila. Psychopharmacology (Berl) [Epub ahead of print]. PubMed ID: 26852229

    Sitnik, J. L., Francis, C., Hens, K., Huybrechts, R., Wolfner, M. F. and Callaerts, P. (2014). Neprilysins: An Evolutionarily Conserved Family of Metalloproteases That Play Important Roles in Reproduction in Drosophila. Genetics. PubMed ID: 24395329

  • Brian Calvi Department of Biology, Indiana University
    Chakravarti, A., Thirimanne, H. N., Brown, S. and Calvi, B. R. (2022). Drosophila p53 isoforms have overlapping and distinct functions in germline genome integrity and oocyte quality control. Elife 11. PubMed ID: 35023826

    Rotelli, M. D., Bolling, A. M., Killion, A. W., Weinberg, A. J., Dixon, M. J. and Calvi, B. R. (2019). An RNAi Screen for Genes Required for Growth of Drosophila Wing Tissue. G3 (Bethesda). PubMed ID: 31387856

    Rotelli, M. D., Policastro, R. A., Bolling, A. M., Killion, A. W., Weinberg, A. J., Dixon, M. J., Zentner, G. E., Walczak, C. E., Lilly, M. A. and Calvi, B. R. (2019). A Cyclin A-Myb-MuvB-Aurora B network regulates the choice between mitotic cycles and polyploid endoreplication cycles. PLoS Genet 15(7): e1008253. PubMed ID: 31291240

    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

    Zhang, C., Montooth, K. L. and Calvi, B. R. (2017). Incompatibility between mitochondrial and nuclear genomes during oogenesis results in ovarian failure and embryonic lethality. Development 144(13): 2490-2503. PubMed ID: 28576772

    Paranjape, N. P. and Calvi, B. R. (2016). The histone variant H3.3 is enriched at Drosophila amplicon origins but does not mark them for activation. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27172191

    Qi, S. and Calvi, B.R. (2016). Different cell cycle modifications repress apoptosis at different steps independent of developmental signaling in Drosophila. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27075174

    Liu, J., Zimmer, K., Rusch, D. B., Paranjape, N., Podicheti, R., Tang, H. and Calvi, B. R. (2015). DNA sequence templates adjacent nucleosome and ORC sites at gene amplification origins in Drosophila. Nucleic Acids Res. PubMed ID: 26227968

    Zhang, B., Rotelli, M., Dixon, M. and Calvi, B. R. (2015). The function of Drosophila p53 isoforms in apoptosis. Cell Death Differ [Epub ahead of print]. PubMed ID: 25882045

    Zhang, B., Mehrotra, S., Ng, W. L., Calvi, B. R. (2014). Low Levels of p53 Protein and Chromatin Silencing of p53 Target Genes Repress Apoptosis in Drosophila Endocycling Cells. PLoS Genet 10: e1004581. PubMed ID: 25211335

    Hassel, C., Zhang, B., Dixon, M. and Calvi, B. R. (2013). Induction of endocycles represses apoptosis independently of differentiation and predisposes cells to genome instability. Development. PubMed ID: 24284207

    McConnell, K. H., Dixon, M. and Calvi, B. R. (2012). The histone acetyltransferases CBP and Chameau integrate developmental and DNA replication programs in Drosophila ovarian follicle cells. Development 139: 3880-3890. PubMed ID: 22951641

  • Jim Camakaris Department of Genetics, University of Melbourne
    Allan, K., Perez, K. A., Barnham, K. J., Camakaris, J. and Burke, R. (2014). A commonly used Drosophila model of Alzheimer's disease generates an aberrant species of amyloid-beta with an additional N-terminal glutamine residue. FEBS Lett. PubMed ID: 25171862

    Southon, A., Greenough, M. A., Ganio, G., Bush, A. I., Burke, R., Camakaris, J. (2013) Presenilin promotes dietary copper uptake. PLoS One 8: e62811. PubMed ID: 23667524

    Binks, T., Lye, J. C., Camakaris, J. and Burke, R. (2010). Tissue-specific interplay between copper uptake and efflux in Drosophila. J Biol Inorg Chem 15: 621-628. PubMed ID: 20151166

    Norgate, M., Southon, A., Greenough, M., Cater, M., Farlow, A., Batterham, P., Bush, A. I., Subramaniam, V. N., Burke, R. and Camakaris, J. (2010). Syntaxin 5 is required for copper homeostasis in Drosophila and mammals. PLoS One 5: e14303. PubMed ID: 21188142

  • Anthony Cammarato Heart & Vascular Institute, Johns Hopkins Medicine, Baltimore
    Viswanathan, M. C., Schmidt, W., Rynkiewicz, M. J., Agarwal, K., Gao, J., Katz, J., Lehman, W. and Cammarato, A. (2017). Distortion of the Actin A-triad results in contractile disinhibition and cardiomyopathy. Cell Rep 20(11): 2612-2625. PubMed ID: 28903042

    Viswanathan, M. C., Blice-Baum, A. C., Sang, T. K. and Cammarato, A. (2016). Cardiac-restricted expression of VCP/TER94 RNAi or disease alleles perturbs Drosophila heart structure and impairs function. J Cardiovasc Dev Dis 3. PubMed ID: 27500162

    Viswanathan, M. C., Blice-Baum, A. C., Schmidt, W., Foster, D. B. and Cammarato, A. (2015). Pseudo-acetylation of K326 and K328 of actin disrupts Drosophila melanogaster indirect flight muscle structure and performance. Front Physiol 6: 116. PubMed ID: 25972811

    Cozhimuttam Viswanathan, M., Kaushik, G., Engler, A. J., Lehman, W. J. and Cammarato, A. (2013). A Drosophila Melanogaster Model of Diastolic Dysfunction and Cardiomyopathy Based on Impaired Troponin-T Function. Circ Res. PubMed ID: 24221941

    Xie, H. B., Cammarato, A., Rajasekaran, N. S., Zhang, H., Suggs, J. A., Lin, H. C., Bernstein, S. I., Benjamin, I. J. and Golic, K. G. (2013). The NADPH metabolic network regulates human alphaB-crystallin cardiomyopathy and reductive stress in Drosophila melanogaster. PLoS Genet 9: e1003544. PubMed ID: 23818860

    Wang, Y., Melkani, G. C., Suggs, J. A., Melkani, A., Kronert, W. A., Cammarato, A. and Bernstein, S. I. (2012). Expression of the inclusion body myopathy 3 mutation in Drosophila depresses myosin function and stability and recapitulates muscle inclusions and weakness. Mol Biol Cell 23: 2057-2065. PubMed ID: 22496423

    Kaushik, G., Zambon, A. C., Fuhrmann, A., Bernstein, S. I., Bodmer, R., Engler, A. J. and Cammarato, A. (2012). Measuring passive myocardial stiffness in Drosophila melanogaster to investigate diastolic dysfunction. J Cell Mol Med 16: 1656-1662. PubMed ID: 22225769

  • Shelagh Campbell University of Alberta
    Willms, R. J., Zeng, J. and Campbell, S. D. (2020). Myt1 Kinase Couples Mitotic Cell Cycle Exit with Differentiation in Drosophila. Cell Rep 33(7): 108400. PubMed ID: 33207203

    Varadarajan, R., Ayeni, J., Jin, Z., Homola, E. and Campbell, S. D. (2016). Myt1 inhibition of Cyclin A/Cdk1 is essential for fusome integrity and pre-meiotic centriole engagement in Drosophila spermatocytes. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27170181

    Ayeni, J. O., Audibert, A., Fichelson, P., Srayko, M., Gho, M. and Campbell, S. D. (2016). G2-phase arrest prevents bristle progenitor self-renewal and synchronizes cell divisions with cell fate differentiation. Development [Epub ahead of print]. PubMed ID: 26893341

    Ayeni, J. O., Varadarajan, R., Mukherjee, O., Stuart, D. T., Sprenger, F., Srayko, M. and Campbell, S. D. (2013). Dual Phosphorylation of Cdk1 Coordinates Cell Proliferation with Key Developmental Processes in Drosophila. Genetics. PubMed ID: 24214341

    Li, X., Zhuo, R., Tiong, S., Di Cara, F., King-Jones, K., Hughes, S. C., Campbell, S. D., Wevrick, R. (2013) The Smc5/Smc6/MAGE Complex Confers Resistance to Caffeine and Genotoxic Stress in Drosophila melanogaster. PLoS One 8: e59866. PubMed ID: 23555814

    Fasulo, B., Koyama, C., Yu, K. R., Homola, E. M., Hsieh, T. S., Campbell, S. D. and Sullivan, W. (2012). Chk1 and Wee1 kinases coordinate DNA replication, chromosome condensation, and anaphase entry. Mol Biol Cell 23: 1047-1057. PubMed ID: 22262459

    Salle, J., Campbell, S. D., Gho, M. and Audibert, A. (2012). CycA is involved in the control of endoreplication dynamics in the Drosophila bristle lineage. Development 139: 547-557. PubMed ID: 22223681

    Joyce, E. F., Pedersen, M., Tiong, S., White-Brown, S. K., Paul, A., Campbell, S. D. and McKim, K. S. (2011). Drosophila ATM and ATR have distinct activities in the regulation of meiotic DNA damage and repair. J Cell Biol 195: 359-367. PubMed ID: 22024169

  • Jorge Campusano Departamento de Biología Celular y Molecular, Pontificia Universidad Catílica de Chile Santiago
    Zarate, R. V., Hidalgo, S., Navarro, N., Molina-Mateo, D., Arancibia, D., Rojo-Cortes, F., Oliva, C., Andres, M. E., Zamorano, P. and Campusano, J. M. (2022). An Early Disturbance in Serotonergic Neurotransmission Contributes to the Onset of Parkinsonian Phenotypes in Drosophila melanogaster. Cells 11(9). PubMed ID: 35563850

    Hidalgo, S., Fuenzalida-Uribe, N., Molina-Mateo, D., Escobar, A. P., Oliva, C., Espana, R. A., Andres, M. E. and Campusano, J. M. (2020). Study of the release of endogenous amines in Drosophila brain in vivo in response to stimuli linked to aversive olfactory conditioning. J Neurochem. PubMed ID: 32596813

    Hidalgo, S., Castro, C., Zarate, R. V., Valderrama, B. P., Hodge, J. J. L. and Campusano, J. M. (2020). The behavioral and neurochemical characterization of a Drosophila dysbindin mutant supports the contribution of serotonin to schizophrenia negative symptoms. Neurochem Int 138: 104753. PubMed ID: 32416114

    Silva, B., Hidalgo, S. and Campusano, J. M. (2020). Dop1R1, a type 1 dopaminergic receptor expressed in Mushroom Bodies, modulates Drosophila larval locomotion. PLoS One 15(2): e0229671. PubMed ID: 32101569

    Fuenzalida-Uribe, N. and Campusano, J. M. (2017). Unveiling the dual role of the dopaminergic system on locomotion and the innate value for an aversive olfactory stimulus in Drosophila. Neuroscience 371: 433-444. PubMed ID: 29292079

    Molina-Mateo, D., Fuenzalida-Uribe, N., Hidalgo, S., Molina-Fernandez, C., Abarca, J., Zarate, R. V., Escandon, M., Figueroa, R., Tevy, M. F. and Campusano, J. M. (2017). Characterization of a presymptomatic stage in a Drosophila Parkinson's disease model: Unveiling dopaminergic compensatory mechanisms. Biochim Biophys Acta. PubMed ID: 28716706

    Hidalgo, S., Molina-Mateo, D., Escobedo, P., Zarate, R. V., Fritz, E., Fierro, A., Perez, E. G., Iturriaga-Vasquez, P., Reyes-Parada, M., Varas, R., Fuenzalida-Uribe, N. and Campusano, J. M. (2017). Characterization of a novel Drosophila SERT mutant: insights on the contribution of the serotonin neural system to behaviors. ACS Chem Neurosci. PubMed ID: 28665105

    Silva, B., Molina-Fernandez, C., Ugalde, M. B., Tognarelli, E. I., Angel, C. and Campusano, J. M. (2015). Muscarinic ACh Receptors Contribute to Aversive Olfactory Learning in Drosophila. Neural Plast 2015: 658918. PubMed ID: 26380118

    Silva, B., Goles, N. I., Varas, R. and Campusano, J. M. (2014). Serotonin receptors expressed in Drosophila mushroom bodies differentially modulate larval locomotion. PLoS One 9: e89641. PubMed ID: 24586928

    Leyton, V., Goles, N. I., Fuenzalida-Uribe, N. and Campusano, J. M. (2013). Octopamine and Dopamine differentially modulate the nicotine-induced calcium response in Drosophila Mushroom Body Kenyon Cells. Neurosci Lett. PubMed ID: 24334164

    Fuenzalida-Uribe, N., Meza, R. C., Hoffmann, H. A., Varas, R., Campusano, J. M. (2013) nAChR-induced octopamine release mediates the effect of nicotine onstartle response in/ofDrosophila melanogaster. J Neurochem. PubMed ID: 23331098

  • Sonsoles Campuzano Centro de Biología Molecular Severo Ochoa, Madrid
    Barrios, N. and Campuzano, S. (2016). Expanding the Iroquois genes repertoire: A non-transcriptional function in cell cycle progression. Fly (Austin) [Epub ahead of print]. PubMed ID: 26760760

    Barrios, N., Gonzalez-Perez, E., Hernandez, R. and Campuzano, S. (2015). The Homeodomain Iroquois Proteins Control Cell Cycle Progression and Regulate the Size of Developmental Fields. PLoS Genet 11: e1005463. PubMed ID: 26305360

    Andreu, M. J., Ajuria, L., Samper, N., Gonzalez-Perez, E., Campuzano, S., Gonzalez-Crespo, S. and Jimenez, G. (2012). EGFR-dependent downregulation of Capicua and the establishment of Drosophila dorsoventral polarity. Fly (Austin) 6: 234-239. PubMed ID: 22878648

    Andreu, M. J., Gonzalez-Perez, E., Ajuria, L., Samper, N., Gonzalez-Crespo, S., Campuzano, S. and Jimenez, G. (2012). Mirror represses pipe expression in follicle cells to initiate dorsoventral axis formation in Drosophila. Development 139: 1110-1114. PubMed ID: 22318229

  • Cao Gang Huazhong Agricultural University. Wuhan, China
    Su, J., Zhang, Y., Su, H., Wang, C., Wang, D., Yang, Y., Li, X., Qi, W., Li, H., Li, X., Song, Y. and Cao, G. (2022). Dosage Compensation of the X Chromosome during Sheep Testis Development Revealed by Single-Cell RNA Sequencing. Animals (Basel) 12(17). PubMed ID: 36077890

    Yu, J., Xiao, K., Chen, X., Deng, L., Zhang, L., Li, Y., Gao, A., Gao, J., Wu, C., Yang, X., Zhou, Q., Yang, J., Bao, C., Jiao, J., Cheng, S., Guo, Z., Xu, W., Cao, X., Guo, Z., Dai, J., Hu, J., Fu, Z. and Cao, G. (2022). Neuron-derived neuropeptide Y fine-tunes the splenic immune responses. Neuron 110(8): 1327-1339 e1326. PubMed ID: 35139365

    Zhou, F., Cao, G., Dai, S., Li, G., Li, H., Ding, Z., Hou, S., Xu, B., You, W., Wiseglass, G., Shi, F., Yang, X., Rubinstein, R. and Jin, Y. (2020). Chelicerata sDscam isoforms combine homophilic specificities to define unique cell recognition. Proc Natl Acad Sci U S A 117(40): 24813-24824. PubMed ID: 32963097

  • Maya Capelson Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
    Pascual-Garcia, P., Little, S. C. and Capelson, M. (2022). Nup98-dependent transcriptional memory is established independently of transcription. Elife 11. PubMed ID: 35289742

    Aleman, J. R., Kuhn, T. M., Pascual-Garcia, P., Gospocic, J., Lan, Y., Bonasio, R., Little, S. C. and Capelson, M. (2021). Correct dosage of X chromosome transcription is controlled by a nuclear pore component. Cell Rep 35(11): 109236. PubMed ID: 34133927

    Gozalo, A., Duke, A., Lan, Y., Pascual-Garcia, P., Talamas, J. A., Nguyen, S. C., Shah, P. P., Jain, R., Joyce, E. F. and Capelson, M. (2019). Core Components of the Nuclear Pore Bind Distinct States of Chromatin and Contribute to Polycomb Repression. Mol Cell. PubMed ID: 31784359

    Kuhn, T. M., Pascual-Garcia, P., Gozalo, A., Little, S. C. and Capelson, M. (2019). Chromatin targeting of nuclear pore proteins induces chromatin decondensation. J Cell Biol. PubMed ID: 31366666

    Pascual-Garcia, P., Debo, B., Aleman, J. R., Talamas, J. A., Lan, Y., Nguyen, N. H., Won, K. J. and Capelson, M. (2017). Metazoan Nuclear Pores Provide a Scaffold for Poised Genes and Mediate Induced Enhancer-Promoter Contacts. Mol Cell 66(1): 63-76 e66. PubMed ID: 28366641

    Pascual-Garcia, P., Jeong, J. and Capelson, M. (2014). Nucleoporin Nup98 associates with Trx/MLL and NSL histone-modifying complexes and regulates Hox gene expression. Cell Rep 9(2): 433-442. PubMed ID: 25310983

  • Maria Capovilla Institut Sophia Agrobiotech, Université, Nice
    Raad, H., Ferveur, J.F., Ledger, N., Capovilla, M. and Robichon, A. (2016). Functional gustatory role of chemoreceptors in Drosophila wings. Cell Rep [Epub ahead of print]. PubMed ID: 27160896

    Valmalette, J. C., Raad, H., Qiu, N., Ohara, S., Capovilla, M. and Robichon, A. (2015). Nano-architecture of gustatory chemosensory bristles and trachea in Drosophila wings. Sci Rep 5: 14198. PubMed ID: 26381332

    Tevy, M. F., Seyres, D., Traina, C., Perrin, L. and Capovilla, M. (2014). Ndae1 expression and regulation in Drosophila embryos. PLoS One 9: e92956. PubMed ID: 24676142

    Amodio, V., Tevy, M. F., Traina, C., Ghosh, T. K. and Capovilla, M. (2012). Transactivation in Drosophila of human enhancers by human transcription factors involved in congenital heart diseases. Dev Dyn 241: 190-199. PubMed ID: 21990232

  • Pau Carazo Institut Cavanilles de Biodiversitat i Biologia Evolutiva, Universitat de Valencia, Spain.
    >Corbel, Q., Londono-Nieto, C. and Carazo, P. (2022). Does perception of female cues modulate male short-term fitness components in Drosophila melanogaster? Ecol Evol 12(9): e9287. PubMed ID: 36177144

    Corbel, Q., Serra, M., Garcia-Roa, R. and Carazo, P. (2022). Male Adaptive Plasticity Can Explain the Evolution of Sexual Perception Costs. Am Nat 200(3): E110-E123. PubMed ID: 35977789

    Marquez-Rosado, A., Garcia-Co, C., Londono-Nieto, C. and Carazo, P. (2022). No evidence that relatedness or familiarity modulates male harm in Drosophila melanogaster flies from a wild population. Ecol Evol 12(4): e8803. PubMed ID: 35432938

  • Gwyneth Card Janelia Research Campus, Ashburn, Va.
    Dombrovski, M., Peek, M. Y., Park, J. Y., Vaccari, A., Sumathipala, M., Morrow, C., Breads, P., Zhao, A., Kurmangaliyev, Y. Z., Sanfilippo, P., Rehan, A., Polsky, J., Alghailani, S., Tenshaw, E., Namiki, S., Zipursky, S. L. and Card, G. M. (2023). Synaptic gradients transform object location to action. Nature 613(7944): 534-542. PubMed ID: 36599984

    Klapoetke, N. C., Nern, A., Rogers, E. M., Rubin, G. M., Reiser, M. B. and Card, G. M. (2022). A functionally ordered visual feature map in the Drosophila brain. Neuron. PubMed ID: 35290791

    Ache, J. M., Polsky, J., Alghailani, S., Parekh, R., Breads, P., Peek, M. Y., Bock, D. D., von Reyn, C. R. and Card, G. M. (2019). Neural basis for looming size and velocity encoding in the Drosophila giant fiber escape pathway. Curr Biol 29(6): 1073-1081.e1074. PubMed ID: 30827912

    Namiki, S., Dickinson, M. H., Wong, A. M., Korff, W. and Card, G. M. (2018). The functional organization of descending sensory-motor pathways in Drosophila. Elife 7. PubMed ID: 29943730

    Cande, J., Namiki, S., Qiu, J., Korff, W., Card, G. M., Shaevitz, J. W., Stern, D. L. and Berman, G. J. (2018). Optogenetic dissection of descending behavioral control in Drosophila. Elife 7. PubMed ID: 29943729

    von Reyn, C. R., Nern, A., Williamson, W. R., Breads, P., Wu, M., Namiki, S. and Card, G. M. (2017). Feature Integration Drives Probabilistic Behavior in the Drosophila Escape Response. Neuron 94(6): 1190-1204 e1196. PubMed ID: 28641115

    Wu, M., Nern, A., Williamson, W. R., Morimoto, M. M., Reiser, M. B., Card, G. M. and Rubin, G. M. (2016). Visual projection neurons in the Drosophila lobula link feature detection to distinct behavioral programs. Elife 5. PubMed ID: 28029094

    Mellert, D. J., Williamson, W. R., Shirangi, T. R., Card, G. M. and Truman, J. W. (2016). Genetic and Environmental Control of Neurodevelopmental Robustness in Drosophila. PLoS One 11(5): e0155957. PubMed ID: 27223118

  • Albert Cardona MRC Laboratory of Molecular Biology, Cambridge, U. K.
    Patel, A. A., Cardona, A. and Cox, D. N. (2023). Neural substrates of cold nociception in Drosophila larva. bioRxiv. PubMed ID: 37577520

    Winding, M., Pedigo, B. D., Barnes, C. L., Patsolic, H. G., Park, Y., Kazimiers, T., Fushiki, A., Andrade, I. V., Khandelwal, A., Valdes-Aleman, J., Li, F., Randel, N., Barsotti, E., Correia, A., Fetter, R. D., Hartenstein, V., Priebe, C. E., Vogelstein, J. T., Cardona, A. and Zlatic, M. (2023). The connectome of an insect brain. Science 379(6636): eadd9330. PubMed ID: 36893230

    Barnes, C. L., Bonnery, D. and Cardona, A. (2022). Synaptic counts approximate synaptic contact area in Drosophila. PLoS One 17(4): e0266064. PubMed ID: 35377898

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

    Eschbach, C., Fushiki, A., Winding, M., Afonso, B., Andrade, I. V., Cocanougher, B. T., Eichler, K., Gepner, R., Si, G., Valdes-Aleman, J., Fetter, R. D., Gershow, M., Jefferis, G. S., Samuel, A. D., Truman, J. W., Cardona, A. and Zlatic, M. (2021). Circuits for integrating learned and innate valences in the insect brain. Elife 10. PubMed ID: 34755599

    Imambocus, B. N., Zhou, F., Formozov, A., Wittich, A., Tenedini, F. M., Hu, C., Sauter, K., Macarenhas Varela, E., Heredia, F., Casimiro, A. P., Macedo, A., Schlegel, P., Yang, C. H., Miguel-Aliaga, I., Wiegert, J. S., Pankratz, M. J., Gontijo, A. M., Cardona, A. and Soba, P. (2021). A neuropeptidergic circuit gates selective escape behavior of Drosophila larvae. Curr Biol. PubMed ID: 34798050

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

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

    Andrade, I. V., Riebli, N., Nguyen, B. M., Omoto, J. J., Cardona, A. and Hartenstein, V. (2019). Developmentally arrested precursors of pontine neurons establish an embryonic blueprint of the Drosophila central complex. Curr Biol 29(3): 412-425.e413. PubMed ID: 30661802

    Tastekin, I., Khandelwal, A., Tadres, D., Fessner, N. D., Truman, J. W., Zlatic, M., Cardona, A. and Louis, M. (2018). Sensorimotor pathway controlling stopping behavior during chemotaxis in the Drosophila melanogaster larva. Elife 7. PubMed ID: 30465650

    Miroschnikow, A., Schlegel, P., Schoofs, A., Hueckesfeld, S., Li, F., Schneider-Mizell, C. M., Fetter, R. D., Truman, J. W., Cardona, A. and Pankratz, M. J. (2018). Convergence of monosynaptic and polysynaptic sensory paths onto common motor outputs in a Drosophila feeding connectome. Elife 7. PubMed ID: 30526854

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

  • John Carlson Dept. of Molecular, Cellular, and Developmental Biology. Yale University
    Wang, W., Dweck, H. K. M., Talross, G. J. S., Zaidi, A., Gendron, J. M. and Carlson, J. R. (2022). Sugar sensation and mechanosensation in the egg-laying preference shift of Drosophila suzukii. Elife 11. PubMed ID: 36398882

    Xiao, S., Baik, L. S., Shang, X. and Carlson, J. R. (2022). Meeting a threat of the Anthropocene: Taste avoidance of metal ions by Drosophila. Proc Natl Acad Sci U S A 119(25): e2204238119. PubMed ID: 35700364

    Dweck, H. K. M., Talross, G. J. S., Luo, Y., Ebrahim, S. A. M. and Carlson, J. R. (2022). Ir56b is an atypical ionotropic receptor that underlies appetitive salt response in Drosophila. Curr Biol 32(8): 1776-1787.e1774. PubMed ID: 35294865

    Dweck, H. K. M., Talross, G. J. S., Luo, Y., Ebrahim, S. A. M. and Carlson, J. R. (2022). Ir56b is an atypical ionotropic receptor that underlies appetitive salt response in Drosophila. Curr Biol. PubMed ID: 35294865

    Ebrahim, S. A. M., Talross, G. J. S. and Carlson, J. R. (2021). Sight of parasitoid wasps accelerates sexual behavior and upregulates a micropeptide gene in Drosophila. Nat Commun 12(1): 2453. PubMed ID: 33907186

    Dweck, H. K. M. and Carlson, J. R. (2019). Molecular Logic and Evolution of Bitter Taste in Drosophila. Curr Biol. PubMed ID: 31839451

    Xiao, S., Sun, J. S. and Carlson, J. R. (2019). Robust olfactory responses in the absence of odorant binding proteins. Elife 8. PubMed ID: 31651397

    Soni, N., Chahda, J. S. and Carlson, J. R. (2019). Odor coding in the antenna of the tsetse fly Glossina morsitans. Proc Natl Acad Sci U S A. PubMed ID: 31221757

    He, Z., Luo, Y., Shang, X., Sun, J. S. and Carlson, J. R. (2019). Chemosensory sensilla of the Drosophila wing express a candidate ionotropic pheromone receptor. PLoS Biol 17(5): e2006619. PubMed ID: 31112532

    Chahda, J. S., Soni, N., Sun, J. S., Ebrahim, S. A. M., Weiss, B. L. and Carlson, J. R. (2019). The molecular and cellular basis of olfactory response to tsetse fly attractants. PLoS Genet 15(3): e1008005. PubMed ID: 30875383

    Sun, J. S., Larter, N. K., Chahda, J. S., Rioux, D., Gumaste, A. and Carlson, J. R. (2018). Humidity response depends on the small soluble protein Obp59a in Drosophila. Elife 7. PubMed ID: 30230472

    Park, J. and Carlson, J. R. (2017). Physiological responses of the Drosophila labellum to amino acids. J Neurogenet: 1-10. PubMed ID: 29191065

    Joseph, R. M., Sun, J. S., Tam, E. and Carlson, J. R. (2017). A receptor and neuron that activate a circuit limiting sucrose consumption. Elife 6. PubMed ID: 28332980

    Delventhal, R., Menuz, K., Joseph, R., Park, J., Sun, J. S. and Carlson, J. R. (2017). The taste response to ammonia in Drosophila. Sci Rep 7: 43754. PubMed ID: 28262698

    Larter, N. K., Sun, J. S. and Carlson, J. R. (2016). Organization and function of Drosophila odorant binding proteins. Elife 5. PubMed ID: 27845621

  • Ana Carmena de la Cruz Instituto de Neurociencia, CSIC-Universidad Miguel Hernández, Alicante, Spain
    de Torres-Jurado, A., Manzanero-Ortiz, S. and Carmena, A. (2022). Glial-secreted Netrins regulate Robo1/Rac1-Cdc42 signaling threshold levels during Drosophila asymmetric neural stem/progenitor cell division. Curr Biol 32(10): 2174-2188.e2173. PubMed ID: 35472309

    Manzanero-Ortiz, S., de Torres-Jurado, A., Hernández-Rojas, R. and Carmena, A. (2021). Pilot RNAi Screen in Drosophila Neural Stem Cell Lineages to Identify Novel Tumor Suppressor Genes Involved in Asymmetric Cell Division. Int J Mol Sci 22(21). PubMed ID: 34768763

    Manzanero-Ortiz, S., de Torres-Jurado, A., Hernandez-Rojas, R. and Carmena, A. (2021). Pilot RNAi Screen in Drosophila Neural Stem Cell Lineages to Identify Novel Tumor Suppressor Genes Involved in Asymmetric Cell Division. Int J Mol Sci 22(21). PubMed ID: 34768763

    Carmena, A. (2018). Compromising asymmetric stem cell division in Drosophila central brain: Revisiting the connections with tumorigenesis. Fly (Austin) 12(1): 71-80. PubMed ID: 29239688

    Perez-Gomez, R., Slovakova, J., Rives-Quinto, N., Krejci, A. and Carmena, A. (2013). A Serrate-Notch-Canoe complex mediates essential interactions between glia and neuroepithelial cells during Drosophila optic lobe development. J Cell Sci 126(Pt 21): 4873-4884. PubMed ID: 23970418

    Carmena, A. (2009). Approaching Drosophila development through proteomic tools and databases: At the hub of the post-genomic era. Mech Dev 126(10): 761-770. PubMed ID: 19679183

  • Ginger Carney Biology, Texas A&M, College Station
    Iftikhar, H., Johnson, N. L., Marlatt, M. L. and Carney, G. E. (2019). The role of miRNAs in Drosophila melanogaster male courtship behavior. Genetics. PubMed ID: 30683757

    Schultzhaus, J. N., Bennett, C. J., Iftikhar, H., Yew, J. Y., Mallett, J. and Carney, G. E. (2018). High fat diet alters Drosophila melanogaster sexual behavior and traits: decreased attractiveness and changes in pheromone profiles. Sci Rep 8(1): 5387. PubMed ID: 29599496

    Schultzhaus, J. N. and Carney, G. E. (2017). Dietary protein content alters both male and female contributions to Drosophila melanogaster female post-mating response traits. J Insect Physiol [Epub ahead of print]. PubMed ID: 28414060

    Schultzhaus, J. N., Saleem, S., Iftikhar, H. and Carney, G. E. (2016). The role of the Drosophila lateral horn in olfactory information processing and behavioral response. J Insect Physiol 98: 29-37. PubMed ID: 27871975

    Saleem, S., Ruggles, P. H., Abbott, W. K. and Carney, G. E. (2014). Sexual Experience Enhances Drosophila melanogaster Male Mating Behavior and Success. PLoS One 9: e96639. PubMed ID: 24805129

    Ratterman, N. L., Rosenthal, G. G., Carney, G. E. and Jones, A. G. (2014). Genetic variation and covariation in male attractiveness and female mating preferences in Drosophila melanogaster. G3 (Bethesda) 4: 79-88. PubMed ID: 24212081

    Saleem, S., Schwedes, C. C., Ellis, L. L., Grady, S. T., Adams, R. L., Johnson, N., Whittington, J. R. and Carney, G. E. (2012). Drosophila melanogaster p24 trafficking proteins have vital roles in development and reproduction. Mech Dev 129: 177-191. PubMed ID: 22554671

  • Robert Carrillo Department of Molecular Genetics and Cell Biology, University of Chicago
    Lobb-Rabe, M., DeLong, K., Salazar, R. J., Zhang, R., Wang, Y. and Carrillo, R. A. (2022). Dpr10 and Nocte are required for Drosophila motor axon pathfinding. Neural Dev 17(1): 10. PubMed ID: 36271407

    Wang, Y., Lobb-Rabe, M., Ashley, J., Chatterjee, P., Anand, V., Bellen, H. J., Kanca, O. and Carrillo, R. A. (2022). Systematic expression profiling of Dpr and DIP genes reveals cell surface codes in Drosophila larval motor and sensory neurons. Development 149(10). PubMed ID: 35502740

    Wang, Y., Lobb-Rabe, M., Ashley, J., Anand, V. and Carrillo, R. A. (2021). Structural and functional synaptic plasticity induced by convergent synapse loss in the Drosophila neuromuscular circuit. J Neurosci. PubMed ID: 33402422

    Ashley, J., Sorrentino, V., Lobb-Rabe, M., Nagarkar-Jaiswal, S., Tan, L., Xu, S., Xiao, Q., Zinn, K. and Carrillo, R. A. (2019). Transsynaptic interactions between IgSF proteins DIP-alpha and Dpr10 are required for motor neuron targeting specificity. Elife 8. PubMed ID: 30714906

    Carrillo, R.A., Özkan, E., Menon, K.P., Nagarkar-Jaiswal, S., Lee, P.T., Jeon, M., Birnbaum, M.E., Bellen, H.J., Garcia, K.C. and Zinn, K. (2015). Control of synaptic connectivity by a network of Drosophila IgSF cell surface proteins. Cell 163: 1770-1782. PubMed ID: 26687361

    Menon, K.P., Carrillo, R.A. and Zinn, K. (2015). The translational regulator Cup controls NMJ presynaptic terminal morphology. Mol Cell Neurosci [Epub ahead of print]. PubMed ID: 26102195

  • Dana Carroll Biological Chemistry, University of Utah
    Beumer, K. J. and Carroll, D. (2014). Targeted genome engineering techniques in Drosophila. Methods. PubMed ID: 24412316

    Beumer, K. J., Trautman, J. K., Christian, M., Dahlem, T. J., Lake, C. M., Hawley, R. S., Grunwald, D. J., Voytas, D. F. and Carroll, D. (2013). Comparing ZFNs and TALENs for Gene Targeting in Drosophila. G3 (Bethesda). PubMed ID: 23979928

    Beumer, K. J., Trautman, J. K., Mukherjee, K., Carroll, D. (2013) Donor DNA Utilization during Gene Targeting with Zinc-finger Nucleases. G3 (Bethesda). PubMed ID: 23550125

    Takasu, Y., Kobayashi, I., Beumer, K., Uchino, K., Sezutsu, H., Sajwan, S., Carroll, D., Tamura, T. and Zurovec, M. (2010). Targeted mutagenesis in the silkworm Bombyx mori using zinc finger nuclease mRNA injection. Insect Biochem Mol Biol 40: 759-765. PubMed ID: 20692340

    Carroll, D., Beumer, K. J. and Trautman, J. K. (2010). High-efficiency gene targeting in Drosophila with zinc finger nucleases. Methods Mol Biol 649: 271-280. PubMed ID: 20680841

  • Sean Carroll Laboratory of Cell and Molecular Biology, University of Wisconsin
    Loehlin, D. W., Ames, J. R., Vaccaro, K. and Carroll, S. B. (2019). A major role for noncoding regulatory mutations in the evolution of enzyme activity. Proc Natl Acad Sci U S A 116(25): 12383-12389. PubMed ID: 31152141

    Loehlin, D. W. and Carroll, S. B. (2016). Expression of tandem gene duplicates is often greater than twofold. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27162370

    Koshikawa, S., Giorgianni, M. W., Vaccaro, K., Kassner, V. A., Yoder, J. H., Werner, T. and Carroll, S. B. (2015). Gain of cis-regulatory activities underlies novel domains of wingless gene expression in Drosophila. Proc Natl Acad Sci U S A 112: 7524-7529. PubMed ID: 26034272

    Wang, W., Kidd, B. J., Carroll, S. B. and Yoder, J. H. (2011). Sexually dimorphic regulation of the Wingless morphogen controls sex-specific segment number in Drosophila. Proc Natl Acad Sci U S A 108: 11139-11144. PubMed ID: 21690416

    Rebeiz, M., Pool, J. E., Kassner, V. A., Aquadro, C. F. and Carroll, S. B. (2009). Stepwise modification of a modular enhancer underlies adaptation in a Drosophila population. Science 326(5960): 1663-7. PubMed ID: 20019281

    Werner, T., Koshikawa, S., Williams, T. M. and Carroll, S. B. (2010). Generation of a novel wing colour pattern by the Wingless morphogen. Nature 464: 1143-1148. PubMed ID: 20376004

  • Richard Carthew Northwestern University
    Bernasek, S. M., Hur, S. S. J., Peláez-Restrepo, N., Lachance, J. B., Bakker, R., Navarro, H. T., Sanchez-Luege, N., Amaral, L. A. N., Bagheri, N., Rebay, I. and Carthew, R. W. (2023). Ratiometric sensing of Pnt and Yan transcription factor levels confers ultrasensitivity to photoreceptor fate transitions in Drosophila. Development. PubMed ID: 36942737

    Giri, R., Brady, S., Papadopoulos, D. K. and Carthew, R. W. (2022). Single-cell Senseless protein analysis reveals metastable states during the transition to a sensory organ fate. iScience 25(10): 105097. PubMed ID: 36157584

    Gallagher, K. D., Mani, M. and Carthew, R. W. (2022). Emergence of a geometric pattern of cell fates from tissue-scale mechanics in the Drosophila eye. Elife 11. PubMed ID: 35037852

    Alba, V., Carthew, J. E., Carthew, R. W. and Mani, M. (2021). Global constraints within the developmental program of the Drosophila wing. Elife 10. PubMed ID: 34180394

    Bakker, R., Mani, M. and Carthew, R. W. (2020). The Wg and Dpp morphogens regulate gene expression by modulating the frequency of transcriptional bursts. Elife 9. PubMed ID: 32568073

    Giri, R., Papadopoulos, D. K., Posadas, D. M., Potluri, H. K., Tomancak, P., Mani, M. and Carthew, R. W. (2020). Ordered patterning of the sensory system is susceptible to stochastic features of gene expression. Elife 9. PubMed ID: 32101167

    Agbu, P., Cassidy, J. J., Braverman, J., Jacobson, A. and Carthew, R. W. (2019). MicroRNA miR-7 regulates secretion of insulin-like peptides. Endocrinology. PubMed ID: 31875904

    Cassidy, J. J., Bernasek, S. M., Bakker, R., Giri, R., Pelaez, N., Eder, B., Bobrowska, A., Bagheri, N., Nunes Amaral, L. A. and Carthew, R. W. (2019). Repressive Gene Regulation Synchronizes Development with Cellular Metabolism. Cell. PubMed ID: 31353220

    Church, V. A., Pressman, S., Isaji, M., Truscott, M., Cizmecioglu, N. T., Buratowski, S., Frolov, M. V. and Carthew, R. W. (2017). Microprocessor recruitment to elongating RNA polymerase II is required for differential expression of microRNAs. Cell Rep 20(13): 3123-3134. PubMed ID: 28954229

    Pelaez, N., Gavalda-Miralles, A., Wang, B., Navarro, H. T., Gudjonson, H., Rebay, I., Dinner, A. R., Katsaggelos, A. K., Amaral, L. A. and Carthew, R. W. (2015). Dynamics and heterogeneity of a fate determinant during transition towards cell differentiation. Elife 4 [Epub ahead of print]. PubMed ID: 26583752

    Qi, J., Wang, B., Pelaez, N., Rebay, I., Carthew, R. W., Katsaggelos, A. K. and Nunes Amaral, L. A. (2013). Drosophila Eye Nuclei Segmentation Based on Graph Cut and Convex Shape Prior. Int Conf Signal Process Proc: 670-674. PubMed ID: 25089515

  • Jose Maria Carvajal-Gonzalez
    Garrido-Jimenez, S., Roman, A. C. and Carvajal-Gonzalez, J. M. (2019). Diminished Expression of Fat and Dachsous PCP Proteins Impaired Centriole Planar Polarization in Drosophila. Front Genet 10: 328. PubMed ID: 31031805

    Garrido-Jimenez, S., Roman, A. C., Alvarez-Barrientos, A. and Carvajal-Gonzalez, J. M. (2018). Centriole planar polarity assessment in Drosophila wings. Development 145(23). PubMed ID: 30389850

    Carvajal-Gonzalez, J. M., Mulero-Navarro, S., Smith, M. and Mlodzik, M. (2016). A Novel Frizzled-Based Screening Tool Identifies Genetic Modifiers of Planar Cell Polarity in Drosophila Wings. G3 (Bethesda) 6(12): 3963-3973. PubMed ID: 27729438

  • Antonio Bernardo de Carvalho Department of Genetics, Federal University of Rio de Janeiro
    Ricchio, J., Uno, F. and Carvalho, A. B. (2021). New Genes in the Drosophila Y Chromosome: Lessons from D. willistoni. Genes (Basel) 12(11). PubMed ID: 34828421

    Vanderlinde, T., Dupim, E. G., Nazario-Yepiz, N. O. and Carvalho, A. B. (2018). An improved genome assembly for Drosophila navojoa, the basal species in the mojavensis cluster. J Hered. PubMed ID: 30423125

    Dupim, E. G., Goldstein, G., Vanderlinde, T., Vaz, S. C., Krsticevic, F., Bastos, A., Pinhao, T., Torres, M., David, J. R., Vilela, C. R. and Carvalho, A. B. (2018). An investigation of Y chromosome incorporations in 400 species of Drosophila and related genera. PLoS Genet 14(11): e1007770. PubMed ID: 30388103

    Carvalho, A. B., Vicoso, B., Russo, C. A., Swenor, B. and Clark, A. G. (2015). Birth of a new gene on the Y chromosome of Drosophila melanogaster. Proc Natl Acad Sci U S A 112(40): 12450-12455. PubMed ID: 26385968

  • Ruben J. Cauchi Faculty of Medicine & Surgery, University of Malta
    Herrera, P. and Cauchi, R. J. (2023). Functional characterisation of the ACE2 orthologues in Drosophila provides insights into the neuromuscular complications of COVID-19. Biochim Biophys Acta Mol Basis Dis 1869(8): 166818. PubMed ID: 37495086

    Servettini, I., Talani, G., Megaro, A., Setzu, M. D., Biggio, F., Briffa, M., Guglielmi, L., Savalli, N., Binda, F., Delicata, F., Bru-Mercier, G., Vassallo, N., Maglione, V., Cauchi, R. J., Di Pardo, A., Collu, M., Imbrici, P., Catacuzzeno, L., D'Adamo, M. C., Olcese, R. and Pessia, M. (2023). An activator of voltage-gated K(+) channels Kv1.1 as a therapeutic candidate for episodic ataxia type 1. Proc Natl Acad Sci U S A 120(31): e2207978120. PubMed ID: 37487086

    >Borg, R., Herrera, P., Purkiss, A., Cacciottolo, R. and Cauchi, R. J. (2023). Reduced levels of ALS gene DCTN1 induce motor defects in Drosophila. Front Neurosci 17: 1164251. PubMed ID: 37360176

    Borg, R., Purkiss, A., Cacciottolo, R., Herrera, P. and Cauchi, R. J. (2023). Loss of amyotrophic lateral sclerosis risk factor SCFD1 causes motor dysfunction in Drosophila. Neurobiol Aging 126: 67-76. PubMed ID: 36944290

  • Elena Casacuberta Consejo Superior de Investigaciones Científicas, Madrid
    Lopez-Panades, E. and Casacuberta, E. (2015). NAP-1, Nucleosome assembly protein 1, a histone chaperone involved in Drosophila telomeres. Insect Biochem Mol Biol [Epub ahead of print]. PubMed ID: 26742602

    Lopez-Panades, E., Gavis, E. R. and Casacuberta, E. (2015). Specific localization of the Drosophila telomere transposon proteins and RNAs, give insight in their behavior, control and telomere biology in this organism. PLoS One 10: e0128573. PubMed ID: 26068215

    Silva-Sousa, R. and Casacuberta, E. (2013). The JIL-1 Kinase Affects Telomere Expression in the Different Telomere Domains of Drosophila. PLoS One 8: e81543. PubMed ID: 24244743

    Silva-Sousa, R., Diaz Varela, M. and Casacuberta, E. (2013). The Putzig partners DREF, TRF2 and KEN are involved in the regulation of the Drosophila telomere retrotransposons, HeT-A and TART. Mob DNA 4: 18. PubMed ID: 23822164

    Silva-Sousa, R., Lopez-Panades, E., Pineyro, D. and Casacuberta, E. (2012). The Chromosomal Proteins JIL-1 and Z4/Putzig Regulate the Telomeric Chromatin in Drosophila melanogaster. PLoS Genet 8: e1003153. PubMed ID: 23271984

    Petit, N., Pineyro, D., Lopez-Panades, E., Casacuberta, E. and Navarro, A. (2012). HeT-A_pi1, a piRNA target sequence in the Drosophila telomeric retrotransposon HeT-A, is extremely conserved across copies and species. PLoS One 7: e37405. PubMed ID: 22629389

    Pineyro, D., Lopez-Panades, E., Lucena-Perez, M. and Casacuberta, E. (2011). Transcriptional analysis of the HeT-A retrotransposon in mutant and wild type stocks reveals high sequence variability at Drosophila telomeres and other unusual features. BMC Genomics 12: 573. PubMed ID: 22111838

  • José Casal Department of Zoology, University of Cambridge
    Pietra, S., Ng, K., Lawrence, P. A. and Casal, J. (2020). Planar cell polarity in the larval epidermis of Drosophila and the role of microtubules. Open Biol 10(12): 200290. PubMed ID: 33295841

    Saavedra, P., Brittle, A., Palacios, I.M., Strutt, D., Casal, J. and Lawrence, P.A. (2016). Planar cell polarity: the Dachsous/Fat system contributes differently to the embryonic and larval stages of Drosophila. Biol Open [Epub ahead of print]. PubMed ID: 26935392

    Medina, I., Casal, J. and Fabre, C.C. (2015). Do circadian genes and ambient temperature affect substrate-borne signalling during Drosophila courtship? Biol Open [Epub ahead of print]. PubMed ID: 26519517

    Saavedra, P., Vincent, J. P., Palacios, I. M., Lawrence, P. A. and Casal, J. (2014). Plasticity of both planar cell polarity and cell identity during the development of Drosophila. Elife 3: e01569. PubMed ID: 24520160

    Lawrence, P. A., Casal, J. (2013) The mechanisms of planar cell polarity, growth and the Hippo pathway: Some known unknowns. Dev Biol 377: 1-8. PubMed ID: 23592229

    Fabre, C. C., Hedwig, B., Conduit, G., Lawrence, P. A., Goodwin, S. F. and Casal, J. (2012). Substrate-borne vibratory communication during courtship in Drosophila melanogaster. Curr Biol 22: 2180-2185. PubMed ID: 23103187

    Struhl, G., Casal, J. and Lawrence, P. A. (2012). Dissecting the molecular bridges that mediate the function of Frizzled in planar cell polarity. Development 139: 3665-3674. PubMed ID: 22949620

    Krzemien, J., Fabre, C. C., Casal, J. and Lawrence, P. A. (2012). The muscle pattern of the Drosophila abdomen depends on a subdivision of the anterior compartment of each segment. Development 139: 75-83. PubMed ID: 22147953

  • Andreu Casali Institute for Research in Biomedicine, Barcelona
    Campbell, K., Rossi, F., Adams, J., Pitsidianaki, I., Barriga, F. M., Garcia-Gerique, L., Batlle, E., Casanova, J. and Casali, A. (2019). Collective cell migration and metastases induced by an epithelial-to-mesenchymal transition in Drosophila intestinal tumors. Nat Commun 10(1): 2311. PubMed ID: 31127094

    Rahman, M. M., Franch-Marro, X., Maestro, J. L., Martin, D. and Casali, A. (2017). Local Juvenile Hormone activity regulates gut homeostasis and tumor growth in adult Drosophila. Sci Rep 7(1): 11677. PubMed ID: 28916802

    Arnal, A., Jacqueline, C., Ujvari, B., Leger, L., Moreno, C., Faugere, D., Tasiemski, A., Boidin-Wichlacz, C., Misse, D., Renaud, F., Montagne, J., Casali, A., Roche, B., Mery, F. and Thomas, F. (2017). Cancer brings forward oviposition in the fly Drosophila melanogaster. Ecol Evol 7(1): 272-276. PubMed ID: 28070290

    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(2): e88413. PubMed ID: 24516653

  • Jordi Casanova Institute for Research in Biomedicine (IRB Barcelona)
    Pino-Jimenez, B., Giannios, P. and Casanova, J. (2023). Polyploidy-associated autophagy promotes larval tracheal histolysis at Drosophila metamorphosis. Autophagy: 1-10. PubMed ID: 37424089

    Fraire-Zamora, J. J., Tosi, S., Solon, J. and Casanova, J. (2021). Control of hormone-driven organ disassembly by ECM remodeling and Yorkie-dependent apoptosis. Curr Biol. PubMed ID: 34666006

    Giannios, P. and Casanova, J. (2021). Systemic and local effect of the Drosophila headcase gene and its role in stress protection of Adult Progenitor Cells. PLoS Genet 17(2): e1009362. PubMed ID: 33556132

    Casani, S., Casanova, J. and Llimargas, M. (2020). Unravelling the distinct contribution of cell shape changes and cell intercalation to tissue morphogenesis: the case of the Drosophila trachea. Open Biol 10(11): 200329. PubMed ID: 33234070

    Ozturk-Colak, A., Stephan-Otto Attolini, C., Casanova, J. and Araujo, S. J. (2018). Blimp-1 mediates tracheal lumen maturation in Drosophila melanogaster. Genetics. PubMed ID: 30082278

    Mineo, A., Fuentes, E., Furriols, M. and Casanova, J. (2018). Holes in the plasma membrane mimic Torso-Like perforin in Torso tyrosine kinase receptor activation in the Drosophila embryo. Genetics [Epub ahead of print]. PubMed ID: 30049783

    Hernandez de Madrid, B. and Casanova, J. (2018). GATA factor genes in the Drosophila midgut embryo. PLoS One 13(3): e0193612. PubMed ID: 29518114
    Campbell, K., Lebreton, G., Franch-Marro, X. and Casanova, J. (2018). Differential roles of the Drosophila EMT-inducing transcription factors Snail and Serpent in driving primary tumour growth. PLoS Genet 14(2): e1007167. PubMed ID: 29420531

    Mineo, A., Furriols, M. and Casanova, J. (2017). Transfer of dorsoventral and terminal information from the ovary to the embryo by a common group of eggshell proteins in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28179368

    Mineo, A., Furriols, M. and Casanova, J. (2017). Transfer of dorsoventral and terminal information from the ovary to the embryo by a common group of eggshell proteins in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28179368

    de Miguel, C., Linsler, F., Casanova, J. and Franch-Marro, X. (2016). Genetic basis for the evolution of organ morphogenesis. The case of spalt and cut in development of insect trachea. Development. PubMed ID: 27578790

    Ricolo, D., Deligiannaki, M., Casanova, J. and Araújo, S.J. (2016). Centrosome amplification increases single-cell branching in post-mitotic cells. Curr Biol [Epub ahead of print]. PubMed ID: 27693136

  • Fernando Casares Centro Andaluz de Biología del Desarrollo, Sevilla, Spain
    Miguez, D. G., Iannini, A., Garcia-Morales, D. and Casares, F. (2022). Patterning on the move: the effects of Hh morphogen source movement on signaling dynamics. Development. PubMed ID: 36355083

    Lobo-Cabrera, F. J., Navarro, T., Iannini, A., Casares, F. and Cuetos, A. (2021). Quantitative Relationships Between Growth, Differentiation, and Shape That Control Drosophila Eye Development and Its Variation. Front Cell Dev Biol 9: 681933. PubMed ID: 34350178
    Buchberger, E., Bilen, A., Ayaz, S., Salamanca, D., Matas de Las Heras, C., Niksic, A., Almudi, I., Torres-Oliva, M., Casares, F. and Posnien, N. (2021). Variation in pleiotropic hub gene expression is associated with interspecific differences in head shape and eye size in Drosophila. Mol Biol Evol. PubMed ID: 33386848

    Garcia-Morales, D., Navarro, T., Iannini, A., Pereira, P. S., Miguez, D. G. and Casares, F. (2019). Dynamic Hh signalling can generate temporal information during tissue patterning. Development 146(8). PubMed ID: 30918051

    Magri, M. S., Dominguez-Cejudo, M. A. and Casares, F. (2018). Wnt controls the medial-lateral subdivision of the Drosophila head. Biol Lett 14(7). PubMed ID: 30045903

    Neto, M., Naval-Sanchez, M., Potier, D., Pereira, P. S., Geerts, D., Aerts, S. and Casares, F. (2017). Nuclear receptors connect progenitor transcription factors to cell cycle control. Sci Rep 7(1): 4845. PubMed ID: 28687780

    Al Khatib, A., Siomava, N., Iannini, A., Posnien, N. and Casares, F. (2017). Specific expression and function of the Six3 optix in Drosophila serially homologous organs. Biol Open. PubMed ID: 28642242

    Vollmer, J., Fried, P., Aguilar-Hidalgo, D., Sanchez-Aragon, M., Iannini, A., Casares, F. and Iber, D. (2017). Growth control in the Drosophila eye disc by the cytokine Unpaired. Development 144(5): 837-843. PubMed ID: 28246213

    Martins, T., Eusebio, N., Correia, A., Marinho, J., Casares, F. and Pereira, P. S. (2017). TGFbeta/Activin signalling is required for ribosome biogenesis and cell growth in Drosophila salivary glands. Open Biol 7(1). PubMed ID: 28123053

    Fried, P., Sanchez-Aragon, M., Aguilar-Hidalgo, D., Lehtinen, B., Casares, F. and Iber, D. (2016). A model of the spatio-temporal dynamics of Drosophila eye disc development. PLoS Comput Biol 12: e1005052. PubMed ID: 27626238

  • Sergio Casas-Tinto Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, CSIC, Madrid
    Jarabo, P., de Pablo, C., Gonzalez-Blanco, A. and Casas-Tinto, S. (2022). Circadian Gene cry Controls Tumorigenesis through Modulation of Myc Accumulation in Glioblastoma Cells. Int J Mol Sci 23(4). PubMed ID: 35216153

    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

    Portela, M., Mitchell, T. and Casas-Tinto, S. (2020). Cell-to-cell communication mediates glioblastoma progression in Drosophila. Biol Open 9(9). PubMed ID: 32878880

    Santana, E., de Los Reyes, T. and Casas-Tinto, S. (2020). Small heat shock proteins determine synapse number and neuronal activity during development. PLoS One 15(5): e0233231. PubMed ID: 32437379

    Arnes, M., Romero, N., Casas-Tinto, S., Acebes, A. and Ferrus, A. (2020). PI3K activation prevents Abeta42-induced synapse loss and favors insoluble amyloid deposit formation. Mol Biol Cell 31(4): 244-260. PubMed ID: 31877058

    Portela, M., Venkataramani, V., Fahey-Lozano, N., Seco, E., Losada-Perez, M., Winkler, F. and Casas-Tinto, S. (2019). Glioblastoma cells vampirize WNT from neurons and trigger a JNK/MMP signaling loop that enhances glioblastoma progression and neurodegeneration. PLoS Biol 17(12): e3000545. PubMed ID: 31846454

  • Ruben Cauchi University of Malta
    Borg, R., Purkiss, A., Cacciottolo, R., Herrera, P. and Cauchi, R. J. (2023). Loss of amyotrophic lateral sclerosis risk factor SCFD1 causes motor dysfunction in Drosophila. Neurobiol Aging 126: 67-76. PubMed ID: 36944290

    Farrugia, M., Vassallo, N. and Cauchi, R. J. (2022). Disruption of Survival Motor Neuron in Glia Impacts Survival but has no Effect on Neuromuscular Function in Drosophila. Neuroscience 491: 32-42. PubMed ID: 35314252

    Aquilina, B. and Cauchi, R. J. (2018). Genetic screen identifies a requirement for SMN in mRNA localisation within the Drosophila oocyte. BMC Res Notes 11(1): 378. PubMed ID: 29895323

    Borg, R. M., Fenech Salerno, B., Vassallo, N., Bordonne, R. and Cauchi, R. J. (2016). Disruption of snRNP biogenesis factors Tgs1 and pICln induces phenotypes that mirror aspects of SMN-Gemins complex perturbation in Drosophila, providing new insights into spinal muscular atrophy. Neurobiol Dis 94: 245-258. PubMed ID: 27388936

    Borg, R. M., Bordonne, R., Vassallo, N. and Cauchi, R. J. (2015). Genetic interactions between the members of the SMN-Gemins complex in Drosophila. PLoS One 10: e0130974. PubMed ID: 26098872

    Borg, R. and Cauchi, R. J. (2013). The gemin associates of survival motor neuron are required for motor function in Drosophila. PLoS One 8: e83878. PubMed ID: 24391840

    Cauchi, R. J. (2012). Conserved requirement for DEAD-box RNA helicase Gemin3 in Drosophila oogenesis. BMC Res Notes 5: 120. PubMed ID: 22361416

    Cauchi, R. J. (2011). Gem formation upon constitutive Gemin3 overexpression in Drosophila. Cell Biol Int 35: 1233-1238. PubMed ID: 21627586

  • Valeria Cavaliere Department of Pharmacy and Biotechnology, Università di Bologna
    Giordani, G., Cattabriga, G., Becchimanzi, A., Di Lelio, I., De Leva, G., Gigliotti, S., Pennacchio, F., Gargiulo, G. and Cavaliere, V. (2022). Role of neuronal and non-neuronal acetylcholine signaling in Drosophila humoral immunity. Insect Biochem Mol Biol 153: 103899. PubMed ID: 36596348

    Mezzofanti, E., Ignesti, M., Hsu, T., Gargiulo, G. and Cavaliere, V. (2019). Vps28 is involved in the intracellular trafficking of Awd, the Drosophila homolog of NME1/2. Front Physiol 10: 983. PubMed ID: 31427986

    Romani, P., Duchi, S., Gargiulo, G. and Cavaliere, V. (2017). Evidence for a novel function of Awd in maintenance of genomic stability. Sci Rep 7(1): 16820. PubMed ID: 29203880

    Valzania, L., Ono, H., Ignesti, M., Cavaliere, V., Bernardi, F., Gamberi, C., Lasko, P. and Gargiulo, G. (2016). Drosophila 4EHP is essential for the larval-pupal transition and required in the prothoracic gland for ecdysone biosynthesis. Dev Biol 410(1): 14-23. PubMed ID: 26721418

    Ignesti, M., Barraco, M., Nallamothu, G., Woolworth, J. A., Duchi, S., Gargiulo, G., Cavaliere, V. and Hsu, T. (2014). Notch signaling during development requires the function of awd, the Drosophila homolog of human metastasis suppressor gene Nm23. BMC Biol 12: 12. PubMed ID: 24528630

  • Giacomo Cavalli Institute of Human Genetics, Montpellier, France
    Bourbon, H. G., Benetah, M. H., Guillou, E., Mojica-Vazquez, L. H., Baanannou, A., Bernat-Fabre, S., Loubiere, V., Bantignies, F., Cavalli, G. and Boube, M. (2022). A shared ancient enhancer element differentially regulates the bric-a-brac tandem gene duplicates in the developing Drosophila leg. PLoS Genet 18(3): e1010083. PubMed ID: 35294439

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

    Loubiere, V., Papadopoulos, G. L., Szabo, Q., Martinez, A. M. and Cavalli, G. (2020). Widespread activation of developmental gene expression characterized by PRC1-dependent chromatin looping. Sci Adv 6(2): eaax4001. PubMed ID: 31950077

    Pal, K., Forcato, M., Jost, D., Sexton, T., Vaillant, C., Salviato, E., Mazza, E. M. C., Lugli, E., Cavalli, G. and Ferrari, F. (2019). Global chromatin conformation differences in the Drosophila dosage compensated chromosome X. Nat Commun 10(1): 5355. PubMed ID: 31767860

    Cardozo Gizzi, A. M., Cattoni, D. I., Fiche, J. B., Espinola, S. M., Gurgo, J., Messina, O., Houbron, C., Ogiyama, Y., Papadopoulos, G. L., Cavalli, G., Lagha, M. and Nollmann, M. (2019). Microscopy-Based Chromosome Conformation Capture Enables Simultaneous Visualization of Genome Organization and Transcription in Intact Organisms. Mol Cell 74(1): 212-222.e215. PubMed ID: 30795893

    Cheutin, T. and Cavalli, G. (2018). Loss of PRC1 induces higher-order opening of Hox loci independently of transcription during Drosophila embryogenesis. Nat Commun 9(1): 3898. PubMed ID: 30254245

    Ogiyama, Y., Schuettengruber, B., Papadopoulos, G. L., Chang, J. M. and Cavalli, G. (2018). Polycomb-dependent chromatin looping contributes to gene silencing during Drosophila development. Mol Cell 71(1): 73-88.e75. PubMed ID: 30008320

    Szabo, Q., Jost, D., Chang, J. M., Cattoni, D. I., Papadopoulos, G. L., Bonev, B., Sexton, T., Gurgo, J., Jacquier, C., Nollmann, M., Bantignies, F. and Cavalli, G. (2018). TADs are 3D structural units of higher-order chromosome organization in Drosophila. Sci Adv 4(2): eaar8082. PubMed ID: 29503869

    Cattoni, D. I., Gizzi, A. M. C., Georgieva, M., Di Stefano, M., Valeri, A., Chamousset, D., Houbron, C., Dejardin, S., Fiche, J. B., Gonzalez, I., Chang, J. M., Sexton, T., Marti-Renom, M. A., Bantignies, F., Cavalli, G. and Nollmann, M. (2017). Single-cell absolute contact probability detection reveals chromosomes are organized by multiple low-frequency yet specific interactions. Nat Commun 8(1): 1753. PubMed ID: 29170434

    Schauer, T., Ghavi-Helm, Y., Sexton, T., Albig, C., Regnard, C., Cavalli, G., Furlong, E. E. and Becker, P. B. (2017). Chromosome topology guides the Drosophila Dosage Compensation Complex for target gene activation. EMBO Rep. PubMed ID: 28794204

    Ciabrelli, F., Comoglio, F., Fellous, S., Bonev, B., Ninova, M., Szabo, Q., Xuereb, A., Klopp, C., Aravin, A., Paro, R., Bantignies, F. and Cavalli, G. (2017). Stable Polycomb-dependent transgenerational inheritance of chromatin states in Drosophila. Nat Genet 49(6): 876-886. PubMed ID: 28436983

  • Arzu Celik Department of Molecular Biology and Genetics, Istanbul
    Sahin, H. B., Sayin, S., Holder, M., Bugra, K. and Celik, A. (2020). Salt inducible kinases as novel Notch interactors in the developing Drosophila retina. PLoS One 15(6): e0234744. PubMed ID: 32542037

    Terzioglu Kara, E., Kiral, F. R., Ozturk Colak, A. and Celik, A. (2020). Generation and characterization of inner photoreceptor-specific enhancer-trap lines using a novel piggyBac-Gal4 element in Drosophila. Arch Insect Biochem Physiol 104(2): e21675. PubMed ID: 32285519

    Koestler, S. A., Alaybeyoglu, B., Weichenberger, C. X. and Celik, A. (2015). FlyOde - a platform for community curation and interactive visualization of dynamic gene regulatory networks in Drosophila eye development. F1000Res 4: 1484. PubMed ID: 26998229

  • Susan Celniker Genome Dynamics Department, Lawrence Berkeley Lab
    Fisher, W. W., Hammonds, A. S., Weiszmann, R., Booth, B. W., Gevirtzman, L., Patton, J., Kubo, C., Waterston, R. H. and Celniker, S. E. (2023). A modERN Resource: Identification of Drosophila Transcription Factor candidate target genes using RNAi. Genetics. PubMed ID: 36652461

    Brown, J. B., Langley, S. A., Snijders, A. M., Wan, K. H., Morris, S. N. S., Booth, B. W., Fisher, W. W., Hammonds, A. S., Park, S., Weiszmann, R., Yu, C., Kirwan, J. A., Weber, R. J. M., Viant, M. R., Mao, J. H. and Celniker, S. E. (2021). An integrated host-microbiome response to atrazine exposure mediates toxicity in Drosophila. Commun Biol 4(1): 1324. PubMed ID: 34819611

    Chen, K., Luan, X., Liu, Q., Wang, J., Chang, X., Snijders, A. M., Mao, J. H., Secombe, J., Dan, Z., Chen, J. H., Wang, Z., Dong, X., Qiu, C., Chang, X., Zhang, D., Celniker, S. E. and Liu, X. (2019). Drosophila Histone Demethylase KDM5 Regulates Social Behavior through Immune Control and Gut Microbiota Maintenance. Cell Host Microbe 25(4): 537-552.e538. PubMed ID: 30902578

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

    Wu, S., Joseph, A., Hammonds, A. S., Celniker, S. E., Yu, B. and Frise, E. (2016). Stability-driven nonnegative matrix factorization to interpret spatial gene expression and build local gene networks. Proc Natl Acad Sci U S A 113: 4290-4295. PubMed ID: 27071099

    Brooks, A. N., Duff, M. O., May, G., Yang, L., Bolisetty, M., Landolin, J., Wan, K., Sandler, J., Celniker, S. E., Graveley, B. R. and Brenner, S. E. (2015). Regulation of alternative splicing in Drosophila by 56 RNA binding proteins. Genome Res [Epub ahead of print]. PubMed ID: 26294686

    Stoiber, M. H., Olson, S., May, G. E., Duff, M. O., Manent, J., Obar, R., Guruharsha, K., Artavanis-Tsakonas, S., Brown, J. B., Graveley, B. R. and Celniker, S. E. (2015). Extensive cross-regulation of post-transcriptional regulatory networks in Drosophila. Genome Res [Epub ahead of print]. PubMed ID: 26294687

    Brown, J. B. and Celniker, S. E. (2015). Lessons from modENCODE. Annu Rev Genomics Hum Genet [Epub ahead of print]. PubMed ID: 26133010

    Duff, M.O., Olson, S., Wei, X., Garrett, S.C., Osman, A., Bolisetty, M., Plocik, A., Celniker, S.E. and Graveley, B.R. (2015). Genome-wide identification of zero nucleotide recursive splicing in Drosophila. Nature [Epub ahead of print]. PubMed ID: 25970244

    Hoskins, R. A., et al. (2015). The Release 6 reference sequence of the Drosophila melanogaster genome. Genome Res. PubMed ID: 25589440

  • Giovanni Cenci Drosophila Genetics Dipartimento di Scienze Cliniche Applicate e biotecnologiche, University of L'Aquila, Italy
    Bosso, G., Cipressa, F., Tullo, L. and Cenci, G. (2023). Co-amplification of CBX3 with EGFR or RAC1 in human cancers corroborated by a conserved genetic interaction among the genes. Cell Death Discov 9(1): 317. PubMed ID: 37633946

    Porrazzo, A., Cipressa, F., De Gregorio, A., De Pitta, C., Sales, G., Ciapponi, L., Morciano, P., Esposito, G., Tabocchini, M. A. and Cenci, G. (2022). Low dose rate γ-irradiation protects fruit fly chromosomes from double strand breaks and telomere fusions by reducing the esi-RNA biogenesis factor Loquacious. Commun Biol 5(1): 905. PubMed ID: 36057690

    Di Giorgio, M. L., Morciano, P., Bucciarelli, E., Porrazzo, A., Cipressa, F., Saraniero, S., Manzi, D., Rong, Y. S. and Cenci, G. (2020). The Drosophila citrate lyase is required for cell division during spermatogenesis. Cells 9(1). PubMed ID: 31947614

    Bosso, G., Cipressa, F., Moroni, M. L., Pennisi, R., Albanesi, J., Brandi, V., Cugusi, S., Renda, F., Ciapponi, L., Polticelli, F., Antoccia, A., di Masi, A. and Cenci, G. (2019). NBS1 interacts with HP1 to ensure genome integrity. Cell Death Dis 10(12): 951. PubMed ID: 31836699

    Morciano, P., Di Giorgio, M. L., Porrazzo, A., Licursi, V., Negri, R., Rong, Y. and Cenci, G. (2019). Depletion of ATP-Citrate Lyase (ATPCL) affects chromosome integrity without altering histone acetylation in Drosophila mitotic cells. Front Physiol 10: 383. PubMed ID: 31019471

    Cicconi, A., Micheli, E., Verni, F., Jackson, A., Gradilla, A. C., Cipressa, F., Raimondo, D., Bosso, G., Wakefield, J. G., Ciapponi, L., Cenci, G., Gatti, M., Cacchione, S. and Raffa, G. D. (2016). The Drosophila telomere-capping protein Verrocchio binds single-stranded DNA and protects telomeres from DNA damage response. Nucleic Acids Res. PubMed ID: 27940556

    Cipressa, F., Morciano, P., Bosso, G., Mannini, L., Galati, A., Daniela Raffa, G., Cacchione, S., Musio, A. and Cenci, G. (2016). A role for Separase in telomere protection. Nat Commun 7: 10405. PubMed ID: 26778495

    Cenci, G., Ciapponi, L., Marzullo, M., Raffa, G.D., Morciano, P., Raimondo, D., Burla, R., Saggio, I. and Gatti, M (2015). The analysis of pendolino (peo) mutants reveals differences in the fusigenic potential among Drosophila telomeres. PLoS Genet 11: e1005260. PubMed ID: 26110638

    Vernì, F. and Cenci, G. (2015). The Drosophila histone variant H2A.V works in concert with HP1 to promote kinetochore-driven microtubule formation. Cell Cycle. [Epub ahead of print]. PubMed ID: 25591068

    Cipressa, F., Di Giorgio, M. L. and Cenci, G. (2013). A Simple Approach for Multicolor Immunofluorescence Staining in Different Drosophila Cell Types. J Cell Physiol. PubMed ID: 24170430

  • Maria Fernanda Ceriani Facultad de Ciencias Exactas y Naturales de la Universidad de Buenos Aires
    Fernandez-Acosta, M., Romero, J. I., Bernabo, G., Velazquez-Campos, G. M., Gonzalez, N., Mares, M. L., Werbajh, S., Avendano-Vazquez, L. A., Rechberger, G. N., KUhnlein, R. P., Marino-Buslje, C., Cantera, R., Rezaval, C. and Ceriani, M. F. (2022). orsai, the Drosophila homolog of human ETFRF1, links lipid catabolism to growth control. BMC Biol 20(1): 233. PubMed ID: 36266680

    Polcowñuk, S., Yoshii, T. and Ceriani, M. F. (2021). DPP acutely defines the connectivity of central pacemaker neurons in Drosophila. J Neurosci. PubMed ID: 34429376

    Fernandez-Chiappe, F., Frenkel, L., Colque, C. C., Ricciuti, A., Hahm, B., Cerredo, K., Muraro, N. I. and Ceriani, M. F. (2020). High frequency neuronal bursting is essential for circadian and sleep behaviors in Drosophila. J Neurosci. PubMed ID: 33262246

    Duhart, J. M., Herrero, A., de la Cruz, G., Ispizua, J. I., Pírez, N. and Ceriani, M. F. (2020). Circadian Structural Plasticity Drives Remodeling of E Cell Output. Curr Biol. PubMed ID: 33065014

    Herrero, A., Yoshii, T., Ispizua, J. I., Colque, C., Veenstra, J. A., Muraro, N. I. and Ceriani, M. F. (2020). Coupling Neuropeptide Levels to Structural Plasticity in Drosophila Clock Neurons. Curr Biol. PubMed ID: 32619484

    Pirez, N., Bernabei-Cornejo, S. G., Fernandez-Acosta, M., Duhart, J. M. and Ceriani, M. F. (2018). Contribution of non-circadian neurons to the temporal organization of locomotor activity. Biol Open. PubMed ID: 30530810

    Herrero, A., Duhart, J. M. and Ceriani, M. F. (2017). Neuronal and glial clocks underlying structural remodeling of pacemaker neurons in Drosophila. Front Physiol 8: 918. PubMed ID: 29184510

    Beckwith, E. J., Hernando, C. E., Polcownuk, S., Bertolin, A. P., Mancini, E., Ceriani, M. F. and Yanovsky, M. J. (2017). Rhythmic behavior is controlled by the SRm160 splicing factor in Drosophila melanogaster. Genetics. PubMed ID: 28801530

    Frenkel, L., Muraro, N. I., Beltran Gonzalez, A. N., Marcora, M. S., Bernabo, G., Hermann-Luibl, C., Romero, J. I., Helfrich-Forster, C., Castano, E. M., Marino-Busjle, C., Calvo, D. J. and Ceriani, M. F. (2017). Organization of circadian behavior relies on glycinergic transmission. Cell Rep 19(1): 72-85. PubMed ID: 28380364

    Muraro, N. I. and Ceriani, M. F. (2015). Acetylcholine from Visual Circuits Modulates the Activity of Arousal Neurons in Drosophila. J Neurosci 35: 16315-16327. PubMed ID: 26674859

  • Sarah Certel University of Montana, Missoula
    Certel, S. J., McCabe, B. D. and Stowers, R. S. (2022). A conditional GABAergic synaptic vesicle marker for Drosophila. J Neurosci Methods 372: 109540. PubMed ID: 35219770

    Sherer, L. M., Catudio Garrett, E., Morgan, H. R., Brewer, E. D., Sirrs, L. A., Shearin, H. K., Williams, J. L., McCabe, B. D., Stowers, R. S. and Certel, S. J. (2020). Octopamine neuron dependent aggression requires dVGLUT from dual-transmitting neurons. PLoS Genet 16(2): e1008609. PubMed ID: 32097408

    McKinney, H., Sherer, L. M., Williams, J. L., Certel, S. J. and Stowers, R. S. (2020). Characterization of Drosophila Octopamine Receptor Neuronal Expression using MiMIC-converted Gal4 lines. J Comp Neurol. PubMed ID: 32060912

    Gupta, T., Morgan, H. R., Andrews, J. C., Brewer, E. R. and Certel, S. J. (2017). Methyl-CpG binding domain proteins inhibit interspecies courtship and promote aggression in Drosophila. Sci Rep 7(1): 5420. PubMed ID: 28710457

    Gupta, T., Morgan, H. R., Bailey, J. A. and Certel, S. J. (2016). Functional conservation of MBD proteins: MeCP2 and Drosophila MBD proteins alter sleep. Genes Brain Behav. PubMed ID: 27489246

    Hess-Homeier, D. L., Fan, C. Y., Gupta, T., Chiang, A. S. and Certel, S. J. (2014). Astrocyte-specific regulation of hMeCP2 expression in Drosophila. Biol Open. PubMed ID: 25305037

    Andrews, J. C., Fernandez, M. P., Yu, Q., Leary, G. P., Leung, A. K., Kavanaugh, M. P., Kravitz, E. A. and Certel, S. J. (2014). Octopamine neuromodulation regulates gr32a-linked aggression and courtship pathways in Drosophila males. PLoS Genet 10: e1004356. PubMed ID: 24852170

    Burke, C. J., Huetteroth, W., Owald, D., Perisse, E., Krashes, M. J., Das, G., Gohl, D., Silies, M., Certel, S. and Waddell, S. (2012). Layered reward signalling through octopamine and dopamine in Drosophila. Nature 492: 433-437. PubMed ID: 23103875

    Certel, S. J. and Kravitz, E. A. (2012). Scoring and analyzing aggression in Drosophila. Cold Spring Harb Protoc 2012: 319-325. PubMed ID: 22383642

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

  • ​Isaac Cervantes-Sandoval
    Martinez-Cervantes, J., Shah, P., Phan, A. and Cervantes-Sandoval, I. (2022). Higher-order unimodal olfactory sensory preconditioning in Drosophila. Elife 11. PubMed ID: 36129180

    >Roselli, C., Ramaswami, M., Boto, T. and Cervantes-Sandoval, I. (2021). The Making of Long-Lasting Memories: A Fruit Fly Perspective. Front Behav Neurosci 15: 662129. PubMed ID: 33859556

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

  • Ronald Chalmers University of Nottingham
    Liu, D. and Chalmers, R. (2013). Hyperactive mariner transposons are created by mutations that disrupt allosterism and increase the rate of transposon end synapsis. Nucleic Acids Res. PubMed ID: 24319144

    Marzo, M., Liu, D., Ruiz, A. and Chalmers, R. (2013). Identification of multiple binding sites for the THAP domain of the Galileo transposase in the long terminal inverted-repeats. Gene. PubMed ID: 23648487

    Munoz-Lopez, M., Siddique, A., Bischerour, J., Lorite, P., Chalmers, R. and Palomeque, T. (2008). Transposition of Mboumar-9: identification of a new naturally active mariner-family transposon. J Mol Biol 382: 567-572. PubMed ID: 18675277

  • Severine Chambeyron Institute of Human Genetics, CNRS, Montpellier, France
    Mohamed, M., Dang, N. T., Ogyama, Y., Burlet, N., Mugat, B., Boulesteix, M., Mérel, V., Veber, P., Salces-Ortiz, J., Severac, D., Pélisson, A., Vieira, C., Sabot, F., Fablet, M. and Chambeyron, S. (2020). A Transposon Story: From TE Content to TE Dynamic Invasion of Drosophila Genomes Using the Single-Molecule Sequencing Technology from Oxford Nanopore. Cells 9(8). PubMed ID: 32722451

    Mugat, B., Nicot, S., Varela-Chavez, C., Jourdan, C., Sato, K., Basyuk, E., Juge, F., Siomi, M. C., Pelisson, A. and Chambeyron, S. (2020). The Mi-2 nucleosome remodeler and the Rpd3 histone deacetylase are involved in piRNA-guided heterochromatin formation. Nat Commun 11(1): 2818. PubMed ID: 32499524

    Barckmann, B., El-Barouk, M., Pelisson, A., Mugat, B., Li, B., Franckhauser, C., Fiston Lavier, A. S., Mirouze, M., Fablet, M. and Chambeyron, S. (2018). The somatic piRNA pathway controls germline transposition over generations. Nucleic Acids Res 46(18): 9524-9536. PubMed ID: 30312469

    Akkouche, A., Mugat, B., Barckmann, B., Varela-Chavez, C., Li, B., Raffel, R., Pelisson, A. and Chambeyron, S. (2017). Piwi is required during Drosophila embryogenesis to license dual-strand piRNA clusters for transposon repression in adult ovaries. Mol Cell 66(3): 411-419.e414. PubMed ID: 28457744

    Lopez-Maestre, H., Carnelossi, E. A., Lacroix, V., Burlet, N., Mugat, B., Chambeyron, S., Carareto, C. M. and Vieira, C. (2017). Identification of misexpressed genetic elements in hybrids between Drosophila-related species. Sci Rep 7: 40618. PubMed ID: 28091568

    Mugat, B., Akkouche, A., Serrano, V., Armenise, C., Li, B., Brun, C., Fulga, T. A., Van Vactor, D., Pelisson, A. and Chambeyron, S. (2015). MicroRNA-Dependent Transcriptional Silencing of Transposable Elements in Drosophila Follicle Cells. PLoS Genet 11(5): e1005194. PubMed ID: 25993106

  • Ho Yin Edwin Chan School of Life Sciences, The Chinese University of Hong Kong
    Chen, Z. S., Wong, A. K. Y., Cheng, T. C., Koon, A. C. and Chan, H. Y. E. (2019). FipoQ/FBXO33, a Cullin-1 based ubiquitin ligase complex component modulates ubiquitination and solubility of polyglutamine disease protein. J Neurochem. PubMed ID: 30685895

    Koon, A. C., Chen, Z. S., Peng, S., Fung, J. M. S., Zhang, X., Lembke, K. M., Chow, H. K., Frank, C. A., Jiang, L., Lau, K. F. and Chan, H. Y. E. (2018). Drosophila Exo70 Is Essential for Neurite Extension and Survival under Thermal Stress. J Neurosci 38(37): 8071-8086. PubMed ID: 30209205

    Chen, Z. S., Li, L., Peng, S., Chen, F. M., Zhang, Q., An, Y., Lin, X., Li, W., Koon, A. C., Chan, T. F., Lau, K. F., Ngo, J. C. K., Wong, W. T., Kwan, K. M. and Chan, H. Y. E. (2018). Planar cell polarity gene Fuz triggers apoptosis in neurodegenerative disease models. EMBO Rep. PubMed ID: 30026307

    Zhang, Q., Tsoi, H., Peng, S., Li, P. P., Lau, K. F., Rudnicki, D. D., Ngo, J. C. and Chan, H. Y. (2016). A peptidylic inhibitor-based therapeutic approach that simultaneously suppresses RNA- and protein-mediated toxicities in polyglutamine diseases. Dis Model Mech [Epub ahead of print]. PubMed ID: 26839389

    Ng, C. F., Ko, C. H., Koon, C. M., Xian, J. W., Leung, P. C., Fung, K. P., Chan, H. Y. and Lau, C. B. (2013). The Aqueous Extract of Rhizome of Gastrodia elata Protected Drosophila and PC12 Cells against Beta-Amyloid-Induced Neurotoxicity. Evid Based Complement Alternat Med 2013: 516741. PubMed ID: 24174977

    Peng, C., Zuo, Y., Kwan, K. M., Liang, Y., Ma, K. Y., Chan, H. Y., Huang, Y., Yu, H. and Chen, Z. Y. (2012). Blueberry extract prolongs lifespan of Drosophila melanogaster. Exp Gerontol 47: 170-178. PubMed ID: 22197903

    Peng, C., Chan, H. Y., Huang, Y., Yu, H. and Chen, Z. Y. (2011). Apple polyphenols extend the mean lifespan of Drosophila melanogaster. J Agric Food Chem 59: 2097-2106. PubMed ID: 21319854

  • Henry Chang Department of Biological Sciences, Purdue University
    Ready, D. F. and Chang, H. C. (2023). Interommatidial cells build a tensile collagen network during Drosophila retinal morphogenesis. Curr Biol 33(11): 2223-2234. PubMed ID: 37209679

    Ready, D. F. and Chang, H. C. (2021). Calcium waves facilitate and coordinate the contraction of endfeet actin stress fibers in Drosophila interommatidial cells. Development. PubMed ID: 34698814

    Abdallah, A. M., Zhou, X., Kim, C., Shah, K. K., Hogden, C., Schoenherr, J. A., Clemens, J. C., Chang, H. C. (2013) Activated Cdc42 kinase regulates Dock localization in male germ cells during Drosophila spermatogenesis. Dev Biol. PubMed ID: 23562806

    Schoenherr, J. A., Drennan, J. M., Martinez, J. S., Chikka, M. R., Hall, M. C., Chang, H. C. and Clemens, J. C. (2012). Drosophila activated Cdc42 kinase has an anti-apoptotic function. PLoS Genet 8: e1002725. PubMed ID: 22615583

    Yang, W. K., Peng, Y. H., Li, H., Lin, H. C., Lin, Y. C., Lai, T. T., Suo, H., Wang, C. H., Lin, W. H., Ou, C. Y., Zhou, X., Pi, H., Chang, H. C. and Chien, C. T. (2011). Nak regulates localization of clathrin sites in higher-order dendrites to promote local dendrite growth. Neuron 72: 285-299. PubMed ID: 22017988

    Burgess, J., Jauregui, M., Tan, J., Rollins, J., Lallet, S., Leventis, P. A., Boulianne, G. L., Chang, H. C., Le Borgne, R., Kramer, H. and Brill, J. A. (2011). AP-1 and clathrin are essential for secretory granule biogenesis in Drosophila. Mol Biol Cell 22: 2094-2105. PubMed ID: 21490149

  • Hui-yun Chang Department of Medical Science, Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan.
    Quinn, J. J., Zhang, Q. C., Georgiev, P., Ilik, I. A., Akhtar, A. and Chang, H. Y. (2016). Rapid evolutionary turnover underlies conserved lncRNA-genome interactions. Genes Dev 30: 191-207. PubMed ID: 26773003

    Quinn, J. J. and Chang, H. Y. (2014). RNA switch at enhancers. Nat Genet 46: 929-931. PubMed ID: 25162802

    Quinn, J. J., Ilik, I. A., Qu, K., Georgiev, P., Chu, C., Akhtar, A. and Chang, H. Y. (2014). Revealing long noncoding RNA architecture and functions using domain-specific chromatin isolation by RNA purification. Nat Biotechnol. PubMed ID: 24997788aW., Cao, L., Lin, X., Ji, S., Huang, S., Zhang, G., Liu, X., Tao, Y., Wu, S. and Chen, D. (2015). Ci antagonizes Hippo signaling in the somatic cells of the ovary to drive germline stem cell differentiation. Cell Res. PubMed ID: 26403189

    Zhang, G., Huang, H., Liu, D., Cheng, Y., Liu, X., Zhang, W., Yin, R., Zhang, D., Zhang, P., Liu, J., Li, C., Liu, B., Luo, Y., Zhu, Y., Zhang, N., He, S., He, C., Wang, H. and Chen, D. (2015). N(6)-methyladenine DNA modification in Drosophila. Cell 161: 893-906. PubMed ID: 25936838

    Huang, H., Li, Y., Szulwach, K. E., Zhang, G., Jin, P. and Chen, D. (2014). AGO3 Slicer activity regulates mitochondria-nuage localization of Armitage and piRNA amplification. J Cell Biol 206: 217-230. PubMed ID: 25049272

    Huang, S., Zhang, Z., Zhang, C., Lv, X., Zheng, X., Chen, Z., Sun, L., Wang, H., Zhu, Y., Zhang, J., Yang, S., Lu, Y., Sun, Q., Tao, Y., Liu, F., Zhao, Y. and Chen, D. (2013). Activation of Smurf E3 ligase promoted by smoothened regulates hedgehog signaling through targeting patched turnover. PLoS Biol 11: e1001721. PubMed ID: 24302888

    Zhang, W., Cheng, Y., Li, Y., Chen, Z., Jin, P. and Chen, D. (2014). A feed-forward mechanism involving Drosophila fragile X mental retardation protein triggers a replication stress-induced DNA damage response. Hum Mol Genet. PubMed ID: 24833720

    Wang, H., Huang, H. and Chen, D. (2014). Effective Gene Knockdown in the Drosophila Germline by Artificial miRNA-Mimicking siRNAs. Methods Mol Biol 1093: 225-234. PubMed ID: 24178569

  • Elizabeth Chen Johns Hopkins University School of Medicine, Department of Molecular Biology & Genetics, Baltimore
    Kim, J. H. and Chen, E. H. (2019). The fusogenic synapse at a glance. J Cell Sci 132(18). PubMed ID: 31527149

    Duan, R., Kim, J. H., Shilagardi, K., Schiffhauer, E. S., Lee, D. M., Son, S., Li, S., Thomas, C., Luo, T., Fletcher, D. A., Robinson, D. N. and Chen, E. H. (2018). Spectrin is a mechanoresponsive protein shaping fusogenic synapse architecture during myoblast fusion. Nat Cell Biol 20(6): 688-698. PubMed ID: 29802406

    Kim, J. H., Ren, Y., Ng, W. P., Li, S., Son, S., Kee, Y. S., Zhang, S., Zhang, G., Fletcher, D. A., Robinson, D. N. and Chen, E. H. (2015). Mechanical tension drives cell membrane fusion. Dev Cell 32: 561-573. PubMed ID: 25684354

    Duan, R., Jin, P., Luo, F., Zhang, G., Anderson, N. and Chen, E. H. (2012). Group I PAKs function downstream of Rac to promote podosome invasion during myoblast fusion in vivo. J Cell Biol 199: 169-185. PubMed ID: 23007650

    Jin, P., Duan, R., Luo, F., Zhang, G., Hong, S. N. and Chen, E. H. (2011). Competition between Blown fuse and WASP for WIP binding regulates the dynamics of WASP-dependent actin polymerization in vivo. Dev Cell 20: 623-638. PubMed ID: 21571220

    Sens, K. L., Zhang, S., Jin, P., Duan, R., Zhang, G., Luo, F., Parachini, L. and Chen, E. H. (2010). An invasive podosome-like structure promotes fusion pore formation during myoblast fusion. J Cell Biol 191: 1013-1027. PubMed ID: 21098115

  • Elizabeth Chen Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine
    Shilagardi, K., Li, S., Luo, F., Marikar, F., Duan, R., Jin, P., Kim, J. H., Murnen, K., Chen, E. H. (2013) Actin-Propelled Invasive Membrane Protrusions Promote Fusogenic Protein Engagement During Cell-Cell Fusion. Science. PubMed ID: 23470732

    Duan, R., Jin, P., Luo, F., Zhang, G., Anderson, N. and Chen, E. H. (2012). Group I PAKs function downstream of Rac to promote podosome invasion during myoblast fusion in vivo. J Cell Biol 199: 169-185. PubMed ID: 23007650

    Jin, P., Duan, R., Luo, F., Zhang, G., Hong, S. N. and Chen, E. H. (2011). Competition between Blown fuse and WASP for WIP binding regulates the dynamics of WASP-dependent actin polymerization in vivo. Dev Cell 20: 623-638. PubMed ID: 21571220

    Sens, K. L., Zhang, S., Jin, P., Duan, R., Zhang, G., Luo, F., Parachini, L. and Chen, E. H. (2010). An invasive podosome-like structure promotes fusion pore formation during myoblast fusion. J Cell Biol 191: 1013-1027. PubMed ID: 21098115

  • Guang-Chao Chen Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
    Pai, Y. L., Lin, Y. J., Peng, W. H., Huang, L. T., Chou, H. Y., Wang, C. H., Chien, C. T. and Chen, G. C. (2023). The deubiquitinase Leon/USP5 interacts with Atg1/ULK1 and antagonizes autophagy. Cell Death Dis 14(8): 540. PubMed ID: 37607937

    Wen, J. K., Wang, Y. T., Chan, C. C., Hsieh, C. W., Liao, H. M., Hung, C. C. and Chen, G. C. (2017). Atg9 antagonizes TOR signaling to regulate intestinal cell growth and epithelial homeostasis in Drosophila. Elife 6. PubMed ID: 29144896

    Amcheslavsky, A., Song, W., Li, Q., Nie, Y., Bragatto, I., Ferrandon, D., Perrimon, N. and Ip, Y. T. (2014). Enteroendocrine Cells Support Intestinal Stem-Cell-Mediated Homeostasis in Drosophila. Cell Rep. PubMed ID: 25263551

    Chen, D. Y., Li, M. Y., Wu, S. Y., Lin, Y. L., Tsai, S. P., Lai, P. L., Lin, Y. T., Kuo, J. C., Meng, T. C. and Chen, G. C. (2012). The Bro1-domain-containing protein Myopic/HDPTP coordinates with Rab4 to regulate cell adhesion and migration. J Cell Sci 125: 4841-4852. PubMed ID: 22825871

    Santhanam, A., Liang, S. Y., Chen, D. Y., Chen, G. C. and Meng, T. C. (2013). Midgut-enriched receptor protein tyrosine phosphatase PTP52F is required for Drosophila development during larva-pupa transition. FEBS J 280: 476-488. PubMed ID: 22780963

    Chen, S. F., Kang, M. L., Chen, Y. C., Tang, H. W., Huang, C. W., Li, W. H., Lin, C. P., Wang, C. Y., Wang, P. Y., Chen, G. C. and Wang, H. D. (2012). Autophagy-related gene 7 is downstream of heat shock protein 27 in the regulation of eye morphology, polyglutamine toxicity, and lifespan in Drosophila. J Biomed Sci 19: 52. PubMed ID: 22621211

    Tang, H. W. and Chen, G. C. (2011). Unraveling the role of myosin in forming autophagosomes. Autophagy 7: 778-779. PubMed ID: 21460626

  • Jiong Chen Model Animal Research Center of Nanjing University
    Huang, Z., Wang, W., Xu, P., Gong, S., Hu, Y., Liu, Y., Su, F., Anjum, K. M., Deng, W. M., Yang, S., Liu, J., Jiao, R. and Chen, J. (2023). Drosophila Ectoderm-expressed 4 modulates JAK/STAT pathway and protects flies against Drosophila C virus infection. Front Immunol 14: 1135625. PubMed ID: 36817462

    Qu, C., Yang, W., Kan, Y., Zuo, H., Wu, M., Zhang, Q., Wang, H., Wang, D. and Chen, J. (2022). RhoA/ROCK Signaling Regulates Drp1-Mediated Mitochondrial Fission During Collective Cell Migration. Front Cell Dev Biol 10: 882581. PubMed ID: 35712666

    Wang, X., Wang, H., Liu, L., Li, S., Emery, G. and Chen, J. (2020). Temporal Coordination of Collective Migration and Lumen Formation by Antagonism between Two Nuclear Receptors. iScience 23(7): 101335. PubMed ID: 32682323

    Wang, H., Guo, X., Wang, X., Wang, X. and Chen, J. (2020). Supracellular Actomyosin Mediates Cell-Cell Communication and Shapes Collective Migratory Morphology. iScience 23(6): 101204. PubMed ID: 32535019

    Luo, J., Zhou, P., Guo, X., Wang, D. and Chen, J. (2019). The polarity protein Dlg5 regulates collective cell migration during Drosophila oogenesis. PLoS One 14(12): e0226061. PubMed ID: 31856229

    Wang, H., Qiu, Z., Xu, Z., Chen, S. J., Luo, J., Wang, X. and Chen, J. (2018). aPKC is a key polarity molecule coordinating the function of three distinct cell polarities during collective migration. Development. PubMed ID: 29636381

    Kang, D., Wang, D., Xu, J., Quan, C., Guo, X., Wang, H., Luo, J., Yang, Z., Chen, S. and Chen, J. (2018). The InR/Akt/TORC1 Growth-Promoting Signaling Negatively Regulates JAK/STAT Activity and Migratory Cell Fate during Morphogenesis. Dev Cell 44(4): 524-531.e525. PubMed ID: 29456138

    Wen, D. T., Zheng, L., Yang, F., Li, H. Z. and Chen, J. (2018). Endurance exercise prevents high-fat-diet induced locomotor impairment, cardiac dysfunction, lifespan shortening, and dSir2 expression decline in aging Drosophila. Exp Gerontol. PubMed ID: 29355704

    Pan, C., Wang, W., Yuan, H., Yang, L., Chen, B., Li, D. and Chen, J. (2017). The immediate early protein WSV187 can influence viral replication via regulation of JAK/STAT pathway in Drosophila. Dev Comp Immunol 72: 89-96. PubMed ID: 28232015

    Wu, J., Wang, H., Guo, X. and Chen, J. (2016). Cofilin-mediated actin dynamics promotes actin bundle formation during Drosophila bristle development. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27385345

  • Xin Chen Department of Biology, Johns Hopkins University
    Ranjan, R., Snedeker, J., Wooten, M., Chu, C., Bracero, S., Mouton, T. and Chen, X. (2022). Differential condensation of sister chromatids acts with Cdc6 to ensure asynchronous S-phase entry in Drosophila male germline stem cell lineage. Dev Cell 57(9): 1102-1118.e1107. PubMed ID: 35483360

    Zhou, J., Dabiri, Y., Gama-Brambila, R. A., Ghafoory, S., Altinbay, M., Mehrabi, A., Golriz, M., Blagojevic, B., Reuter, S., Han, K., Seidel, A., Dikic, I., Wolfl, S. and Cheng, X. (2022). pVHL-mediated SMAD3 degradation suppresses TGF-β signaling. J Cell Biol 221(1). PubMed ID: 34860252

    Vidaurre, V. and Chen, X. (2021). Epigenetic regulation of Drosophila germline stem cell maintenance and differentiation. Dev Biol 473: 105-118. PubMed ID: 33610541

    Shi, Z., Lim, C., Tran, V., Cui, K., Zhao, K. and Chen, X. (2020). Single-cyst transcriptome analysis of Drosophila male germline stem cell lineage. Development. PubMed ID: 32122991

    Ranjan, R., Snedeker, J. and Chen, X. (2019). Asymmetric centromeres differentially coordinate with mitotic machinery to ensure biased sister chromatid segregation in germline stem cells. Cell Stem Cell. PubMed ID: 31564548

    Wooten, M., Snedeker, J., Nizami, Z. F., Yang, X., Ranjan, R., Urban, E., Kim, J. M., Gall, J., Xiao, J. and Chen, X. (2019). Asymmetric histone inheritance via strand-specific incorporation and biased replication fork movement. Nat Struct Mol Biol. PubMed ID: 31358945

    Feng, L., Shi, Z., Xie, J., Ma, B. and Chen, X. (2018). Enhancer of polycomb maintains germline activity and genome integrity in Drosophila testis. Cell Death Differ. PubMed ID: 29362481

    Eun, S.H., Feng, L., Cedeno-Rosario, L., Gan, Q., Wei, G., Cui, K., Zhao, K. and Chen, X. (2017). Polycomb group gene E(z) is required for spermatogonial dedifferentiation in Drosophila adult testis. J Mol Biol [Epub ahead of print] . PubMed ID: 28434938

    Feng, L., Shi, Z. and Chen, X. (2017). Enhancer of polycomb coordinates multiple signaling pathways to promote both cyst and germline stem cell differentiation in the Drosophila adult testis. PLoS Genet 13: e1006571. PubMed ID: 28196077

    Xie, J., Wooten, M., Tran, V., Chen, B.C., Pozmanter, C., Simbolon, C., Betzig, E. and Chen, X. (2015). Histone H3 threonine phosphorylation regulates asymmetric histone inheritance in the Drosophila male germline. Cell [Epub ahead of print]. PubMed ID: 26522592

    Eun, S. H., Shi, Z., Cui, K., Zhao, K. and Chen, X. (2014). A Non-Cell Autonomous Role of E(z) to Prevent Germ Cells from Turning on a Somatic Cell Marker. Science 343: 1513-1516. PubMed ID: 24675960

  • Jun Cheng Department of Bioengineering, University of Illinois, Chicago
    Wang, Z., Oppegard, S. C., Eddington, D. T. and Cheng, J. (2017). Effect of localized hypoxia on Drosophila embryo development. PLoS One 12(9): e0185267. PubMed ID: 28934338

    Bang, C. and Cheng, J. (2015). Dynamic interplay of spectrosome and centrosome organelles in asymmetric stem cell divisions. PLoS One 10: e0123294. PubMed ID: 25849996

    Zhang, B., Xu, Z., Zhang, Y., Shao, X., Xu, X., Cheng, J. and Li, Z. (2015). Fipronil induces apoptosis through caspase-dependent mitochondrial pathways in Drosophila S2 cells. Pestic Biochem Physiol 119: 81-89. PubMed ID: 25868821

    Salzmann, V., Inaba, M., Cheng, J. and Yamashita, Y. M. (2013). Lineage tracing quantification reveals symmetric stem cell division in Drosophila male germline stem cells. Cell Mol Bioeng 6: 441-448. PubMed ID: 24465278

    Sun, L., Johnson, A. F., Li, J., Lambdin, A. S., Cheng, J. and Birchler, J. A. (2013). Differential effect of aneuploidy on the X chromosome and genes with sex-biased expression in Drosophila. Proc Natl Acad Sci U S A 110: 16514-16519. PubMed ID: 24062456

  • Louise Cheng Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
    Veen, K., Nguyen, P. K., Froldi, F., Dong, Q., Alvarez-Ochoa, E., Harvey, K. F., McMullen, J. P., Marshall, O., Jusuf, P. R. and Cheng, L. Y. (2023). Dedifferentiation-derived neural stem cells exhibit perturbed temporal progression. EMBO Rep: e55837. PubMed ID: 37039033

    Lodge, W., Zavortink, M., Golenkina, S., Froldi, F., Dark, C., Cheung, S., Parker, B. L., Blazev, R., Bakopoulos, D., Christie, E. L., Wimmer, V. C., Duckworth, B. C., Richardson, H. E. and Cheng, L. Y. (2021). Tumor-derived MMPs regulate cachexia in a Drosophila cancer model. Dev Cell 56(18): 2664-2680 e2666. PubMed ID: 34473940

    Dong, Q., Zavortink, M., Froldi, F., Golenkina, S., Lam, T. and Cheng, L. Y. (2021). Glial Hedgehog signalling and lipid metabolism regulate neural stem cell proliferation in Drosophila. EMBO Rep 22(5): e52130. PubMed ID: 33751817

  • Steve Chenoweth University of Queensland, Brisbane
    Rusuwa, B. B., Chung, H., Allen, S. L., Frentiu, F. D. and Chenoweth, S. F. (2022). Natural variation at a single gene generates sexual antagonism across fitness components in Drosophila. Curr Biol. PubMed ID: 35700732

    Narayan, V. P., Wilson, A. J. and Chenoweth, S. F. (2022). Genetic and social contributions to sex differences in lifespan in Drosophila serrata. J Evol Biol 35(4): 657-663. PubMed ID: 35290690

    Reddiex, A. J. and Chenoweth, S. F. (2021). Integrating genomics and multivariate evolutionary quantitative genetics: a case study of constraints on sexual selection in Drosophila serrata. Proc Biol Sci 288(1960): 20211785. PubMed ID: 34641732

    Gosden, T. P., Reddiex, A. J. and Chenoweth, S. F. (2018). Artificial selection reveals sex differences in the genetic basis of sexual attractiveness. Proc Natl Acad Sci U S A 115(21): 5498-5503. PubMed ID: 29735676

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

    Allen, S. L., McGuigan, K., Connallon, T., Blows, M. W. and Chenoweth, S. F. (2017). Sexual selection on spontaneous mutations strengthens the between-sex genetic correlation for fitness. Evolution [Epub ahead of print]. PubMed ID: 28722119

    Allen, S. L., Bonduriansky, R., Sgro, C. M. and Chenoweth, S. F. (2017). Sex-biased transcriptome divergence along a latitudinal gradient. Mol Ecol. PubMed ID: 28100025

    Gosden, T. P., Thomson, J. R., Blows, M. W., Schaul, A. and Chenoweth, S. F. (2016). Testing for a genetic response to sexual selection in a wild Drosophila population. J Evol Biol. PubMed ID: 26914275

    Latimer, C. A., Foley, B. and Chenoweth, S. F. (2014). Connecting thermal performance curve variation to the genotype: a multivariate QTL approach. J Evol Biol. PubMed ID: 25403928

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

    Allen, S. L., Bonduriansky, R. and Chenoweth, S. F. (2013). The genomic distribution of sex-biased genes in Drosophila serrata: X chromosome demasculinization, feminization, and hyper-expression in both sexes. Genome Biol Evol. PubMed ID: 24084777

  • Peter Cherbas Department of Biology, Indiana University
    Hackney, J. F. and Cherbas, P. (2015). Injury response checkpoint and developmental timing in insects. Fly (Austin) [Epub ahead of print]. PubMed ID: 25833067

    Hackney, J. F., Zolali-Meybodi, O. and Cherbas, P. (2012). Tissue damage disrupts developmental progression and ecdysteroid biosynthesis in Drosophila. PLoS One 7: e49105. PubMed ID: 23166607

    Gauthier, S. A., VanHaaften, E., Cherbas, L., Cherbas, P. and Hewes, R. S. (2012). Cryptocephal, the Drosophila melanogaster ATF4, is a specific coactivator for ecdysone receptor isoform B2. PLoS Genet 8: e1002883. PubMed ID: 22912598

  • Sara Cherry Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
    Liu, Y., Gordesky-Gold, B., Leney-Greene, M., Weinbren, N. L., Tudor, M. and Cherry, S. (2018). Inflammation-induced, STING-dependent autophagy restricts Zika virus infection in the Drosophila brain. Cell Host Microbe. PubMed ID: 29934091

    Liang, D., Tatomer, D. C., Luo, Z., Wu, H., Yang, L., Chen, L. L., Cherry, S. and Wilusz, J. E. (2017). The Output of Protein-Coding Genes Shifts to Circular RNAs When the Pre-mRNA Processing Machinery Is Limiting. Mol Cell. PubMed ID: 29174924

    Sansone, C. L., Cohen, J., Yasunaga, A., Xu, J., Osborn, G., Subramanian, H., Gold, B., Buchon, N. and Cherry, S. (2015). Microbiota-dependent priming of antiviral intestinal immunity in Drosophila. Cell Host Microbe 18: 571-581. PubMed ID: 26567510

    Kramer, M. C., Liang, D., Tatomer, D. C., Gold, B., March, Z. M., Cherry, S. and Wilusz, J. E. (2015). Combinatorial control of Drosophila circular RNA expression by intronic repeats, hnRNPs, and SR proteins. Genes Dev. PubMed ID: 26450910

    Hackett, B. A., Yasunaga, A., Panda, D., Tartell, M. A., Hopkins, K. C., Hensley, S. E. and Cherry, S. (2015). RNASEK is required for internalization of diverse acid-dependent viruses. Proc Natl Acad Sci U S A. PubMed ID: 26056282

    Panda, D., Gold, B., Tartell, M. A., Rausch, K., Casas-Tinto, S. and Cherry, S. (2015). The transcription factor FoxK participates with Nup98 to regulate antiviral gene expression. MBio 6. PubMed ID: 25852164

    Buchon, N., Silverman, N. and Cherry, S. (2014). Immunity in Drosophila melanogaster - from microbial recognition to whole-organism physiology. Nat Rev Immunol 14: 796-810. PubMed ID: 25421701

    Panda, D., Pascual-Garcia, P., Dunagin, M., Tudor, M., Hopkins, K. C., Xu, J., Gold, B., Raj, A., Capelson, M., Cherry, S. (2014). Nup98 promotes antiviral gene expression to restrict RNA viral infection in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 25197089

    Moy, R. H., Cole, B. S., Yasunaga, A., Gold, B., Shankarling, G., Varble, A., Molleston, J. M., tenOever, B. R., Lynch, K. W. and Cherry, S. (2014). Stem-Loop Recognition by DDX17 Facilitates miRNA Processing and Antiviral Defense. Cell 158: 764-777. PubMed ID: 25126784

    Panda, D., Rose, P. P., Hanna, S. L., Gold, B., Hopkins, K. C., Lyde, R. B., Marks, M. S. and Cherry, S. (2013). Genome-wide RNAi Screen Identifies SEC61A and VCP as Conserved Regulators of Sindbis Virus Entry. Cell Rep. PubMed ID: 24332855

  • Igor Chesnokov, Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham
    Akhmetova, K., Balasov, M. and Chesnokov, I. (2021). Drosophila STING protein has a role in lipid metabolism. Elife 10. PubMed ID: 34467853

    Balasov, M., Akhmetova, K. and Chesnokov, I. (2020). Humanized Drosophila Model of the Meier-Gorlin Syndrome Reveals Conserved and Divergent Features of the Orc6 Protein. Genetics 216(4): 995-1007. PubMed ID: 33037049

    Akhmetova, K., Balasov, M., Svitin, A., Chesnokova, E., Renfrow, M. and Chesnokov, I. (2018). Phosphorylation of Pnut in the Early Stages of Drosophila Embryo Development Affects Association of the Septin Complex with the Membrane and Is Important for Viability. G3 (Bethesda) 8(1): 27-38. PubMed ID: 29079679

    Akhmetova, K., Balasov, M., Huijbregts, R. P. and Chesnokov, I. (2015). Functional insight into the role of Orc6 in septin complex filament formation in Drosophila. Mol Biol Cell 26(1): 15-28. PubMed ID: 25355953

  • Darya Chetverina Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
    Erokhin, M., Mogila, V., Lomaev, D. and Chetverina, D. (2023). Polycomb Recruiters Inside and Outside of the Repressed Domains. Int J Mol Sci 24(14). PubMed ID: 37511153

    Chetverina, D. A., P. V. Elizar'ev, P. G. Georgiev and M. M. Erokhin (2015). SV40 transcription terminators stabilize the activity of regulatory elements in Drosophila melanogaster. Dokl Biochem Biophys 463: 251-254. PubMed ID: 26335824

    Erokhin, M., A. Davydova, A. Parshikov, V. M. Studitsky, P. Georgiev and D. Chetverina (2013). Transcription through enhancers suppresses their activity in Drosophila. Epigenetics Chromatin 6(1): 31. PubMed ID: 24279291

  • Ann-Shyn Chiang Institute of Biotechnology, Taiwan
    Chen, C. C., Lin, H. W., Feng, K. L., Tseng, D. W., de Belle, J. S. and Chiang, A. S. (2023). A subset of cholinergic mushroom body neurons blocks long-term memory formation in Drosophila. Cell Rep 42(8): 112974. PubMed ID: 37590142

    Lin, H. W., Chen, C. C., Jhang, R. Y., Chen, L., de Belle, J. S., Tully, T. and Chiang, A. S. (2022). CREBB repression of protein synthesis in mushroom body gates long-term memory formation in Drosophila. Proc Natl Acad Sci U S A 119(50): e2211308119. PubMed ID: 36469774

    Feng, K. L., Weng, J. Y., Chen, C. C., Abubaker, M. B., Lin, H. W., Charng, C. C., Lo, C. C., de Belle, J. S., Tully, T., Lien, C. C. and Chiang, A. S. (2021). Neuropeptide F inhibits dopamine neuron interference of long-term memory consolidation in Drosophila. iScience 24(12): 103506. PubMed ID: 34934925

    Lin, H. W., Chen, C. C., de Belle, J. S., Tully, T. and Chiang, A. S. (2021). CREBA and CREBB in two identified neurons gate long-term memory formation in Drosophila. Proc Natl Acad Sci U S A 118(37). PubMed ID: 34507985

    Tai, C. Y., Chin, A. L. and Chiang, A. S. (2020). A Comprehensive Map of Visual Projection Neurons for Processing Ultraviolet Information in the Drosophila Brain. J Comp Neurol. PubMed ID: 33174208

    Wu, J. K., Tai, C. Y., Feng, K. L., Chen, S. L., Chen, C. C. and Chiang, A. S. (2017). Long-term memory requires sequential protein synthesis in three subsets of mushroom body output neurons in Drosophila. Sci Rep 7(1): 7112. PubMed ID: 28769066

    Hsiao, P. Y., Tsai, C. L., Chen, M. C., Lin, Y. Y., Yang, S. D. and Chiang, A. S. (2015). Non-invasive manipulation of Drosophila behavior by two-photon excited red-activatable channelrhodopsin. Biomed Opt Express 6: 4344-4352. PubMed ID: 26601000

    Lin, C. W., Lin, H. W., Chiu, M. T., Shih, Y. H., Wang, T. Y., Chang, H. M. and Chiang, A. S. (2015). Automated in situ brain imaging for mapping the Drosophila connectome. J Neurogenet: 1-31. PubMed ID: 26223305

    Shih, H.W., Wu, C.L., Chang, S.W., Liu, T.H., Sih-Yu Lai, J., Fu, T.F., Fu, C.C. and Chiang, A.S. (2015). Parallel circuits control temperature preference in Drosophila during ageing. Nat Commun 6: 7775. PubMed ID: 26178754

    Shih, C.T., Sporns, O., Yuan, S.L., Su, T.S., Lin, Y.J., Chuang, C.C., Wang, T.Y., Lo, C.C., Greenspan, R.J. and Chiang, A.S. (2015). Connectomics-based analysis of information flow in the Drosophila brain. Curr Biol 25: 1249-1258. PubMed ID: 25866397

    Shih, C. T., Sporns, O., Yuan, S. L., Su, T. S., Lin, Y. J., Chuang, C. C., Wang, T. Y., Lo, C. C., Greenspan, R. J. and Chiang, A. S. (2015). Connectomics-based analysis of information flow in the Drosophila brain. Curr Biol. PubMed ID: 25866397

  • M. Eugenia Chiappe
    Erginkaya, M. and Chiappe, M. E. (2022). Motion vision: Drosophila neural pathways that go with the visual flow. Curr Biol 32(16): R881-R883. PubMed ID: 35998597

    Fujiwara, T., Brotas, M. and Chiappe, M. E. (2022). Walking strides direct rapid and flexible recruitment of visual circuits for course control in Drosophila. Neuron 110(13): 2124-2138 e2128. PubMed ID: 35525243

    Cruz, T. L., Perez, S. M. and Chiappe, M. E. (2021). Fast tuning of posture control by visual feedback underlies gaze stabilization in walking Drosophila. Curr Biol 31(20): 4596-4607 e4595. PubMed ID: 34499851

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

    Sharifai, N., Samarajeewa, H., Kamiyama, D., Deng, T. C., Boulina, M. and Chiba, A. (2014). Imaging dynamic molecular signaling by the Cdc42 GTPase within the developing CNS. PLoS One 9: e88870. PubMed ID: 24586421

    Chang, X., Kim, M. D., Stephens, R., Qu, T., Chiba, A. and Tsechpenakis, G. (2013). Part-based motor neuron recognition in the Drosophila ventral nerve cord. Neuroimage. PubMed ID: 24373882

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

    Tsechpenakis, G., Mukherjee, P., Kim, M. D. and Chiba, A. (2012). Three-dimensional motor neuron morphology estimation in the Drosophila ventral nerve cord. IEEE Trans Biomed Eng 59: 1253-1263. PubMed ID: 22203698

    Tsechpenakis, G., Gamage, R. E., Kim, M. D. and Chiba, A. (2011). Motor neuron morphology estimation for its classification in the Drosophila brain. Conf Proc IEEE Eng Med Biol Soc 2011: 7755-7758. PubMed ID: 22256136

  • Cheng-Ting Chien Institute of Molecular Biology, Taiwan
    Tsai, Y. W., Sung, H. H., Li, J. C., Yeh, C. Y., Chen, P. Y., Cheng, Y. J., Chen, C. H., Tsai, Y. C. and Chien, C. T. (2019). Glia-derived exosomal miR-274 targets Sprouty in trachea and synaptic boutons to modulate growth and responses to hypoxia. Proc Natl Acad Sci U S A. PubMed ID: 31666321

    Chen, P. Y., Tsai, Y. W., Cheng, Y. J., Giangrande, A. and Chien, C. T. (2019). Glial response to hypoxia in mutants of NPAS1/3 homolog Trachealess through Wg signaling to modulate synaptic bouton organization. PLoS Genet 15(8): e1007980. PubMed ID: 31381576

    Yang, W. K., Chueh, Y. R., Cheng, Y. J., Siegenthaler, D., Pielage, J. and Chien, C. T. (2019). Epidermis-derived L1CAM homolog Neuroglian mediates dendrite enclosure and blocks heteroneuronal dendrite bundling. Curr Biol. PubMed ID: 31006568

    Wang, C. H., Huang, Y. C., Chen, P. Y., Cheng, Y. J., Kao, H. H., Pi, H. and Chien, C. T. (2017). USP5/Leon deubiquitinase confines postsynaptic growth by maintaining ubiquitin homeostasis through Ubiquilin. Elife 6. PubMed ID: 28489002

    Wang, M., Chen, P. Y., Wang, C. H., Lai, T. T., Tsai, P. I., Cheng, Y. J., Kao, H. H. and Chien, C. T. (2016). Dbo/Henji Modulates Synaptic dPAK to Gate Glutamate Receptor Abundance and Postsynaptic Response. PLoS Genet 12: e1006362. PubMed ID: 27736876

    Lin, C. H., Li, H., Lee, Y. N., Cheng, Y. J., Wu, R. M. and Chien, C. T. (2015). Lrrk regulates the dynamic profile of dendritic Golgi outposts through the golgin Lava lamp. J Cell Biol 210: 471-483. PubMed ID: 26216903

    Wang, C. H., Chen, G. C. and Chien, C. T. (2014). The deubiquitinase Leon/USP5 regulates ubiquitin homeostasis during Drosophila development. Biochem Biophys Res Commun. PubMed ID: 25152394

    Tsai, P. I., Wang, M., Kao, H. H., Cheng, Y. J., Walker, J. A., Chen, R. H. and Chien, C. T. (2012). Neurofibromin mediates FAK signaling in confining synapse growth at Drosophila neuromuscular junctions. J Neurosci 32: 16971-16981. PubMed ID: 23175848

    Tsai, P. I., Wang, M., Kao, H. H., Cheng, Y. J., Lin, Y. J., Chen, R. H. and Chien, C. T. (2012). Activity-dependent retrograde laminin A signaling regulates synapse growth at Drosophila neuromuscular junctions. Proc Natl Acad Sci U S A 109: 17699-17704. PubMed ID: 23054837

    Chen, C. K., Chen, W. Y. and Chien, C. T. (2012). The POU-domain protein Pdm3 regulates axonal targeting of R neurons in the Drosophila ellipsoid body. Dev Neurobiol 72: 1422-1432. PubMed ID: 22190420

  • Takahiro Chihara The University of Tokyo
    Kamemura, K., Moriya, H., Ukita, Y., Okumura, M., Miura, M. and Chihara, T. (2022). Endoplasmic reticulum proteins Meigo and Gp93 govern dendrite targeting by regulating Toll-6 localization. Dev Biol 484: 30-39. PubMed ID: 35134382

    Takeuchi, K. I., Honda, D., Okumura, M., Miura, M. and Chihara, T. (2021). Systemic innate immune response induces death of olfactory receptor neurons in Drosophila. Genes Cells. PubMed ID: 34921694

    Kamemura, K., Chen, C. A., Okumura, M., Miura, M. and Chihara, T. (2021). Amyotrophic lateral sclerosis-associated VAP33 is required for maintaining neuronal dendrite morphology and organelle distribution in Drosophila. Genes Cells. PubMed ID: 33548103

    Anzo, M., Sekine, S., Makihara, S., Chao, K., Miura, M. and Chihara, T. (2017). Dendritic Eph organizes dendrodendritic segregation in discrete olfactory map formation in Drosophila. Genes Dev 31(10): 1054-1065. PubMed ID: 28637694

    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

    Kashio, S., Obata, F., Zhang, L., Katsuyama, T., Chihara, T. and Miura, M. (2016). Tissue nonautonomous effects of fat body methionine metabolism on imaginal disc repair in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 26831070

    Sakuma, C., Okumura, M., Umehara, T., Miura, M. and Chihara, T. (2015). A STRIPAK component Strip regulates neuronal morphogenesis by affecting microtubule stability. Sci Rep 5: 17769. PubMed ID: 26644129

    Okumura, M., Kato, T., Miura, M. and Chihara, T. (2015). Hierarchical axon targeting of Drosophila olfactory receptor neurons specified by the proneural transcription factors Atonal and Amos. Genes Cells. PubMed ID: 26663477

    Sakuma, C., Kawauchi, T., Haraguchi, S., Shikanai, M., Yamaguchi, Y., Gelfand, V. I., Luo, L., Miura, M. and Chihara, T. (2014). Drosophila Strip serves as a platform for early endosome organization during axon elongation. Nat Commun 5: 5180. PubMed ID: 25312435

    Chihara, T., Kitabayashi, A., Morimoto, M., Takeuchi, K., Masuyama, K., Tonoki, A., Davis, R. L., Wang, J. W. and Miura, M. (2014). Caspase inhibition in select olfactory neurons restores innate attraction behavior in aged Drosophila. PLoS Genet 10: e1004437. PubMed ID: 24967585

  • Irene Chiolo Department of Biological Sciences, University of Southern California
    Caridi, C. P., D'Agostino, C., Ryu, T., Zapotoczny, G., Delabaere, L., Li, X., Khodaverdian, V. Y., Amaral, N., Lin, E., Rau, A. R. and Chiolo, I. (2018). Nuclear F-actin and myosins drive relocalization of heterochromatic breaks. Nature. PubMed ID: 29925946

    Ryu, T., Bonner, M. and Chiolo, I. (2016). Cervantes and Quijote protect heterochromatin from aberrant recombination and lead the way to the nuclear periphery. Nucleus [Epub ahead of print] PubMed ID: 27673416

    Ryu, T., Spatola, B., Delabaere, L., Bowlin, K., Hopp, H., Kunitake, R., Karpen, G. H. and Chiolo, I. (2015). Heterochromatic breaks move to the nuclear periphery to continue recombinational repair. Nat. Cell Biol. 17(11): 1401-11. PubMed ID: 26502056

    Chiolo, I., Minoda, A., Colmenares, S. U., Polyzos, A., Costes, S. V., Karpen, G. H. (2011). Double-strand breaks in heterochromatin move outside of a dynamic HP1a domain to complete recombinational repair. Cell 144: 732-44.

  • Adam Chippindale Department of Biology, Queen's University Kingston, Ontario
    Robinson, C. E., Thyagarajan, H. and Chippindale, A. K. (2023). Evolution of reproductive isolation in a long-term evolution experiment with Drosophila melanogaster: 30 years of divergent life-history selection. Evolution 77(8): 1756-1768. PubMed ID: 37256776

    Xiao, C., Bayat Fard, N., Brzezinski, K., Robertson, R. M. and Chippindale, A. K. (2019). Experimental evolution of response to anoxia in Drosophila melanogaster: recovery of locomotion following CO2 or N2 exposure. J Exp Biol 222(Pt 14). PubMed ID: 31285245

    Pischedda, A. and Chippindale, A. K. (2017). Direct benefits of choosing a high fitness mate can offset the indirect costs associated with intralocus sexual conflict. Evolution [Epub ahead of print]. PubMed ID: 28369895

    Kimber, C. M. and Chippindale, A. K. (2013). Mutation, Condition, and the Maintenance of Extended Lifespan in Drosophila. Curr Biol. PubMed ID: 24210612

    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

    Chippindale, A. K. (2013). A cryptic rock-paper-scissors game between Drosophila males. Mol Ecol 22: 1190-1192. PubMed ID: 23437838

  • Joanna Chiu Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis
    Tabuloc, C. A., Cai, Y. D., Kwok, R. S., Chan, E. C., Hidalgo, S. and Chiu, J. C. (2023). CLOCK and TIMELESS regulate rhythmic occupancy of the BRAHMA chromatin-remodeling protein at clock gene promoters. PLoS Genet 19(2): e1010649. PubMed ID: 36809369

    Hidalgo, S., Anguiano, M., Tabuloc, C. A. and Chiu, J. C. (2023). Seasonal cues act through the circadian clock and pigment-dispersing factor to control EYES ABSENT and downstream physiological changes. Curr Biol. PubMed ID: 36708710

    Liu, X. and Chiu, J. C. (2022). Nutrient-sensitive protein O-GlcNAcylation shapes daily biological rhythms. Open Biol 12(9): 220215. PubMed ID: 36099933

    Abrieux, A., Xue, Y., Cai, Y., Lewald, K. M., Nguyen, H. N., Zhang, Y. and Chiu, J. C. (2020). EYES ABSENT and TIMELESS integrate photoperiodic and temperature cues to regulate seasonal physiology in Drosophila. Proc Natl Acad Sci U S A 117(26): 15293-15304. PubMed ID: 32541062

    Lam, V. H., Li, Y. H., Liu, X., Murphy, K. A., Diehl, J. S., Kwok, R. S. and Chiu, J. C. (2018). CK1alpha collaborates with DOUBLETIME to regulate PERIOD function in the Drosophila circadian clock. J Neurosci. PubMed ID: 30373768

    Kwok, R. S., Lam, V. H. and Chiu, J. C. (2016). Understanding the role of chromatin remodeling in the regulation of circadian transcription in Drosophila. Fly (Austin): [Epub ahead of print]. PubMed ID: 26926115

    Kwok, R. S., Li, Y. H., Lei, A. J., Edery, I. and Chiu, J. C. (2015). The catalytic and non-catalytic functions of the Brahma chromatin-remodeling protein collaborate to fine-tune circadian transcription in Drosophila. PLoS Genet 11: e1005307. PubMed ID: 26132408

    Hamby, K. A., Kwok, R. S., Zalom, F. G. and Chiu, J. C. (2013). Integrating Circadian Activity and Gene Expression Profiles to Predict Chronotoxicity of Drosophila suzukii Response to Insecticides. PLoS One 8: e68472. PubMed ID: 23861907

    Chiu, J. C., Low, K. H., Pike, D. H., Yildirim, E. and Edery, I. (2010). Assaying locomotor activity to study circadian rhythms and sleep parameters in Drosophila. J Vis Exp. PubMed ID: 20972399

  • Dmitri ‘Mitya’ Chklovskii Simons Foundation, New York
    Lipshutz, D., Kashalikar, A., Farashahi, S. and Chklovskii, D. B. (2023). A linear discriminant analysis model of imbalanced associative learning in the mushroom body compartment. PLoS Comput Biol 19(2): e1010864. PubMed ID: 36745688

    Chapochnikov, N. M., Pehlevan, C. and Chklovskii, D. B. (2023). Normative and mechanistic model of an adaptive circuit for efficient encoding and feature extraction. Proc Natl Acad Sci U S A 120(29): e2117484120. PubMed ID: 37428907

    Chapochnikov, N. M., Pehlevan, C. and Chklovskii, D. B. (2023). Normative and mechanistic model of an adaptive circuit for efficient encoding and feature extraction. Proc Natl Acad Sci U S A 120(29): e2117484120. PubMed ID: 37428907

    Lipshutz, D., Kashalikar, A., Farashahi, S. and Chklovskii, D. B. (2023). A linear discriminant analysis model of imbalanced associative learning in the mushroom body compartment. PLoS Comput Biol 19(2): e1010864. PubMed ID: 36745688

    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. PubMed ID: 28432786

  • Kyoung Sang Cho Department of Biological Sciences, Konkuk University, Seoul, 05029, Republic of Korea; Korea Hemp Institute, Konkuk University, Seoul, 05029, Republic of Korea
    Lee, B., Choi, B., Park, Y., Jang, S., Yuan, C., Lim, C., Lee, J. H., Song, G. J. and Cho, K. S. (2022). Roles of ZnT86D in Neurodevelopment and Pathogenesis of Alzheimer Disease in a Drosophila melanogaster Model. Int J Mol Sci 23(19). PubMed ID: 36233134

    Choi, B., Lim, C., Lee, H., Lee, J. E., Kim, J., Chung, C. and Cho, K. S. (2022). Neuroprotective effects of linear ubiquitin E3 ligase against aging-induced DNA damage and amyloid β neurotoxicity in the brain of Drosophila melanogaster. Biochem Biophys Res Commun 637: 196-202. PubMed ID: 36403483

    Lee, B., Shin, C., Shin, M., Choi, B., Yuan, C. and Cho, K. S. (2021). The linear ubiquitin E3 ligase-Relish pathway is involved in the regulation of proteostasis in Drosophila muscle during aging. Biochem Biophys Res Commun 550: 184-190. PubMed ID: 33706102

    Kim, T., Shin, H., Song, B., Won, C., Yoshida, H., Yamaguchi, M., Cho, K. S. and Lee, I. S. (2020). Overexpression of H3K36 methyltransferase NSD in glial cells affects brain development in Drosophila. Glia 68(12): 2503-2516. PubMed ID: 32531091

    Jeon, Y., Lee, J. H., Choi, B., Won, S. Y. and Cho, K. S. (2020). Genetic Dissection of Alzheimer's Disease Using Drosophila Models. Int J Mol Sci 21(3). PubMed ID: 32019113

    Hwang, S., Jeong, H., Hong, E. H., Joo, H. M., Cho, K. S. and Nam, S. Y. (2019). Low-dose ionizing radiation alleviates Abeta42-induced cell death via regulating AKT and p38 pathways in Drosophila Alzheimer's disease models. Biol Open 8(2). PubMed ID: 30670376


  • Kwang-Min Choe Yonsei University, Department of Systems Biology
    Park, S. H., Lee, C. W. and Choe, K. M. (2019). Interplay between integrins and PI4P5K Sktl is crucial for cell polarization and reepithelialisation during Drosophila wound healing. Sci Rep 9(1): 16331. PubMed ID: 31704968

    Lee, C. W., Kwon, Y. C., Lee, Y., Park, M. Y. and Choe, K. M. (2019). cdc37 is essential for JNK pathway activation and wound closure in Drosophila. Mol Biol Cell: mbcE18120822. PubMed ID: 31483695

    Ri, H., Lee, J., Sonn, J. Y., Yoo, E., Lim, C. and Choe, J. (2019). Drosophila CrebB is a substrate of the nonsense-mediated mRNA decay pathway that sustains circadian behaviors. Mol Cells 42(4): 301-312. PubMed ID: 31091556

    Park, S. H., Lee, C. W., Lee, J. H., Park, J. Y., Roshandell, M., Brennan, C. A. and Choe, K. M. (2018). Requirement for and polarized localization of integrin proteins during Drosophila wound closure. Mol Biol Cell: mbcE17110635. PubMed ID: 29995573

    Lee, J. H., Lee, C. W., Park, S. H. and Choe, K. M. (2017). Spatiotemporal regulation of cell fusion by JNK and JAK/STAT signaling during Drosophila wound healing. J Cell Sci 130(11): 1917-1928. PubMed ID: 28424232

    Kim, M. J. and Choe, K. M. (2014). Basement membrane and cell integrity of self-tissues in maintaining Drosophila immunological tolerance. PLoS Genet 10(10): e1004683. PubMed ID: 25329560

  • Kwang-Wook Choi Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
    Mok, J. W. and Choi, K. W. (2021). Ex-vivo Microtubule Stability Assay Using Drosophila Wing Disc. Bio Protoc 11(23): e4241. PubMed ID: 35005086

    Yang, D. W., Mok, J. W., Telerman, S. B., Amson, R., Telerman, A. and Choi, K. W. (2021). Topoisomerase II is regulated by translationally controlled tumor protein for cell survival during organ growth in Drosophila. Cell Death Dis 12(9): 811. PubMed ID: 34453033

    Mok, J. W. and Choi, K. W. (2021). Novel function of N-acetyltransferase for microtubule stability and JNK signaling in Drosophila organ development. Proc Natl Acad Sci U S A 118(4). PubMed ID: 33479178

    Vuong, L. T., Won, J. H., Nguyen, M. B. and Choi, K. W. (2020). Role of Armadillo repeat 2 and kinesin-II motor subunit Klp64D for wingless signaling in Drosophila. Sci Rep 10(1): 13864. PubMed ID: 32807823

    Yang, D. W. and Choi, K. W. (2020). Suppression of Patronin deficiency by altered Hippo signaling in Drosophila organ development. Cell Death Differ. PubMed ID: 32737445

    Son, W. and Choi, K. W. (2020). The Classic Lobe Eye Phenotype of Drosophila Is Caused by Transposon Insertion-Induced Misexpression of a Zinc-Finger Transcription Factor. Genetics. PubMed ID: 32641295

    Jung, J., Yeom, E. and Choi, K. W. (2020). Ciao1 interacts with Crumbs and Xpd to regulate organ growth in Drosophila. Cell Death Dis 11(5): 365. PubMed ID: 32404863

    Kim, L. H., Hong, S. T. and Choi, K. W. (2019). Protein phosphatase 2A interacts with Verthandi/Rad21 to regulate mitosis and organ development in Drosophila. Sci Rep 9(1): 7624. PubMed ID: 31110215

    Kim, A. R. and Choi, K. W. (2019). TRiC/CCT chaperonins are essential for organ growth by interacting with insulin/TOR signaling in Drosophila. Oncogene. PubMed ID: 30792539

    Kim, A. R., Choi, E. B., Kim, M. Y. and Choi, K. W. (2017). Angiotensin-converting enzyme Ance is cooperatively regulated by Mad and Pannier in Drosophila imaginal discs. Sci Rep 7(1): 13174. PubMed ID: 29030610

    Nguyen, M.B., Vuong, L.T. and Choi, K.W. (2016). Ebi modulates wing growth by ubiquitin-dependent downregulation of Crumbs in Drosophila. Development 143: 3506-3513. PubMed ID: 27702784

    Hong, S.T. and Choi, K.W. (2016). Antagonistic roles of Drosophila Tctp and Brahma in chromatin remodelling and stabilizing repeated sequences. Nat Commun 7: 12988. PubMed ID: 27687497

    Le, T.P., Vuong, L.T., Kim, A.R., Hsu, Y.C. and Choi, K.W. (2016). 14-3-3 proteins regulate Tctp-Rheb interaction for organ growth in Drosophila. Nat Commun 7: 11501. PubMed ID: 27151460

  • Clement Chow Department of Human Genetics at the University of Utah School of Medicine
    Owings, K. G. and Chow, C. Y. (2023). A Drosophila screen identifies a role for histone methylation in ER stress preconditioning. bioRxiv. PubMed ID: 36945590

    Hope, K. A., Berman, A. R., Peterson, R. T. and Chow, C. Y. (2022). An in vivo drug repurposing screen and transcriptional analyses reveals the serotonin pathway and GSK3 as major therapeutic targets for NGLY1 deficiency. PLoS Genet 18(6): e1010228. PubMed ID: 35653343

    Talsness, D. M., Owings, K. G., Coelho, E., Mercenne, G., Pleinis, J. M., Partha, R., Hope, K. A., Zuberi, A. R., Clark, N. L., Lutz, C. M., Rodan, A. R. and Chow, C. Y. (2020). A Drosophila screen identifies NKCC1 as a modifier of NGLY1 deficiency. Elife 9. PubMed ID: 33315011

    Palu, R. A. S., Dalton, H. M. and Chow, C. Y. (2020). Decoupling of Apoptosis from Activation of the ER Stress Response by the Drosophila Metallopeptidase superdeath. Genetics. PubMed ID: 32047096

    Palu, R. A. S., Ong, E., Stevens, K., Chung, S., Owings, K. G., Goodman, A. G. and Chow, C. Y. (2019). Natural Genetic Variation Screen in Drosophila Identifies Wnt Signaling, Mitochondrial Metabolism, and Redox Homeostasis Genes as Modifiers of Apoptosis. G3 (Bethesda). PubMed ID: 31570502

    Lavoy, S., Chittoor-Vinod, V. G., Chow, C. Y. and Martin, I. (2018). Genetic Modifiers of Neurodegeneration in a Drosophila Model of Parkinson's Disease. Genetics 209(4): 1345-1356. PubMed ID: 29907646

    Owings, K. G., Lowry, J. B., Bi, Y., Might, M. and Chow, C. Y. (2018). Transcriptome and functional analysis in a Drosophila model of NGLY1 deficiency provides insight into therapeutic approaches. Hum Mol Genet 27(6): 1055-1066. PubMed ID: 29346549

  • Stanislava Chtarbanova Department of Biological Sciences, College of Arts & Sciences, University of Alabama, Tuscaloosa, AL,
    Lu, M. Y. and Chtarbanova, S. (2022). The role of micro RNAs (miRNAs) in the regulation of Drosophila melanogaster's innate immunity. Fly (Austin) 16(1): 382-396. PubMed ID: 36412256

    Goto, A., Ji, S., Chtarbanova, S. and Kuraishi, T. (2023). Editorial: Inflammatory and inflammatory-like responses in insects. Front Immunol 14: 1184429. PubMed ID: 37143679

    >Tafesh-Edwards, G., Kalukin, A., Bunnell, D., Chtarbanova, S. and Eleftherianos, I. (2023). Temperature and sex shape Zika virus pathogenicity in the adult Brat (cheesehead) brain: A Drosophila model for virus-associated neurological diseases. iScience 26(4): 106424. PubMed ID: 37009222

    Hagedorn, E., Bunnell, D., Henschel, B., Smith, D. L., Dickinson, S., Brown, A. W., De Luca, M., Turner, A. N. and Chtarbanova, S. (2023). RNA virus-mediated changes in organismal oxygen consumption rate in young and old Drosophila melanogaster males. Aging (Albany NY) 15(6): 1748-1767. PubMed ID: 36947702

    Lu, M. Y. and Chtarbanova, S. (2022). The role of micro RNAs (miRNAs) in the regulation of Drosophila melanogaster's innate immunity. Fly (Austin) 16(1): 382-396. PubMed ID: 36412256

  • Debapratim Chowdhuri Embryotoxicology Section, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
    Vimal, D., Kumar, S., Pandey, A., Sharma, D., Saini, S., Gupta, S., Ravi Ram, K. and Chowdhuri, D. K. (2017). Mlh1 is required for female fertility in Drosophila melanogaster: An outcome of effects on meiotic crossing over, ovarian follicles and egg activation. Eur J Cell Biol. PubMed ID: 29290392

    Singh, P. and Chowdhuri, D. K. (2018). Modulation of sestrin confers protection to Cr(VI) induced neuronal cell death in Drosophila melanogaster. Chemosphere 191: 302-314. PubMed ID: 29045932

    Chandra, S., Khatoon, R., Pandey, A., Saini, S., Vimal, D., Singh, P. and Chowdhuri, D. K. (2015). dme-miR-314-3p modulation in Cr(VI) exposed Drosophila affects DNA damage repair by targeting mus309. J Hazard Mater 304: 360-369. PubMed ID: 26590872

    Shukla, A. K., Pragya, P., Chaouhan, H. S., Tiwari, A. K., Patel, D. K., Abdin, M. Z. and Chowdhuri, D. K. (2014). Heat Shock Protein-70 (Hsp-70) Suppresses Paraquat-Induced Neurodegeneration by Inhibiting JNK and Caspase-3 Activation in Drosophila Model of Parkinson's Disease. PLoS One 9: e98886. PubMed ID: 24887138

    Shukla, A. K., Pragya, P., Chaouhan, H. S., Tiwari, A. K., Patel, D. K., Abdin, M. Z. and Chowdhuri, D. K. (2014). Heat Shock Protein-70 (Hsp-70) Suppresses Paraquat-Induced Neurodegeneration by Inhibiting JNK and Caspase-3 Activation in Drosophila Model of Parkinson's Disease. PLoS One 9: e98886. PubMed ID: 24887138

    Sonane, M., Goyal, R., Chowdhuri, D. K., Ram, K. R. and Gupta, K. C. (2013). Enhanced efficiency of P-element mediated transgenesis in Drosophila: Microinjection of DNA complexed with nanomaterial. Sci Rep 3: 3408. PubMed ID: 24296454

    Mishra, M., Sharma, A., Shukla, A. K., Pragya, P., Murthy, R. C., de Pomerai, D., Dwivedi, U. N. and Chowdhuri, D. K. (2013). Transcriptomic analysis provides insights on hexavalent chromium induced DNA double strand breaks and their possible repair in midgut cells of Drosophila melanogaster larvae. Mutat Res 747-748: 28-39. PubMed ID: 23628323

  • Jongkyeong Chung Molecular Genomics Lab, Seoul National University, Seoul
    Ham, S. J., Yoo, H., Woo, D., Lee, D. H., Park, K. S. and Chung, J. (2023). PINK1 and Parkin regulate IP(3)R-mediated ER calcium release. Nat Commun 14(1): 5202. PubMed ID: 37626046

    Park, J., Jun, K., Choi, Y., Yoon, E., Kim, W., Jang, Y. G. and Chung, J. (2020). CORO7 functions as a scaffold protein for the core kinase complex assembly of the Hippo pathway. J Biol Chem 296: 100040. PubMed ID: 33410403

    Jang, Y. G., Choi, Y., Jun, K. and Chung, J. (2020). Mislocalization of TORC1 to Lysosomes Caused by KIF11 Inhibition Leads to Aberrant TORC1 Activity. Mol Cells 43(8): 705-717. PubMed ID: 32759469

    Hwang, S. H., Bang, S., Kim, W. and Chung, J. (2020). Von Hippel-Lindau tumor suppressor (VHL) stimulates TOR signaling by interacting with phosphoinositide 3-kinase (PI3K). J Biol Chem. PubMed ID: 31959630

    Ham, S. J., Lee, D., Yoo, H., Jun, K., Shin, H. and Chung, J. (2020). Decision between mitophagy and apoptosis by Parkin via VDAC1 ubiquitination. Proc Natl Acad Sci U S A. PubMed ID: 32047033

    Hwang, S. H., Bang, S., Kang, K. S., Kang, D. and Chung, J. (2018). ULK1 negatively regulates Wnt signaling by phosphorylating Dishevelled. Biochem Biophys Res Commun. PubMed ID: 30497781

    Min, S., Yoon, W., Cho, H. and Chung, J. (2017). Misato underlies visceral myopathy in Drosophila. Sci Rep 7(1): 17700. PubMed ID: 29255146

    Choi, S., Quan, X., Bang, S., Yoo, H., Kim, J., Park, J., Park, K. S. and Chung, J. (2017). Mitochondrial calcium uniporter in Drosophila transfers calcium between the endoplasmic reticulum and mitochondria in oxidative stress-induced cell death. J Biol Chem. PubMed ID: 28726639

    Min, S. and Chung, J. (2016). Identification of a neural pathway governing satiety in Drosophila. BMB Rep [Epub ahead of print]. PubMed ID: 26949022

    Min, S., Chae, H.S., Jang, Y.H., Choi, S., Lee, S., Jeong, Y.T., Jones, W.D., Moon, S.J., Kim, Y.J. and Chung, J. (2016). Identification of a peptidergic pathway critical to satiety responses in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26948873

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

    Patlar, B., Jayaswal, V., Ranz, J. M. and Civetta, A. (2021). Nonadaptive molecular evolution of seminal fluid proteins in Drosophila. Evolution. PubMed ID: 34184267

    Grewal, G., Patlar, B. and Civetta, A. (2021). Expression of Mst89B and CG31287 is Needed for Effective Sperm Storage and Egg Fertilization in Drosophila. Cells 10(2). PubMed ID: 33535499

    Go, A. C. and Civetta, A. (2020). Hybrid Incompatibilities and Transgressive Gene Expression Between Two Closely Related Subspecies of Drosophila. Front Genet 11: 599292. PubMed ID: 33362859

    Patlar, B. and Civetta, A. (2020). Speciation and changes in male-gene expression in Drosophila. Genome. PubMed ID: 32730707

    Ferguson, J., Gomes, S., Civetta, A. (2013) Rapid male-specific regulatory divergence and down regulation of spermatogenesis genes in Drosophila species hybrids. PLoS One 8: e61575. PubMed ID: 23593487

    Minuk, L. and Civetta, A. (2011). Drosophila genomes and the development of affordable molecular markers for species genotyping. Genome 54: 341-347. PubMed ID: 21491976

    Sundararajan, V. and Civetta, A. (2011). Male sex interspecies divergence and down regulation of expression of spermatogenesis genes in Drosophila sterile hybrids. J Mol Evol 72: 80-89. PubMed ID: 21079940

  • Chiara Cirelli Center for Sleep and Consciousness, Department of Psychiatry, University of Wisconsin - Madison School of Medicine
    Flores, C. C., Loschky, S. S., Marshall, W., Spano, G. M., Massaro Cenere, M., Tononi, G. and Cirelli, C. (2021). Identification of Ultrastructural Signatures of Sleep and Wake in the Fly Brain. Sleep. PubMed ID: 34536282

    Bushey, D., Tononi, G. and Cirelli, C. (2015). Sleep- and wake-dependent changes in neuronal activity and reactivity demonstrated in fly neurons using in vivo calcium imaging. Proc Natl Acad Sci U S A 112: 4785-4790. PubMed ID: 25825756

    Bushey, D., Tononi, G. and Cirelli, C. (2011). Sleep and synaptic homeostasis: structural evidence in Drosophila. Science 332: 1576-1581. PubMed ID: 21700878

    Gilestro, G. F., Tononi, G. and Cirelli, C. (2009). Widespread changes in synaptic markers as a function of sleep and wakefulness in Drosophila. Science 324: 109-112. PubMed ID: 19342593

  • Tom Clandinin Department of Neurobiology, Stanford University
    Vaughen, J. P., Theisen, E., Rivas-Serna, I. M., Berger, A. B., Kalakuntla, P., Anreiter, I., Mazurak, V. C., Rodriguez, T. P., Mast, J. D., Hartl, T., Perlstein, E. O., Reimer, R. J., Clandinin, M. T. and Clandinin, T. R. (2022). Glial control of sphingolipid levels sculpts diurnal remodeling in a circadian circuit. Neuron. PubMed ID: 35961319

    Turner, M. H., Krieger, A., Pang, M. M. and Clandinin, T. R. (2022). Visual and motor signatures of locomotion dynamically shape a population code for feature detection in Drosophila. Elife 11. PubMed ID: 36300621

    York, R. A., Brezovec, L. E., Coughlan, J., Herbst, S., Krieger, A., Lee, S. Y., Pratt, B., Smart, A. D., Song, E., Suvorov, A., Matute, D. R., Tuthill, J. C. and Clandinin, T. R. (2022). The evolutionary trajectory of drosophilid walking. Curr Biol. PubMed ID: 35671756

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

    Turner, M. H., Mann, K. and Clandinin, T. R. (2021). The connectome predicts resting-state functional connectivity across the Drosophila brain. Curr Biol. PubMed ID: 33770490

    Mann, K., Deny, S., Ganguli, S. and Clandinin, T. R. (2021). Coupling of activity, metabolism and behaviour across the Drosophila brain. Nature 593(7858): 244-248. PubMed ID: 33911283

    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

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

    Wienecke, C. F. R., Leong, J. C. S. and Clandinin, T. R. (2018). Linear summation underlies direction selectivity in Drosophila. Neuron 99(4): 680-688.e684. PubMed ID: 30057202

    Mann, K., Gallen, C. L. and Clandinin, T. R. (2017). Whole-brain calcium imaging reveals an intrinsic functional network in Drosophila. Curr Biol 27(15): 2389-2396.e2384. PubMed ID: 28756955

    Chamberland, S., Yang, H. H., Pan, M. M., Evans, S. W., Guan, S., Chavarha, M., Yang, Y., Salesse, C., Wu, H., Wu, J. C., Clandinin, T. R., Toth, K., Lin, M. Z. and St-Pierre, F. (2017). Fast two-photon imaging of subcellular voltage dynamics in neuronal tissue with genetically encoded indicators. Elife 6. PubMed ID: 28749338

    Fisher, Y. E., Yang, H. H., Isaacman-Beck, J., Xie, M., Gohl, D. M. and Clandinin, T. R. (2017). FlpStop, a tool for conditional gene control in Drosophila. Elife 6. PubMed ID: 28211790

    Leong, J. C., Esch, J. J., Poole, B., Ganguli, S. and Clandinin, T. R. (2016). Direction selectivity in Drosophila emerges from preferred-direction enhancement and null-direction suppression. J Neurosci 36: 8078-8092. PubMed ID: 27488629

    Yang, H. H., St-Pierre, F., Sun, X., Ding, X., Lin, M. Z. and Clandinin, T. R. (2016). Subcellular Imaging of Voltage and Calcium Signals Reveals Neural Processing In Vivo. Cell 166: 245-257. PubMed ID: 27264607

  • Adam Claridge-Chang IMCB/Duke-NUS Medical School, Singapore
    Tumkaya, T., Burhanudin, S., Khalilnezhad, A., Stewart, J., Choi, H. and Claridge-Chang, A. (2022). Most primary olfactory neurons have individually neutral effects on behavior. Elife 11. PubMed ID: 35044905

    Eriksson, A., Anand, P., Gorson, J., Grijuc, C., Hadelia, E., Stewart, J. C., Holford, M. and Claridge-Chang, A. (2018). Using Drosophila behavioral assays to characterize terebrid venom-peptide bioactivity. Sci Rep 8(1): 15276. PubMed ID: 30323294

    Tumkaya, T., Ott, S. and Claridge-Chang, A. (2018). A systematic review of Drosophila short-term-memory genetics: Meta-analysis reveals robust reproducibility. Neurosci Biobehav Rev 95: 361-382. PubMed ID: 30077573

    Selkrig, J., Mohammad, F., Ng, S. H., Chua, J. Y., Tumkaya, T., Ho, J., Chiang, Y. N., Rieger, D., Pettersson, S., Helfrich-Forster, C., Yew, J. Y. and Claridge-Chang, A. (2018). The Drosophila microbiome has a limited influence on sleep, activity, and courtship behaviors. Sci Rep 8(1): 10646. PubMed ID: 30006625

  • Andrew Clark Department of Molecular Biology and Genetics, Cornell University
    Srivastav, S., Feschotte, C. and Clark, A. G. (2023). Rapid evolution of piRNA clusters in the Drosophila melanogaster ovary. bioRxiv. PubMed ID: 37214865

    Hafezi, Y., Omurzakov, A., Carlisle, J. A., Caldas, I. V., Wolfner, M. F. and Clark, A. G. (2023). The Drosophila melanogaster Y-linked gene, WDY , is required for sperm to swim in the female reproductive tract. bioRxiv. PubMed ID: 36778485

    Delbare, S. Y. N., Jain, A. M., Clark, A. G. and Wolfner, M. F. (2023). Transcriptional programs are activated and microRNAs are repressed within minutes after mating in the Drosophila melanogaster female reproductive tract. BMC Genomics 24(1): 356. PubMed ID: 37370014
    BR<>Brown, N. C., Gordon, B., McDonough-Goldstein, C. E., Misra, S., Findlay, G. D., Clark, A. G. and Wolfner, M. F. (2023). The seminal odorant binding protein Obp56g is required for mating plug formation and male fertility in Drosophila melanogaster. bioRxiv. PubMed ID: 36798169

    Delbare, S. Y. N., Venkatraman, S., Scuderi, K., Wells, M. T., Wolfner, M. F., Basu, S. and Clark, A. G. (2023). Time series transcriptome analysis implicates the circadian clock in the Drosophila melanogaster female's response to sex peptide. Proc Natl Acad Sci U S A 120(5): e2214883120. PubMed ID: 36706221

    Flynn, J. M., Hu, K. B. and Clark, A. G. (2023). Three recent sex chromosome-to-autosome fusions in a Drosophila virilis strain with high satellite DNA content. Genetics. PubMed ID: 37052958

    Yang, E., Metzloff, M., Langmuller, A. M., Xu, X., Clark, A. G., Messer, P. W. and Champer, J. (2022). A homing suppression gene drive with multiplexed gRNAs maintains high drive conversion efficiency and avoids functional resistance alleles. G3 (Bethesda) 12(6). PubMed ID: 35394026

    Shahrestani, P., King, E., Ramezan, R., Phillips, M., Riddle, M., Thornburg, M., Greenspan, Z., Estrella, Y., Garcia, K., Chowdhury, P., Malarat, G., Zhu, M., Rottshaefer, S. M., Wraight, S., Griggs, M., Vandenberg, J., Long, A. D., Clark, A. G. and Lazzaro, B. P. (2021). The molecular architecture of Drosophila melanogaster defense against Beauveria bassiana explored through evolve and resequence and quantitative trait locus mapping. G3 (Bethesda). PubMed ID: 34534291

    Schlamp, F., Delbare, S. Y. N., Early, A. M., Wells, M. T., Basu, S. and Clark, A. G. (2021). Dense time-course gene expression profiling of the Drosophila melanogaster innate immune response. BMC Genomics 22(1): 304. PubMed ID: 33902461

    Clark, A. G., Wigby, S., Brown, N. C., Allen, S. E., Misra, S., Sitnik, J. L., Sepil, I., Clark, A. G. and Wolfner, M. F. (2020). The Drosophila seminal proteome and its role in postcopulatory sexual selectioCavalliCavalliCavalliCavalliCavallin. Philos Trans R Soc Lond B Biol Sci 375(1813): 20200072. PubMed ID: 33070726

    Delbare, S. Y. N., Ahmed-Braimah, Y. H., Wolfner, M. F. and Clark, A. G. (2020). Interactions between the microbiome and mating influence the female's transcriptional profile in Drosophila melanogaster. Sci Rep 10(1): 18168. PubMed ID: 33097776

  • Damon Clark Department of Molecular, Cellular, and Developmental Biology, Yale University
    Mano, O., Choi, M., Tanaka, R., Creamer, M. S., Matos, N. C. B., Shomar, J., Badwan, B. A.,

    Clandinin, T. R. and Clark, D. A. (2023). Long timescale anti-directional rotation in Drosophila optomotor behavior. bioRxiv. PubMed ID: 36711627 Tanaka, R., Zhou, B., Agrochao, M., Badwan, B. A., Au, B., Matos, N. C. B. and Clark, D. A. (2023). Drosophila integrates visual evidence and counterevidence in self motion estimation. bioRxiv. PubMed ID: 36711843

    Kadakia, N., Demir, M., Michaelis, B. T., DeAngelis, B. D., Reidenbach, M. A., Clark, D. A. and Emonet, T. (2022). Odour motion sensing enhances navigation of complex plumes. Nature. PubMed ID: 36352224

    Gonzalez-Suarez, A. D., Zavatone-Veth, J. A., Chen, J., Matulis, C. A., Badwan, B. A. and Clark, D. A. (2022). Excitatory and inhibitory neural dynamics jointly tune motion detection. Curr Biol 32(17): 3659-3675.e3658. PubMed ID: 35868321

    Tanaka, R. and Clark, D. A. (2022). Neural mechanisms to exploit positional geometry for collision avoidance. Curr Biol 32(11): 2357-2374.e2356. PubMed ID: 35508172

    Tanaka, R. and Clark, D. A. (2022). Identifying inputs to visual projection neurons in Drosophila lobula by analyzing connectomic data. eNeuro. PubMed ID: 35410869

    Zhou, B., Li, Z., Kim, S., Lafferty, J. and Clark, D. A. (2022). Shallow neural networks trained to detect collisions recover features of visual loom-selective neurons. Elife 11. PubMed ID: 35023828

    Mano, O., Creamer, M. S., Badwan, B. A. and Clark, D. A. (2021). Predicting individual neuron responses with anatomically constrained task optimization. Curr Biol. PubMed ID: 34324832

    Demir, M., Kadakia, N., Anderson, H. D., Clark, D. A. and Emonet, T. (2020). Walking Drosophila navigate complex plumes using stochastic decisions biased by the timing of odor encounters. Elife 9. PubMed ID: 33140723

    Tanaka, R. and Clark, D. A. (2020). Object-Displacement-Sensitive Visual Neurons Drive Freezing in Drosophila. Curr Biol. PubMed ID: 32442466

    DeAngelis, B. D., Zavatone-Veth, J. A., Gonzalez-Suarez, A. D. and Clark, D. A. (2020). Spatiotemporally precise optogenetic activation of sensory neurons in freely walking Drosophila. Elife 9. PubMed ID: 32319425

    Zavatone-Veth, J. A., Badwan, B. A. and Clark, D. A. (2020). A minimal synaptic model for direction selective neurons in Drosophila. J Vis 20(2): 2. PubMed ID: 32040161

  • Anne-Kathrin Classen Center for Biological Systems Analysis, Albert Ludwigs University of Freiburg
    Jaiswal, J., Egert, J., Engesser, R., Peyroton, A. A., Nogay, L., Weichselberger, V., Crucianelli, C., Grass, I., Kreutz, C., Timmer, J. and Classen, A. K. (2023). Mutual repression between JNK/AP-1 and JAK/STAT stratifies senescent and proliferative cell behaviors during tissue regeneration. PLoS Biol 21(5): e3001665. PubMed ID: 37252939

    Prasad, D., Illek, K., Fischer, F., Holstein, K. and Classen, A. K. (2023). Bilateral JNK activation is a hallmark of interface surveillance and promotes elimination of aberrant cells. Elife 12. PubMed ID: 36744859

    Crucianelli, C., Jaiswal, J., Vijayakumar Maya, A., Nogay, L., Cosolo, A., Grass, I. and Classen, A. K. (2022). Distinct signaling signatures drive compensatory proliferation via S-phase acceleration. PLoS Genet 18(12): e1010516. PubMed ID: 36520882

    Weichselberger, V., Dondl, P. and Classen, A. K. (2022). Eya-controlled affinity between cell lineages drives tissue self-organization during Drosophila oogenesis. Nat Commun 13(1): 6377. PubMed ID: 36289235

    Balaji, R., Weichselberger, V. and Classen, A. K. (2019). Response of Drosophila epithelial cell and tissue shape to external forces in vivo. Development 146(17). PubMed ID: 31399470

    Cosolo, A., Jaiswal, J., Csordas, G., Grass, I., Uhlirova, M. and Classen, A. K. (2019). JNK-dependent cell cycle stalling in G2 promotes survival and senescence-like phenotypes in tissue stress. Elife 8. PubMed ID: 30735120
    La Fortezza, M., Grigolon, G., Cosolo, A., Pindyurin, A., Breimann, L., Blum, H., van Steensel, B. and Classen, A. K. (2018). DamID profiling of dynamic Polycomb-binding sites in Drosophila imaginal disc development and tumorigenesis. Epigenetics Chromatin 11(1): 27. PubMed ID: 29871666

    Schunter, S., Villa, R., Flynn, V., Heidelberger, J. B., Classen, A. K., Beli, P. and Becker, P. B. (2017). Ubiquitylation of the acetyltransferase MOF in Drosophila melanogaster. PLoS One 12(5): e0177408. PubMed ID: 28510597

    Bunker, B. D., Nellimoottil, T. T., Boileau, R. M., Classen, A. K. and Bilder, D. (2015). The transcriptional response to tumorigenic polarity loss in Drosophila. Elife 4. PubMed ID: 25719210

  • Denise V. Clark University of New Brunswick, Fredericton
    O'Neill, R. S. and Clark, D. V. (2016). Partial functional diversification of Drosophila melanogaster septins Sep2 and Sep5. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27172205

    O'Neill, R. S. and Clark, D. V. (2013). Evolution of three parent genes and their retrogene copies in Drosophila species. Int J Evol Biol 2013: 693085. PubMed ID: 23841016

    Merzetti, E., Hackett, J. M., Clark, D. V. (2013) Transcriptional regulation of the purine de novo synthesis gene Prat in Drosophila melanogaster. Gene. PubMed ID: 23357222

    Holland, C., Lipsett, D. B. and Clark, D. V. (2011). A link between impaired purine nucleotide synthesis and apoptosis in Drosophila melanogaster. Genetics 188: 359-367. PubMed ID: 21441212

    Hackett, J. M. and Clark, D. V. (2009). Modifiers of Prat, a de novo purine synthesis gene, in Drosophila melanogaster. Genome 52: 957-967. PubMed ID: 19935919

  • Michael Cleary Health Sciences Research Institute, U. C. Merced, Merced, CA
    Aboukilila, M. Y., Sami, J. D., Wang, J., England, W., Spitale, R. C. and Cleary, M. D. (2020). Identification of novel regulators of dendrite arborization using cell type-specific RNA metabolic labeling. PLoS One 15(12): e0240386. PubMed ID: 33264304

    Burow, D. A., Martin, S., Quail, J. F., Alhusaini, N., Coller, J. and Cleary, M. D. (2018). Attenuated Codon Optimality Contributes to Neural-Specific mRNA Decay in Drosophila. Cell Rep 24(7): 1704-1712. PubMed ID: 30110627

    Hida, N., Aboukilila, M. Y., Burow, D. A., Paul, R., Greenberg, M. M., Fazio, M., Beasley, S., Spitale, R. C. and Cleary, M. D. (2017). EC-tagging allows cell type-specific RNA analysis. Nucleic Acids Res 45(15): e138. PubMed ID: 28641402

    Burow, D. A., Umeh-Garcia, M. C., True, M. B., Bakhaj, C. D., Ardell, D. H. and Cleary, M. D. (2015). Dynamic regulation of mRNA decay during neural development. Neural Dev 10: 11. PubMed ID: 25896902

  • Tom Cline University of California, Berkeley
    Evans, D. S. and Cline, T. W. (2013). Drosophila switch gene Sex-lethal can bypass its switch-gene target transformer to regulate aspects of female behavior. Proc Natl Acad Sci U S A. PubMed ID: 24191002

    Cline, T. W., Dorsett, M., Sun, S., Harrison, M. M., Dines, J., Sefton, L. and Megna, L. (2010). Evolution of the Drosophila feminizing switch gene Sex-lethal. Genetics 186: 1321-1336. PubMed ID: 20837995

    Harrison, M. M., Botchan, M. R. and Cline, T. W. (2010). Grainyhead and Zelda compete for binding to the promoters of the earliest-expressed Drosophila genes. Dev Biol 345: 248-255. PubMed ID: 20599892

  • Eleanor J. "Josie" Clowney Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor
    Ahmed, M., Rajagopalan, A. E., Pan, Y., Li, Y., Williams, D. L., Pedersen, E. A., Thakral, M., Previero, A., Close, K. C., Christoforou, C. P., Cai, D., Turner, G. C. and Clowney, E. J. (2023). Input density tunes Kenyon cell sensory responses in the Drosophila mushroom body. Curr Biol 33(13): 2742-2760 e2712. PubMed ID: 37348501

    Brovkina, M. V., Duffie, R., Burtis, A. E. C. and Clowney, E. J. (2021). Fruitless decommissions regulatory elements to implement cell-type-specific neuronal masculinization. PLoS Genet 17(2): e1009338. PubMed ID: 33600447

    Punal, V. M., Ahmed, M., Thornton-Kolbe, E. M. and Clowney, E. J. (2021). Untangling the wires: development of sparse, distributed connectivity in the mushroom body calyx. Cell Tissue Res 383(1): 91-112. PubMed ID: 33404837

    Ryba, A. R., McKenzie, S. K., Olivos-Cisneros, L., Clowney, E. J., Pires, P. M. and Kronauer, D. J. C. (2020). Comparative Development of the Ant Chemosensory System. Curr Biol 30(16): 3223-3230 e3224. PubMed ID: 32559450

  • Matthew Cobb Faculty of Life Sciences, University of Manchester
    Grillet, M., Campagner, D., Petersen, R., McCrohan, C. and Cobb, M. (2016). The peripheral olfactory code in Drosophila larvae contains temporal information and is robust over multiple timescales. Proc Biol Sci 283. PubMed ID: 27194701

    Hoare, D. J., Humble, J., Jin, D., Gilding, N., Petersen, R., Cobb, M. and McCrohan, C. (2011). Modeling peripheral olfactory coding in Drosophila larvae. PLoS One 6: e22996. PubMed ID: 21857978

    Everaerts, C., Farine, J. P., Cobb, M. and Ferveur, J. F. (2010). Drosophila cuticular hydrocarbons revisited: mating status alters cuticular profiles. PLoS One 5: e9607. PubMed ID: 20231905

  • Clark Coffman Department of Genetics, Development of Cell Biology, Iowa State University, Ames
    Pruitt, M. M., Lamm, M. H. and Coffman, C. R. (2013). Molecular dynamics simulations on the Tre1 G protein-coupled receptor: exploring the role of the arginine of the NRY motif in Tre1 structure. BMC Struct Biol 13: 15. PubMed ID: 24044607

    Kamps, A. R., Pruitt, M. M., Herriges, J. C. and Coffman, C. R. (2010). An evolutionarily conserved arginine is essential for Tre1 G protein-coupled receptor function during germ cell migration in Drosophila melanogaster. PLoS One 5: e11839. PubMed ID: 20676220

  • Stephen Cohen Department of Cellular and Molecular Medicine, University of Copenhagen
    Nagarkar, S., Wasnik, R., Govada, P., Cohen, S. and Shashidhara, L. S. (2020). Promoter Proximal Pausing Limits Tumorous Growth Induced by the Yki Transcription Factor in Drosophila. Genetics. PubMed ID: 32737120

    Groth, C., Vaid, P., Khatpe, A., Prashali, N., Ahiya, A., Andrejeva, D., Chakladar, M., Nagarkar, S., Paul, R., Eichenlaub, T., Herranz, H., Sridhar, T. S., Cohen, S. and Shashidhara, L. S. (2020). Genome Wide Screen for Context-Dependent Tumor Suppressors Identified Using in Vivo Models for Neoplasia in Drosophila. G3 (Bethesda). PubMed ID: 32737065

    Hanyu-Nakamura, K., Matsuda, K., Cohen, S. M. and Nakamura, A. (2019). Pgc suppresses the zygotically-acting RNA decay pathway to protect germ plasm RNAs in the Drosophila embryo. Development. PubMed ID: 30890569

    Kirsch, R., Seemann, S. E., Ruzzo, W. L., Cohen, S. M., Stadler, P. F. and Gorodkin, J. (2018). Identification and characterization of novel conserved RNA structures in Drosophila. BMC Genomics 19(1): 899. PubMed ID: 30537930

    Andrejeva, D., Kugler, J. M., Nguyen, H. T., Malmendal, A., Holm, M. L., Toft, B. G., Loya, A. C. and Cohen, S. M. (2018). Metabolic control of PPAR activity by aldehyde dehydrogenase regulates invasive cell behavior and predicts survival in hepatocellular and renal clear cell carcinoma. BMC Cancer 18(1): 1180. PubMed ID: 30486822

    Eichenlaub, T., Villadsen, R., Freitas, F. C. P., Andrejeva, D., Aldana, B. I., Nguyen, H. T., Petersen, O. W., Gorodkin, J., Herranz, H. and Cohen, S. M. (2018). Warburg effect metabolism drives neoplasia in a Drosophila genetic model of epithelial cancer. Curr Biol. PubMed ID: 30293715

    Aw, S. S., Lim, I. K. H., Tang, M. X. M. and Cohen, S. M. (2017). A glio-protective role of mir-263a by tuning sensitivity to Glutamate. Cell Rep 19(9): 1783-1793. PubMed ID: 28564598

    Foo, L. C., Song, S. and Cohen, S. M. (2017). miR-31 mutants reveal continuous glial homeostasis in the adult Drosophila brain. Embo J. PubMed ID: 28320737

    Herranz, H., Eichenlaub, T. and Cohen, S. M. (2016). Cancer in Drosophila: Imaginal Discs as a Model for Epithelial Tumor Formation. Curr Top Dev Biol 116: 181-199. PubMed ID: 26970620

    Eichenlaub, T., Cohen, S.M. and Herranz, H. (2016). Cell competition drives the formation of metastatic tumors in a Drosophila model of epithelial tumor formation. Curr Biol [Epub ahead of print]. PubMed ID: 26853367

    Weng, R. and Cohen, S. M. (2015). Control of Drosophila type I and type II central brain neuroblast proliferation by bantam microRNA. Development [Epub ahead of print]. PubMed ID: 26395494

    Verma, P. and Cohen, S. M. (2015). miR-965 controls cell proliferation and migration during tissue morphogenesis in the abdomen. Elife 4. PubMed ID: 26226636

  • Hervé Colinet Université de Rennes
    Morimoto, J., Wenzel, M., Derous, D., Henry, Y. and Colinet, H. (2022). The transcriptomic signature of responses to larval crowding in Drosophila melanogaster. Insect Sci. PubMed ID: 36115064

    Henry, Y. and Colinet, H. (2018). Microbiota disruption leads to reduced cold tolerance in Drosophila flies. Naturwissenschaften 105(9-10): 59. PubMed ID: 30291448

    Le Bourg, E., Gauthier, T. and Colinet, H. (2015). Feeding on frozen live yeast has some deleterious effects in Drosophila melanogaster. Exp Gerontol [Epub ahead of print]. PubMed ID: 26163343

    Overgaard, J., Sorensen, J. G., Com, E. and Colinet, H. (2014). The rapid cold hardening response of Drosophila melanogaster: Complex regulation across different levels of biological organization. J Insect Physiol. PubMed ID: 24508557

    Colinet, H. and Renault, D. (2014). Dietary live yeast alters metabolic profiles, protein biosynthesis and thermal stress tolerance of Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 170C: 6-14. PubMed ID: 24434805

    Colinet, H., Overgaard, J., Com, E. and Sorensen, J. G. (2013). Proteomic profiling of thermal acclimation in Drosophila melanogaster. Insect Biochem Mol Biol 43: 352-365. PubMed ID: 23416132

  • Nansi Colley Laboratory of Genetics, University of Wisconsin, Madison
    Rosenbaum, E. E., Vasiljevic, E., Cleland, S. C., Flores, C. and Colley, N. J. (2014). The Gos28 SNARE Mediates Intra-Golgi Transport of Rhodopsin and is Required for Photoreceptor Survival. J Biol Chem. PubMed ID: 25261468

    Rosenbaum, E. E., Vasiljevic, E., Brehm, K. S. and Colley, N. J. (2014). Mutations in four glycosyl hydrolases reveal a highly coordinated pathway for rhodopsin biosynthesis and N-glycan trimming in Drosophila melanogaster. PLoS Genet 10: e1004349. PubMed ID: 24785692

    Rosenbaum, E. E., Brehm, K. S., Vasiljevic, E., Gajeski, A. and Colley, N. J. (2012). Drosophila GPI-mannosyltransferase 2 is required for GPI anchor attachment and surface expression of chaoptin. Vis Neurosci 29: 143-156. PubMed ID: 22575127

  • Catherine Collins Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor
    Ir, Waller, T. J., Nye, D. M., Li, J., Zhang, Y., Hume, R. I., Rolls, M. M. and Collins, C. A. (2019). Degeneration of injured axons and dendrites requires restraint of a protective JNK signaling pathway by the transmembrane protein Raw. J Neurosci. PubMed ID: 31492772

    Li, J., Zhang, Y. V., Asghari Adib, E., Stanchev, D. T., Xiong, X., Klinedinst, S., Soppina, P., Jahn, T. R., Hume, R. I., Rasse, T. M. and Collins, C. A. (2017). Restraint of presynaptic protein levels by Wnd/DLK signaling mediates synaptic defects associated with the kinesin-3 motor Unc-104. Elife 6. PubMed ID: 28925357

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

    Mishra, B., Ghannad-Rezaie, M., Li, J., Wang, X., Hao, Y., Ye, B., Chronis, N. and Collins, C. A. (2014). Using microfluidics chips for live imaging and study of injury responses in Drosophila larvae. J Vis Exp. PubMed ID: 24562098

    Mishra, B., Carson, R., Hume, R. I. and Collins, C. A. (2013). Sodium and potassium currents influence wallerian degeneration of injured Drosophila axons. J Neurosci 33: 18728-18739. PubMed ID: 24285879

    Frank, C. A., Wang, X., Collins, C. A., Rodal, A. A., Yuan, Q., Verstreken, P. and Dickman, D. K. (2013). New approaches for studying synaptic development, function, and plasticity using Drosophila as a model system. J Neurosci 33: 17560-17568. PubMed ID: 24198346

  • Barry Condron University of Virginia School of Medicine
    Williamson, M., Mitchell, A. and Condron, B. (2021). Birth temperature followed by a visual critical period determines cooperative group membership. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 34611741

    Dombrovski, M., Kuhar, R., Mitchell, A., Shelton, H. and Condron, B. (2020). Cooperative foraging during larval stage affects fitness in Drosophila. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 206(5): 743-755. PubMed ID: 32623493

    Dombrovski, M., Kim, A., Poussard, L., Vaccari, A., Acton, S., Spillman, E., Condron, B. and Yuan, Q. (2019). A plastic visual pathway regulates cooperative behavior in Drosophila larvae. Curr Biol 29(11): 1866-1876.e1865. PubMed ID: 31130457

    Dombrovski, M., Poussard, L., Moalem, K., Kmecova, L., Hogan, N., Schott, E., Vaccari, A., Acton, S. and Condron, B. (2017). Cooperative behavior emerges among Drosophila larvae. Curr Biol 27(18): 2821-2826.e2822. PubMed ID: 28918946

    Slepian, Z., Sundby, K., Glier, S., McDaniels, J., Nystrom, T., Mukherjee, S., Acton, S. T. and Condron, B. (2015). Visual attraction in Drosophila larvae develops during a critical period and is modulated by crowding conditions. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 26265464

    Basu, S., Condron, B., Aksel, A. and Acton, S. (2013). Segmentation and tracing of single neurons from 3D confocal microscope images. IEEE J Biomed Health Inform 17: 319-335. PubMed ID: 22835569

    Borue, X., Condron, B. and Venton, B. J. (2010). Both synthesis and reuptake are critical for replenishing the releasable serotonin pool in Drosophila. J Neurochem 113: 188-199. PubMed ID: 20070864

  • Boaz Cook Scripps Institute, La Jolla
    Chadha, A., Kaneko, M. and Cook, B. (2015). NOMPC-dependent mechanotransduction shapes the dendrite of proprioceptive neurons. Neurosci Lett. PubMed ID: 25916878

    Chadha, A. and Cook, B. (2014). The effect of stress on motor function in Drosophila. PLoS One 9: e112076. PubMed ID: 25375106

    Desai, B. S., Chadha, A. and Cook, B. (2014). The stum gene is essential for mechanical sensing in proprioceptive neurons. Science 343: 1256-1259. PubMed ID: 24626929

    Kaneko, M., Desai, B. S. and Cook, B. (2013). Ionic leakage underlies a gain-of-function effect of dominant disease mutations affecting diverse P-type ATPases. Nat Genet. PubMed ID: 24336169

    Chadha, A. and Cook, B. (2012). Dissection of gain control mechanisms in Drosophila mechanotransduction. J Neurosci 32: 13052-13061. PubMed ID: 22993423

    Kim, S. E., Coste, B., Chadha, A., Cook, B. and Patapoutian, A. (2012). The role of Drosophila Piezo in mechanical nociception. Nature 483: 209-212. PubMed ID: 22343891

  • Tiffany Cook Center for Molecular Medicine and Genetics, Wayne State University School of Medicine
    Xie, B., Morton, D. B. and Cook, T. A. (2019). Opposing transcriptional and post-transcriptional roles for Scalloped in binary Hippo-dependent neural fate decisions. Dev Biol. PubMed ID: 31265830

    Charlton-Perkins, M. A., Sendler, E. D., Buschbeck, E. K. and Cook, T. A. (2017). Multifunctional glial support by Semper cells in the Drosophila retina. PLoS Genet 13(5): e1006782. PubMed ID: 28562601

    Gresser, A. L., Gutzwiller, L. M., Gauck, M. K., Hartenstein, V., Cook, T. A. and Gebelein, B. (2015). Rhomboid enhancer activity defines a subset of Drosophila neural precursors required for proper feeding, growth and viability. PLoS One 10: e0134915. PubMed ID: 26252385

    Stahl, A. L., Charlton-Perkins, M., Buschbeck, E. K. and Cook, T. A. (2017). The cuticular nature of corneal lenses in Drosophila melanogaster. Dev Genes Evol [Epub ahead of print]. PubMed ID: 28477155

    Jukam, D., Xie, B., Rister, J., Terrell, D., Charlton-Perkins, M., Pistillo, D., Gebelein, B., Desplan, C. and Cook, T. (2013). Opposite Feedbacks in the Hippo Pathway for Growth Control and Neural Fate. Science. PubMed ID: 23989952

    Li-Kroeger, D., Cook, T. A. and Gebelein, B. (2012). Integration of an abdominal Hox complex with Pax2 yields cell-specific EGF secretion from Drosophila sensory precursor cells. Development 139: 1611-1619. PubMed ID: 22438572

    Terrell, D., Xie, B., Workman, M., Mahato, S., Zelhof, A., Gebelein, B. and Cook, T. (2012). OTX2 and CRX rescue overlapping and photoreceptor-specific functions in the Drosophila eye. Dev Dyn 241: 215-228. PubMed ID: 22113834

  • Lynn Cooley Yale University, School of Medicine
    Price, K. L., Tharakan, D. M. and Cooley, L. (2023). Evolutionarily conserved midbody remodeling precedes ring canal formation during gametogenesis. Dev Cell 58(6): 474-488.e475. PubMed ID: 36898376

    Hudson, A. M., Szabo, N. L., Loughran, G., Wills, N. M., Atkins, J. F. and Cooley, L. (2021). Tissue-specific dynamic codon redefinition in Drosophila. Proc Natl Acad Sci U S A 118(5). PubMed ID: 33500350

    Kaufman, R. S., Price, K. L., Mannix, K. M., Ayers, K. M., Hudson, A. M. and Cooley, L. (2020). Drosophila sperm development and intercellular cytoplasm sharing through ring canals do not require an intact fusome. Development 147(22). PubMed ID: 33033119

    Gerdes, J. A., Mannix, K. M., Hudson, A. M. and Cooley, L. (2020). HtsRC-Mediated Accumulation of F-actin Regulates Ring Canal Size During Drosophila melanogaster Oogenesis. Genetics. PubMed ID: 32883702

    Mannix, K. M., Starble, R. M., Kaufman, R. S. and Cooley, L. (2019). Proximity labeling reveals novel interactomes in live Drosophila tissue. Development. PubMed ID: 31208963

    Hudson, A. M., Mannix, K. M., Gerdes, J. A., Kottemann, M. C. and Cooley, L. (2019). Targeted substrate degradation by Kelch controls the actin cytoskeleton during ring canal expansion. Development 146(1). PubMed ID: 30559276

    Hudson, A. M., Mannix, K. M. and Cooley, L. (2015). Actin Cytoskeletal Organization in Drosophila Germline Ring Canals Depends on Kelch Function in a Cullin-RING E3 Ligase. Genetics [Epub ahead of print]. PubMed ID: 26384358

    Mahala Burn, K., Shimada, Y., Ayers, K., Lu, F., Hudson, A. M. and Cooley, L. (2014). Somatic insulin signaling regulates a germline starvation response in Drosophila egg chambers. Dev Biol. PubMed ID: 25481758

    Yan, D., Neumuller, R. A., Buckner, M., Ayers, K., Li, H., Hu, Y., Yang-Zhou, D., Pan, L., Wang, X., Kelley, C., Vinayagam, A., Binari, R., Randklev, S., Perkins, L. A., Xie, T., Cooley, L. and Perrimon, N. (2014). A regulatory network of Drosophila germline stem cell self-renewal. Dev Cell 28: 459-473. PubMed ID: 24576427

    Hudson, A. M. and Cooley, L. (2014). Methods for studying oogenesis. Methods. PubMed ID: 24440745

    McLean, P. F. and Cooley, L. (2013). Bridging the divide: Illuminating the path of intercellular exchange through ring canals. Fly (Austin) 8. PubMed ID: 24406334

    McLean, P. F., Cooley, L. (2013) Protein Equilibration Through Somatic Ring Canals in Drosophila. Science. PubMed ID: 23704373

  • Robin Cooper Department of Biology, University of Kentucky, Lexington
    Vacassenno, R. M., Haddad, C. N. and Cooper, R. L. (2023). The effects of doxapram (blocker of K2p channels) on resting membrane potential and synaptic transmission at the Drosophila neuromuscular junction. Comp Biochem Physiol C Toxicol Pharmacol 263: 109497. PubMed ID: 36306997

    Potter, R., Meade, A., Potter, S. and Cooper, R. L. (2021). Rapid and Direct Action of Lipopolysaccharide (LPS) on Skeletal Muscle of Larval Drosophila. Biology (Basel) 10(12). PubMed ID: 34943150

    Marguerite, N. T., Bernard, J., Harrison, D. A., Harris, D. and Cooper, R. L. (2021). Effect of Temperature on Heart Rate for Phaenicia sericata and Drosophila melanogaster with Altered Expression of the TrpA1 Receptors. Insects 12(1). PubMed ID: 33418937

    Greenhalgh, A., Istas, O. and Cooper, R. L. (2020). Bacterial endotoxin lipopolysaccharide enhances synaptic transmission at low-output glutamatergic synapse. Neurosci Res. PubMed ID: 32987087

    Potter, S., Sifers, J., Yocom, E., Blumich, S. L. E., Potter, R., Nadolski, J., Harrison, D. A. and Cooper, R. L. (2019). Effects of inhibiting mTOR with rapamycin on behavior, development, neuromuscular physiology and cardiac function in larval Drosophila. Biol Open 8(11). PubMed ID: 31704693

    Malloy, C. A., Somasundaram, E., Omar, A., Bhutto, U., Medley, M., Dzubuk, N. and Cooper, R. L. (2019). Pharmacological identification of cholinergic receptor subtypes: modulation of locomotion and neural circuit excitability in Drosophila larvae. Neuroscience. PubMed ID: 31102763

    Istas, O., Greenhalgh, A. and Cooper, R. (2019). The effects of a bacterial endotoxin on behavior and sensory-CNS-Motor circuits in Drosophila melanogaster. Insects 10(4). PubMed ID: 31013568

    Mattingly, M., Weineck, K., Costa, J. and Cooper, R. L. (2018). Hyperpolarization by activation of halorhodopsin results in enhanced synaptic transmission: Neuromuscular junction and CNS circuit. PLoS One 13(7): e0200107. PubMed ID: 29969493

    Malloy, C., Sifers, J., Mikos, A., Samadi, A., Omar, A., Hermanns, C. and Cooper, R. L. (2017). Using optogenetics to assess neuroendocrine modulation of heart rate in Drosophila melanogaster larvae. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 28612236

    Majeed, Z. R., Abdeljaber, E., Soveland, R., Cornwell, K., Bankemper, A., Koch, F. and Cooper, R. L. (2016). Modulatory action by the serotonergic system: behavior and neurophysiology in Drosophila melanogaster. Neural Plast 2016: 7291438. PubMed ID: 26989517

  • Victor Corces Emory University, Department of Biology
    Fresan, U., Rodriguez-Sanchez, M. A., Reina, O., Corces, V. G. and Espinas, M. L. (2020). Haspin kinase modulates nuclear architecture and Polycomb-dependent gene silencing. PLoS Genet 16(8): e1008962. PubMed ID: 32750047

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

    Rowley, M. J., Lyu, X., Rana, V., Ando-Kuri, M., Karns, R., Bosco, G. and Corces, V. G. (2019). Condensin II counteracts Cohesin and RNA Polymerase II in the establishment of 3D chromatin organization. Cell Rep 26(11): 2890-2903.e2893. PubMed ID: 30865881

    Rowley, M. J., Nichols, M. H., Lyu, X., Ando-Kuri, M., Rivera, I. S. M., Hermetz, K., Wang, P., Ruan, Y. and Corces, V. G. (2017). Evolutionarily conserved principles predict 3D chromatin organization. Mol Cell 67(5): 837-852.e837. PubMed ID: 28826674

    Cubenas-Potts, C., Rowley, M. J., Lyu, X., Li, G., Lei, E. P. and Corces, V. G. (2016). Different enhancer classes in Drosophila bind distinct architectural proteins and mediate unique chromatin interactions and 3D architecture. Nucleic Acids Res. PubMed ID: 27899590

    Van Bortle, K., Nichols, M. H., Ramos, E. and Corces, V. G. (2015). Integrated tRNA, transcript, and protein profiles in response to steroid hormone signaling. RNA. PubMed ID: 26289344

    Van Bortle, K., Peterson, A. J., Takenaka, N., O'Connor, M. B. and Corces, V. G. (2015). CTCF-dependent co-localization of canonical Smad signaling factors at architectural protein binding sites in D. melanogaster. Cell Cycle: [Epub ahead of print]. PubMed ID: 26125535

    Gomez-Diaz, E., Rivero, A., Chandre, F. and Corces, V. G. (2014). Insights into the epigenomic landscape of the human malaria vector Anopheles gambiae. Front Genet 5: 277. PubMed ID: 25177345

    Van Bortle, K., Nichols, M. H., Li, L., Ong, C. T., Takenaka, N., Qin, Z. S. and Corces, V. G. (2014). Insulator function and topological domain border strength scale with architectural protein occupancy. Genome Biol 15: R82. PubMed ID: 24981874

    Ong, C. T., Van Bortle, K., Ramos, E. and Corces, V. G. (2013). Poly(ADP-ribosyl)ation Regulates Insulator Function and Intrachromosomal Interactions in Drosophila. Cell. PubMed ID: 24055367

  • Montserrat Corominas Department of Genetics, University of Barcelona
    Camilleri-Robles, C., Serras, F. and Corominas, M. (2019). Role of D-GADD45 in JNK-Dependent Apoptosis and Regeneration in Drosophila. Genes (Basel) 10(5). PubMed ID: 31109086

    Vizcaya-Molina, E., Klein, C. C., Serras, F., Mishra, R. K., Guigo, R. and Corominas, M. (2018). Damage-responsive elements in Drosophila regeneration. Genome Res 28(12): 1852-1866. PubMed ID: 30459214

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

    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

    Turkel, N., Sahota, V. K., Bolden, J. E., Goulding, K. R., Doggett, K., Willoughby, L. F., Blanco, E., Martin-Blanco, E., Corominas, M., Ellul, J., Aigaki, T., Richardson, H. E. and Brumby, A. M. (2013). The BTB-zinc Finger Transcription Factor Abrupt Acts as an Epithelial Oncogene in Drosophila melanogaster through Maintaining a Progenitor-like Cell State. PLoS Genet 9: e1003627. PubMed ID: 23874226

    Blanco, E. and Corominas, M. (2012). CBS: an open platform that integrates predictive methods and epigenetics information to characterize conserved regulatory features in multiple Drosophila genomes. BMC Genomics 13: 688. PubMed ID: 23228284

    Mora, N., Almudi, I., Alsina, B., Corominas, M. and Serras, F. (2013). beta amyloid protein precursor-like (Appl) is a Ras1/MAPK-regulated gene required for axonal targeting in Drosophila photoreceptor neurons. J Cell Sci 126: 53-59. PubMed ID: 23178937

  • Rudi Costa University of Padua
    Andreatta, G., Montagnese, S. and Costa, R. (2022). Natural alleles of the clock gene timeless differentially affect life-history traits in Drosophila. Front Physiol 13: 1092951. PubMed ID: 36703932

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

    Cusumano, P., Damulewicz, M., Carbognin, E., Caccin, L., Puricella, A., Specchia, V., Bozzetti, M. P., Costa, R. and Mazzotta, G. M. (2019). The RNA helicase BELLE is involved in circadian rhythmicity and in transposons regulation in Drosophila melanogaster. Front Physiol 10: 133. PubMed ID: 30842743

    Mazzotta, G. M., Bellanda, M., Minervini, G., Damulewicz, M., Cusumano, P., Aufiero, S., Stefani, M., Zambelli, B., Mammi, S., Costa, R. and Tosatto, S. C. E. (2018). Calmodulin Enhances Cryptochrome Binding to INAD in Drosophila Photoreceptors. Front Mol Neurosci 11: 280. PubMed ID: 30177872

    Cusumano, P., Biscontin, A., Sandrelli, F., Mazzotta, G. M., Tregnago, C., De Pitta, C. and Costa, R. (2018). Modulation of miR-210 alters phasing of circadian locomotor activity and impairs projections of PDF clock neurons in Drosophila melanogaster. PLoS Genet 14(7): e1007500. PubMed ID: 30011269

    Nagy, D., Andreatta, G., Bastianello, S., Martin Anduaga, A., Mazzotta, G., Kyriacou, C. P. and Costa, R. (2018). A semi-natural approach for studying seasonal diapause in Drosophila melanogaster reveals robust photoperiodicity. J Biol Rhythms: 748730417754116. PubMed ID: 29415605
    Andreatta, G., Kyriacou, C. P., Flatt, T. and Costa, R. (2018). Aminergic signaling controls ovarian dormancy in Drosophila. Sci Rep 8(1): 2030. PubMed ID: 29391447

    Damulewicz, M., Mazzotta, G. M., Sartori, E., Rosato, E., Costa, R. and Pyza, E. M. (2017). Cryptochrome is a regulator of synaptic plasticity in the visual system of Drosophila melanogaster. Front Mol Neurosci 10: 165. PubMed ID: 28611590

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

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

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

  • Sue Cotterill St George's, University of London
    Vo, N., Anh Suong, D. N., Yoshino, N., Yoshida, H., Cotterill, S. and Yamaguchi, M. (2016). Novel roles of HP1a and Mcm10 in DNA replication, genome maintenance and photoreceptor cell differentiation. Nucleic Acids Res. PubMed ID: 27903903

    Sahashi, R., Crevel, G., Pasko, J., Suyari, O., Nagai, R., Saura, M. M., Yamaguchi, M. and Cotterill, S. (2014). DNA polymerase alpha interacts with PrSet7 and mediates H4K20 monomethylation in Drosophila. J Cell Sci. PubMed ID: 24806961

    Vo, N., Taga, A., Inaba, Y., Yoshida, H., Cotterill, S. and Yamaguchi, M. (2014). Drosophila Mcm10 is required for DNA replication and differentiation in the compound eye. PLoS One 9: e93450. PubMed ID: 24686397

    Sahashi, R., Matsuda, R., Suyari, O., Kawai, M., Yoshida, H., Cotterill, S. and Yamaguchi, M. (2013). Functional analysis of Drosophila DNA polymerase epsilon p58 subunit. Am J Cancer Res 3: 478-489. PubMed ID: 24224125

  • Albert Courey Department of Chemistry & Biochemistry, UCLA
    Chambers, M., Turki-Judeh, W., Kim, M. W., Chen, K., Gallaher, S. D. and Courey, A. J. (2017). Mechanisms of Groucho-mediated repression revealed by genome-wide analysis of Groucho binding and activity. BMC Genomics 18(1): 215. PubMed ID: 28245789

    Kwong, P. N., Chambers, M., Vashisht, A. A., Turki-Judeh, W., Yau, T. Y., Wohlschlegel, J. A. and Courey, A. J. (2015). The central region of the Drosophila co-repressor Groucho as a regulatory hub. J Biol Chem [Epub ahead of print]. PubMed ID: 26483546

    Kwong, P. N., Chambers, M., Vashisht, A. A., Turki-Judeh, W., Yau, T. Y., Wohlschlegel, J. A. and Courey, A. J. (2015). The Central Region of the Drosophila Co-Repressor Groucho as a Regulatory Hub. J Biol Chem. PubMed ID: 26483546

    Smith, M., Turki-Judeh, W. and Courey, A. J. (2012). SUMOylation in Drosophila Development. Biomolecules 2: 331-349. PubMed ID: 24970141

    Ajuria, L., Nieva, C., Winkler, C., Kuo, D., Samper, N., Andreu, M. J., Helman, A., Gonzalez-Crespo, S., Paroush, Z., Courey, A. J. and Jimenez, G. (2011). Capicua DNA-binding sites are general response elements for RTK signaling in Drosophila. Development 138: 915-924. PubMed ID: 21270056

  • Juan Couso University of Sussex
    Magny, E. G., Platero, A. I., Bishop, S. A., Pueyo, J. I., Aguilar-Hidalgo, D. and Couso, J. P. (2021). Pegasus, a small extracellular peptide enhancing short-range diffusion of Wingless. Nat Commun 12(1): 5660. PubMed ID: 34580289

    Patraquim, P., Mumtaz, M. A. S., Pueyo, J. I., Aspden, J. L. and Couso, J. P. (2020). Developmental regulation of canonical and small ORF translation from mRNAs. Genome Biol 21(1): 128. PubMed ID: 32471506

    Aspden, J. L., Eyre-Walker, Y. C., Philips, R. J., Amin, U., Mumtaz, M. A., Brocard, M. and Couso, J. P. (2014). Extensive translation of small ORFs revealed by Poly-Ribo-Seq. Elife: e03528. PubMed ID: 25144939

    Sampson, C. J., Amin, U. and Couso, J. P. (2013). Activation of Drosophila hemocyte motility by the ecdysone hormone. Biol Open. PubMed ID: 24285708

    Magny, E. G., Pueyo, J. I., Pearl, F. M., Cespedes, M. A., Niven, J. E., Bishop, S. A. and Couso, J. P. (2013). Conserved Regulation of Cardiac Calcium Uptake by Peptides Encoded in Small Open Reading Frames. Science. PubMed ID: 23970561

    Ladoukakis, E., Pereira, V., Magny, E. G., Eyre-Walker, A. and Couso, J. P. (2011). Hundreds of putatively functional small open reading frames in Drosophila. Genome Biol 12: R118. PubMed ID: 22118156

    Galindo, M. I., Fernandez-Garza, D., Phillips, R. and Couso, J. P. (2011). Control of Distal-less expression in the Drosophila appendages by functional 3' enhancers. Dev Biol 353: 396-410. PubMed ID: 21320482

    Bosch, M., Bishop, S. A., Baguna, J. and Couso, J. P. (2010). Leg regeneration in Drosophila abridges the normal developmental program. Int J Dev Biol 54: 1241-1250. PubMed ID: 20563988

  • Virginie Courtier-Orgogozo Institut Jacques Monod, Universite Paris Diderot
    Suvorov, A., Kim, B. Y., Wang, J., Armstrong, E. E., Peede, D., D'Agostino, E. R. R., Price, D. K., Waddell, P., Lang, M., Courtier-Orgogozo, V., David, J. R., Petrov, D., Matute, D. R., Schrider, D. R. and Comeault, A. A. (2021). Widespread introgression across a phylogeny of 155 Drosophila genomes. Curr Biol. PubMed ID: 34788634

    Borne, F., Kulathinal, R. J. and Courtier-Orgogozo, V. (2021). Glue genes are subjected to diverse selective forces during Drosophila development. Genome Biol Evol. PubMed ID: 34788814

    Peluffo, A. E., Hamdani, M., Vargas-Valderrama, A., David, J. R., Mallard, F., Graner, F. and Courtier-Orgogozo, V. (2021). A morphological trait involved in reproductive isolation between Drosophila sister species is sensitive to temperature. Ecol Evol 11(12): 7492-7506. PubMed ID: 34188829

    Borne, F., Prigent, S. R., Molet, M. and Courtier-Orgogozo, V. (2021). Drosophila glue protects from predation. Proc Biol Sci 288(1947): 20210088. PubMed ID: 33726597

    Borne, F., Kovalev, A., Gorb, S. and Courtier-Orgogozo, V. (2020). The glue produced by Drosophila melanogaster for pupa adhesion is universal. J Exp Biol. PubMed ID: 32165432

    Da Lage, J. L., Thomas, G. W. C., Bonneau, M. and Courtier-Orgogozo, V. (2019). Evolution of salivary glue genes in Drosophila species. BMC Evol Biol 19(1): 36. PubMed ID: 30696414

    Nagy, O., Nuez, I., Savisaar, R., Peluffo, A. E., Yassin, A., Lang, M., Stern, D. L., Matute, D. R., David, J. R. and Courtier-Orgogozo, V. (2018). Correlated evolution of two copulatory organs via a single cis-regulatory nucleotide change. Curr Biol 28(21): 3450-3457.e3413. PubMed ID: 30344115

    Lang, M., Nagy, O., Lang, C. and Orgogozo, V. (2016). High throughput preparation of fly genomic DNA in 96-well format using a paint-shaker. Fly (Austin): 0. PubMed ID: 26818699

    Peluffo, A. E., Nuez, I., Debat, V., Savisaar, R., Stern, D. L. and Orgogozo, V. (2015). A major locus controls a genital shape difference involved in reproductive isolation between Drosophila yakuba and Drosophila santomea. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26511499

    Yassin, A., Orgogozo, V. (2013) Coevolution between Male and Female Genitalia in the Drosophila melanogaster Species Subgroup. PLoS One 8: e57158. Pubmed ID: 23451172

    Lang, M., Murat, S., Clark, A. G., Gouppil, G., Blais, C., Matzkin, L. M., Guittard, E., Yoshiyama-Yanagawa, T., Kataoka, H., Niwa, R., Lafont, R., Dauphin-Villemant, C. and Orgogozo, V. (2012). Mutations in the neverland gene turned Drosophila pachea into an obligate specialist species. Science 337: 1658-1661. PubMed ID: 23019649

    Lang, M. and Orgogozo, V. (2011). Identification of homologous gene sequences by PCR with degenerate primers. Methods Mol Biol 772: 245-256. PubMed ID: 22065442

  • Daniel Cox Georgia State University, Atlanta
    Lottes, E. N., Ciger, F. H., Bhattacharjee, S., Timmins-Wilde, E. A., Tete, B., Tran, T., Matta, J., Patel, A. A. and Cox, D. N. (2023). CCT and Cullin1 regulate the TORC1 pathway to promote dendritic arborization in health and disease. bioRxiv. PubMed ID: 37577581

    Patel, A. A., Cardona, A. and Cox, D. N. (2023). Neural substrates of cold nociception in Drosophila larva. bioRxiv. PubMed ID: 37577520

    Nanda, S., Bhattacharjee, S., Cox, D. N. and Ascoli, G. A. (2023). Local microtubule and F-actin distributions fully determine the spatial geometry of Drosophila sensory dendritic arbors. bioRxiv. PubMed ID: 36909461

    Bhattacharjee, S., Iyer, E. P. R., Iyer, S. C., Nanda, S., Rubaharan, M., Ascoli, G. A. and Cox, D. N. (2023). The Zinc-BED Transcription Factor Bedwarfed Promotes Proportional Dendritic Growth and Branching through Transcriptional and Translational Regulation in Drosophila. Int J Mol Sci 24(7). PubMed ID: 37047316

    Himmel, N. J., Sakurai, A., Patel, A. A., Bhattacharjee, S., Letcher, J. M., Benson, M. N., Gray, T. R., Cymbalyuk, G. S. and Cox, D. N. (2023). Chloride-dependent mechanisms of multimodal sensory discrimination and nociceptive sensitization in Drosophila. Elife 12. PubMed ID: 36688373

    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

    Himmel, N. J., Letcher, J. M., Sakurai, A., Gray, T. R., Benson, M. N., Donaldson, K. J. and Cox, D. N. (2021). Identification of a neural basis for cold acclimation in Drosophila larvae. iScience 24(6): 102657. PubMed ID: 34151240

    Nanda, S., Bhattacharjee, S., Cox, D. N. and Ascoli, G. A. (2020). Distinct Relations of Microtubules and Actin Filaments with Dendritic Architecture. iScience 23(12): 101865. PubMed ID: 33319182

    Hartwig, C., Mrndez, G. M., Bhattacharjee, S., Vrailas-Mortimer, A. D., Zlatic, S. A., Freeman, A. A. H., Gokhale, A., Concilli, M., Werner, E., Sapp Savas, C., Rudin-Rush, S., Palmer, L., Shearing, N., Margewich, L., McArthy, J., Taylor, S., Roberts, B., Lupashin, V., Polishchuk, R. S., Cox, D. N., Jorquera, R. A. and Faundez, V. (2020). Golgi-Dependent Copper Homeostasis Sustains Synaptic Development and Mitochondrial Content. J Neurosci. PubMed ID: 33208468

    Himmel, N. J., Letcher, J. M., Sakurai, A., Gray, T. R., Benson, M. N. and Cox, D. N. (2019). Drosophila menthol sensitivity and the Precambrian origins of transient receptor potential-dependent chemosensation. Philos Trans R Soc Lond B Biol Sci 374(1785): 20190369. PubMed ID: 31544603

    Clark, S. G., Graybeal, L. L., Bhattacharjee, S., Thomas, C., Bhattacharya, S. and Cox, D. N. (2018). Basal autophagy is required for promoting dendritic terminal branching in Drosophila sensory neurons. PLoS One 13(11): e0206743. PubMed ID: 30395636

    Im, S. H., Patel, A. A., Cox, D. N. and Galko, M. J. (2018). Drosophila Insulin receptor regulates the persistence of injury-induced nociceptive sensitization. Dis Model Mech 11(5). PubMed ID: 29752280

    Nanda, S., Das, R., Bhattacharjee, S., Cox, D. N. and Ascoli, G. A. (2017). Morphological determinants of dendritic arborization neurons in Drosophila larva. Brain Struct Funct. PubMed ID: 29094302

  • Rachael Cox USUHS, Department of Biochemistry and Molecular Biology, Bethesda
    Sen, A. and Cox, R. T. (2022). Loss of Drosophila Clueless differentially affects the mitochondrial proteome compared to loss of Sod2 and Pink1. Front Physiol 13: 1004099. PubMed ID: 36388112

    Saoji, M., Petersen, C. E., Sen, A., Tripoli, B. A., Smyth, J. T. and Cox, R. T. (2022). Reduction of Drosophila Mitochondrial RNase P in Skeletal and Heart Muscle Causes Muscle Degeneration, Cardiomyopathy, and Heart Arrhythmia. Front Cell Dev Biol 10: 788516. PubMed ID: 35663400

    Sheard, K. M., Thibault-Sennett, S. A., Sen, A., Shewmaker, F. and Cox, R. T. (2019). Clueless forms dynamic, insulin-responsive bliss particles sensitive to stress. Dev Biol. PubMed ID: 31837288

    Sen, A., Karasik, A., Shanmuganathan, A., Mirkovic, E., Koutmos, M. and Cox, R.T. (2016). Loss of the mitochondrial protein-only ribonuclease P complex causes aberrant tRNA processing and lethality in Drosophila. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 27131785

    Sen, A. and Cox, R. T. (2016). Clueless is a conserved ribonucleoprotein that binds the ribosome at the mitochondrial outer membrane. Biol Open [Epub ahead of print]. PubMed ID: 26834020

    Sen, A., Kalvakuri, S., Bodmer, R. and Cox, R. T. (2015). Clueless, a protein required for mitochondrial function, interacts with the PINK1-Parkin complex in Drosophila. Dis Model Mech 8: 577-589. PubMed ID: 26035866

    Cox, R. T. and Spradling, A. C. (2009). Clueless, a conserved Drosophila gene required for mitochondrial subcellular localization, interacts genetically with parkin. Dis Model Mech 2: 490-499. PubMed ID: 19638420

  • Brian Crane Department of Chemistry and Chemical Biology and Biotechnology Resource Center, Cornell University, Ithaca
    Lin, C., Schneps, C. M., Chandrasekaran, S., Ganguly, A. and Crane, B. R. (2022). Mechanistic insight into light-dependent recognition of Timeless by Drosophila Cryptochrome. Structure. PubMed ID: 35397203

    Chandrasekaran, S., Schneps, C. M., Dunleavy, R., Lin, C., DeOliveira, C. C., Ganguly, A. and Crane, B. R. (2021). Tuning flavin environment to detect and control light-induced conformational switching in Drosophila cryptochrome. Commun Biol 4(1): 249. PubMed ID: 33637846

    Lin, C., Top, D., Manahan, C. C., Young, M. W. and Crane, B. R. (2018). Circadian clock activity of cryptochrome relies on tryptophan-mediated photoreduction. Proc Natl Acad Sci U S A 115(15): 3822-3827. PubMed ID: 29581265

    Vaidya, A. T., Top, D., Manahan, C. C., Tokuda, J. M., Zhang, S., Pollack, L., Young, M. W. and Crane, B. R. (2013). Flavin reduction activates Drosophila cryptochrome. Proc Natl Acad Sci U S A. PubMed ID: 24297896

    Levy, C., Zoltowski, B. D., Jones, A. R., Vaidya, A. T., Top, D., Widom, J., Young, M. W., Scrutton, N. S., Crane, B. R. and Leys, D. (2013). Updated structure of Drosophila cryptochrome. Nature 495: E3-4. PubMed ID: 23518567

    Zoltowski, B. D., Vaidya, A. T., Top, D., Widom, J., Young, M. W. and Crane, B. R. (2011). Structure of full-length Drosophila cryptochrome. Nature 480: 396-399. PubMed ID: 22080955

    King, H. A., Hoelz, A., Crane, B. R. and Young, M. W. (2011). Structure of an enclosed dimer formed by the Drosophila period protein. J Mol Biol 413: 561-572. PubMed ID: 21907720

  • Steven Crews School of Medicine, University of North Carolina at Chapel Hill
    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: 3723-3732. PubMed ID: 27802137

    Heckscher, E. S., Long, F., Layden, M. J., Chuang, C. H., Manning, L., Richart, J., Pearson, J. C., Crews, S. T., Peng, H., Myers, E. and Doe, C. Q. (2014). Atlas-builder software and the eNeuro atlas: resources for developmental biology and neuroscience. Development 141: 2524-2532. PubMed ID: 24917506

    Pearson, J. C. and Crews, S. T. (2014). Enhancer diversity and the control of a simple pattern of Drosophila CNS midline cell expression. Dev Biol. PubMed ID: 24854999

    Pearson, J. C. and Crews, S. T. (2014). Enhancer diversity and the control of a simple pattern of drosophila CNS midline cell expression. Dev Biol. PubMed ID: 24854999

    Fontana, J. R. and Crews, S. T. (2012). Transcriptome analysis of Drosophila CNS midline cells reveals diverse peptidergic properties and a role for castor in neuronal differentiation. Dev Biol 372: 131-142. PubMed ID: 23010511

    Watson, J. D. and Crews, S. T. (2012). Formation and specification of a Drosophila dopaminergic precursor cell. Development 139: 3316-3325. PubMed ID: 22874915

    Pearson, J. C., Watson, J. D. and Crews, S. T. (2012). Drosophila melanogaster Zelda and Single-minded collaborate to regulate an evolutionarily dynamic CNS midline cell enhancer. Dev Biol 366: 420-432. PubMed ID: 22537497

  • Richard Cripps University of New Mexico
    Vaziri, P., Ryan, D., Johnston, C. A. and Cripps, R. M. (2020). A Novel Mechanism for Activation of Myosin Regulatory Light Chain by Protein Kinase C-Delta in Drosophila. Genetics 216(1): 177-190. PubMed ID: 32753389

    Czajkowski, E. R., Cisneros, M., Garcia, B. S., Shen, J. and Cripps, R. M. (2020). The Drosophila CG1674 gene encodes a synaptopodin 2-like related protein that localizes to the Z-disc and is required for normal flight muscle development and function. Dev Dyn. PubMed ID: 32893414

    Vishal, K., Lovato, T. L., Bragg, C., Chechenova, M. B. and Cripps, R. M. (2020). FGF signaling promotes myoblast proliferation through activation of wingless signaling. Dev Biol. PubMed ID: 32445643

    Lovato, C. V., Lovato, T. L. and Cripps, R. M. (2019). Crossveinless is a direct transcriptional target of Trachealess and Tango in Drosophila tracheal precursor cells. PLoS One 14(6): e0217906. PubMed ID: 31158257

    DeAguero, A. A., Castillo, L., Oas, S. T., Kiani, K., Bryantsev, A. L. and Cripps, R. M. (2019). Regulation of fiber-specific actin expression by the Drosophila SRF ortholog Blistered. Development. PubMed ID: 30872277

    Dohn, T. E. and Cripps, R. M. (2018). Absence of the Drosophila jump muscle actin Act79B is compensated by up-regulation of Act88F. Dev Dyn [Epub ahead of print]. PubMed ID: 29318731

    Chechenova, M. B., Maes, S. and Cripps, R. M. (2015). Expression of the Troponin C at 41C gene in adult Drosophila tubular muscles depends upon both positive and negative regulatory inputs. PLoS One 10: e0144615. PubMed ID: 26641463

    Lovato, T. L., Sensibaugh, C. A., Swingle, K. L., Martinez, M. M. and Cripps, R. M. (2015). The Drosophila transcription factors Tinman and Pannier activate and collaborate with Myocyte enhancer factor-2 to promote heart cell fate. PLoS One 10: e0132965. PubMed ID: 26225919

    Brunetti, T.M., Fremin, B.J. and Cripps, R.M. (2015). Identification of singles bar as a direct transcriptional target of Drosophila Myocyte enhancer factor-2 and a regulator of adult myoblast fusion. Dev Biol [Epub ahead of print]. PubMed ID: 25797154

    Elwell, J. A., Lovato, T. L., Adams, M. M., Baca, E. M., Lee, T. and Cripps, R. M. (2015). The myogenic repressor gene Holes in muscles is a direct transcriptional target of Twist and Tinman in the Drosophila embryonic mesoderm. Dev Biol. PubMed ID: 25704510

    Oas, S. T., Bryantsev, A. L. and Cripps, R. M. (2014). Arrest is a regulator of fiber-specific alternative splicing in the indirect flight muscles of Drosophila. J Cell Biol 206: 895-908. PubMed ID: 25246617

    Morriss, G. R., Jaramillo, C. T., Mikolajczak, C. M., Duong, S., Jaramillo, M. S. and Cripps, R. M. (2013). The Drosophila wings apart Gene Anchors a Novel, Evolutionarily-Conserved Pathway of Neuromuscular Development. Genetics. PubMed ID: 24026097

  • Justin Crocker European Molecular Biology Laboratory, Heidelberg, Germany
    Li, X. C., Fuqua, T., van Breugel, M. E. and Crocker, J. (2023). Mutational scans reveal differential evolvability of Drosophila promoters and enhancers. Philos Trans R Soc Lond B Biol Sci 378(1877): 20220054. PubMed ID: 37004721

    Gandara, L., Tsai, A., Ekelöf, M., Galupa, R., Preger-Ben Noon, E., Alexandrov, T. and Crocker, J. (2022). Developmental phenomics suggests that H3K4 monomethylation confers multi-level phenotypic robustness. Cell Rep 41(11): 111832. PubMed ID: 36516782

    Galupa, R., Alvarez-Canales, G., Borst, N. O., Fuqua, T., Gandara, L., Misunou, N., Richter, K., Alves, M. R. P., Karumbi, E., Perkins, M. L., Kocijan, T., Rushlow, C. A. and Crocker, J. (2023). Enhancer architecture and chromatin accessibility constrain phenotypic space during Drosophila development. Dev Cell 58(1): 51-62. PubMed ID: 36626871

    Tsai, A. and Crocker, J. (2022). Nuclear morphogenesis: forming a heterogeneous nucleus during embryogenesis. Development 149(4). PubMed ID: 35142344

    Fuqua, T., Jordan, J., Halavatyi, A., Tischer, C., Richter, K. and Crocker, J. (2021). An open-source semi-automated robotics pipeline for embryo immunohistochemistry. Sci Rep 11(1): 10314. PubMed ID: 33986394

    Fuqua, T., Jordan, J., van Breugel, M. E., Halavatyi, A., Tischer, C., Polidoro, P., Abe, N., Tsai, A., Mann, R. S., Stern, D. L. and Crocker, J. (2020). Dense and pleiotropic regulatory information in a developmental enhancer. Nature 587(7833): 235-239. PubMed ID: 33057197

    Tsai, A., Galupa, R. and Crocker, J. (2020). Robust and efficient gene regulation through localized nuclear microenvironments. Development 147(19). PubMed ID: 33020073


  • Damian Crowther Cambridge Neuroscience
    Younan, N. D., Chen, K. F., Rose, R. S., Crowther, D. C. and Viles, J. H. (2018). Prion protein stabilizes amyloid-beta (Abeta) oligomers and enhances Abeta neurotoxicity in a Drosophila model of Alzheimer disease. J Biol Chem. PubMed ID: 29887525

    Pavel, M., Imarisio, S., Menzies, F. M., Jimenez-Sanchez, M., Siddiqi, F. H., Wu, X., Renna, M., O'Kane, C. J., Crowther, D. C. and Rubinsztein, D. C. (2016). CCT complex restricts neuropathogenic protein aggregation via autophagy. Nat Commun 7: 13821. PubMed ID: 27929117

    Ott, S., Vishnivetskaya, A., Malmendal, A. and Crowther, D.C. (2016). Metabolic changes may precede proteostatic dysfunction in a Drosophila model of amyloid beta peptide toxicity. Neurobiol Aging 41: 39-52. PubMed ID: 27103517

    Lim, J. Y., Ott, S. and Crowther, D. C. (2016). Drosophila melanogaster as a Model for Studies on the Early Stages of Alzheimer's Disease. Methods Mol Biol 1303: 227-239. PubMed ID: 26235070

    Ott, S., Dziadulewicz, N. and Crowther, D. C. (2015). Iron is a specific cofactor for distinct oxidation- and aggregation-dependent Aβ toxicity mechanisms. Dis Model Mech [Epub ahead of print]. PubMed ID: 26035384

    Lim, J. Y., Reighard, C. P. and Crowther, D. C. (2015). The pro-domains of neurotrophins, including BDNF, are linked to Alzheimer's disease through a toxic synergy with Abeta. Hum Mol Genet. PubMed ID: 25954034

    Chen, K. F. and Crowther, D. C. (2014). Insights into amyloid disease from fly models. Essays Biochem 56: 69-83. PubMed ID: 25131587

    Chen, K. F., Possidente, B., Lomas, D. A. and Crowther, D. C. (2014). The central molecular clock is robust in the face of behavioural arrhythmia in a Drosophila model of Alzheimer's disease. Dis Model Mech. PubMed ID: 24574361

    Kruppa, A. J., Ott, S., Chandraratna, D. S., Irving, J. A., Page, R. M., Speretta, E., Camargo, L. M., Marciniak, S. J., Lomas, D. A. and Crowther, D. C. (2013). Suppression of Abeta toxicity by puromycin-sensitive aminopeptidase is independent of its proteolytic activity. Biochim Biophys Acta. PubMed ID: 23911349

    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

    Chen, K. F. and Crowther, D. C. (2012). Functional genomics in Drosophila models of human disease. Brief Funct Genomics 11: 405-415. PubMed ID: 22914042

  • Michéle Crozatier Centre de Biologie du Développement, Toulouse
    Morin-Poulard, I., Destalminil-Letourneau, M., Bataille, L., Frendo, J. L., Lebreton, G., Vanzo, N. and Crozatier, M. (2022). Identification of Bipotential Blood Cell/Nephrocyte Progenitors in Drosophila: Another Route for Generating Blood Progenitors. Front Cell Dev Biol 10: 834720. PubMed ID: 35237606

    Morin-Poulard, I., Tian, Y., Vanzo, N. and Crozatier, M. (2021). Drosophila as a Model to Study Cellular Communication Between the Hematopoietic Niche and Blood Progenitors Under Homeostatic Conditions and in Response to an Immune Stress. Front Immunol 12: 719349. PubMed ID: 34484226

    Leitao, A. B., Arunkumar, R., Day, J. P., Geldman, E. M., Morin-Poulard, I., Crozatier, M. and Jiggins, F. M. (2020). Constitutive activation of cellular immunity underlies the evolution of resistance to infection in Drosophila. Elife 9. PubMed ID: 33357377

    Destalminil-Letourneau, M., Morin-Poulard, I., Tian, Y., Vanzo, N. and Crozatier, M. (2021). The vascular niche controls Drosophila hematopoiesis via fibroblast growth factor signaling. Elife 10. PubMed ID: 33395389

    Louradour, I., Sharma, A., Morin-Poulard, I., Letourneau, M., Vincent, A., Crozatier, M. and Vanzo, N. (2017). Reactive oxygen species-dependent Toll/NF-kappaB activation in the Drosophila hematopoietic niche confers resistance to wasp parasitism. Elife 6. PubMed ID: 29091025

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

    Dubois, L., Frendo, J. L., Chanut-Delalande, H., Crozatier, M. and Vincent, A. (2016). Genetic dissection of the transcription factor code controlling serial specification of muscle identities in Drosophila. Elife 5 [Epub ahead of print]. PubMed ID: 27438571

    Morin-Poulard, I., Sharma, A., Louradour, I., Vanzo, N., Vincent, A. and Crozatier, M. (2016). Vascular control of the Drosophila haematopoietic microenvironment by Slit/Robo signalling. Nat Commun 7: 11634. PubMed ID: 27193394

    Oyallon, J., Vanzo, N., Krzemien, J., Morin-Poulard, I., Vincent, A. and Crozatier, M. (2016). Two independent functions of Collier/Early B Cell Factor in the control of Drosophila blood cell homeostasis. PLoS One 11: e0148978. PubMed ID: 26866694

    de Taffin, M., Carrier, Y., Dubois, L., Bataille, L., Painset, A., Le Gras, S., Jost, B., Crozatier, M. and Vincent, A. (2015). Genome-Wide Mapping of Collier In Vivo Binding Sites Highlights Its Hierarchical Position in Different Transcription Regulatory Networks. PLoS One 10: e0133387. PubMed ID: 26204530

  • Jennifer Curtiss New Mexico State University
    Banerjee, S., Woods, C., Burnett, M., Park, S. J., Ja, W. W. and Curtiss, J. (2021). The Drosophila melanogaster Neprilysin Nepl15 is involved in lipid and carbohydrate storage. Sci Rep 11(1): 2099. PubMed ID: 33483521

    Nfonsam, L. E., Cano, C., Mudge, J., Schilkey, F. D. and Curtiss, J. (2012). Analysis of the transcriptomes downstream of Eyeless and the Hedgehog, Decapentaplegic and Notch signaling pathways in Drosophila melanogaster. PLoS One 7(8): e44583. PubMed ID: 22952997

    O'Keefe, D. D., Gonzalez-Nino, E., Edgar, B. A. and Curtiss, J. (2012). Discontinuities in Rap1 activity determine epithelial cell morphology within the developing wing of Drosophila. Dev Biol 369(2): 223-234. PubMed ID: 22776378

  • James Curtsinger Department of Ecology, Evolution and Behavior, University of Minnesota, Saint Paul
    Curtsinger, J. W. (2013). Late-life fecundity plateaus in Drosophila melanogaster can be explained by variation in reproductive life spans. Exp Gerontol. PubMed ID: 24012993

    Khazaeli, A. A. and Curtsinger, J. W. (2013). Pleiotropy and life history evolution in Drosophila melanogaster: uncoupling life span and early fecundity. J Gerontol A Biol Sci Med Sci 68: 546-553. PubMed ID: 23160365

    Khazaeli, A. A. and Curtsinger, J. W. (2010). Life history variation in an artificially selected population of Drosophila melanogaster: pleiotropy, superflies, and age-specific adaptation. Evolution 64: 3409-3416. PubMed ID: 20874737

  • Olivier Cuvier Laboratoire de Biologie, Moléculaire Eucaryote, Universite Toulouse
    Mourad, R., Li, L. and Cuvier, O. (2017). Uncovering direct and indirect molecular determinants of chromatin loops using a computational integrative approach. PLoS Comput Biol 13(5): e1005538. PubMed ID: 28542178

    Mourad, R. and Cuvier, O. (2016). Computational identification of genomic features that influence 3D chromatin domain formation. PLoS Comput Biol 12: e1004908. PubMed ID: 27203237

    Lhoumaud, P., Hennion, M., Gamot, A., Cuddapah, S., Queille, S., Liang, J., Micas, G., Morillon, P., Urbach, S., Bouchez, O., Severac, D., Emberly, E., Zhao, K. and Cuvier, O. (2014). Insulators recruit histone methyltransferase dMes4 to regulate chromatin of flanking genes. EMBO J. PubMed ID: 24916307

    Liang, J., Lacroix, L., Gamot, A., Cuddapah, S., Queille, S., Lhoumaud, P., Lepetit, P., Martin, P. G., Vogelmann, J., Court, F., Hennion, M., Micas, G., Urbach, S., Bouchez, O., Nollmann, M., Zhao, K., Emberly, E. and Cuvier, O. (2014). Chromatin Immunoprecipitation Indirect Peaks Highlight Long-Range Interactions of Insulator Proteins and Pol II Pausing. Mol Cell. PubMed ID: 24486021

    Jiang, N., Emberly, E., Cuvier, O. and Hart, C. M. (2009). Genome-wide mapping of boundary element-associated factor (BEAF) binding sites in Drosophila melanogaster links BEAF to transcription. Mol Cell Biol 29: 3556-3568. PubMed ID: 19380483

  • Benjamin Czech, Cancer Research UK Cambridge Institute, University of Cambridge
    Munafo, M., Lawless, V. R., Passera, A., MacMillan, S., Bornelov, S., Haussmann, I. U., Soller, M., Hannon, G. J. and Czech, B. (2021). Channel nuclear pore complex subunits are required for transposon silencing in Drosophila. Elife 10. PubMed ID: 33856346

    Eastwood, E. L., Jara, K. A., Bornelov, S., Munafo, M., Frantzis, V., Kneuss, E., Barbar, E. J., Czech, B. and Hannon, G. J. (2021). Dimerisation of the PICTS complex via LC8/Cut-up drives co-transcriptional transposon silencing in Drosophila. Elife 10. PubMed ID: 33538693

    Kneuss, E., Munafo, M., Eastwood, E. L., Deumer, U. S., Preall, J. B., Hannon, G. J. and Czech, B. (2019). Specialization of the Drosophila nuclear export family protein Nxf3 for piRNA precursor export. Genes Dev 33(17-18): 1208-1220. PubMed ID: 31416967

    Fagegaltier, D., Falciatori, I., Czech, B., Castel, S., Perrimon, N., Simcox, A. and Hannon, G. J. (2016). Oncogenic transformation of Drosophila somatic cells induces a functional piRNA pathway. Genes Dev 30(14): 1623-1635. PubMed ID: 27474441


  • Return to The Interactive Fly