Department of Biology

Michael Rosbash

Michael Rosbash, Biology faculty member, Brandeis UniversityProfessor of Biology and Investigator, Howard Hughes Medical Institute

Research Description

Circadian Rhythms, Behavior, and Gene Expression

We began our studies of Drosophila circadian rhythms almost 30 years ago, as a collaboration between my lab and that of my friend and Brandeis colleague Jeff Hall. Our more short-term goal at that time was to clone the period locus, and our more long-term goal was to define the machinery that underlies the almost ubiquitous process of circadian rhythmicity. Our entrée into this problem was the period gene (per) of Drosophila melanogaster, discovered more than 10 years earlier in pioneering behavioral genetic experiments by Ronald Konopka and Seymour Benzer. Our cloning and gene rescue efforts were published in 1984, but it wasn’t until 1990 that some mechanistic understanding of circadian rhythmicity was achieved. Paul Hardin, a post-doc in my lab, discovered that per mRNA as well as its encoded protein (PER) undergoes fluctuations in level during the circadian cycle. These observations and others indicated that PER is important for a negative-feedback loop of gene expression and that transcription of the per gene itself is affected. As PER was shown to be nuclear, we proposed that it rather directly inhibits the transcription of its own mRNA. Temporally controlled negative feedback at the transcriptional level is now an accepted feature of circadian timekeeping in mammals, plants, Neurospora, and even cyanobacteria. Moreover, PER as well as the many other clock components defined genetically over the past 1015 years are largely conserved and perform similar functions in the mammalian clock. This indicates that the machinery as well as the principles of the Drosophila clock is widely conserved in animals.

Although transcription factors and transcriptional regulation play key roles in circadian rhythmicity, post-transcriptional regulation has also received prominent attention. This is due to the potent effect of kinase mutants on Drosophila and mammalian timekeeping as well as some remarkable results from the cyanobacterial circadian system. In addition, very recent results indicate that mammals have a circadian clock that functions in the absence of transcription and translation. Nonetheless, our results on the first goal mentioned above continue to reinforce the importance of transcriptional regulation. For example, only a few fly genes have been shown to affect circadian timing when their dose is manipulated (1 gene instead of the normal 2; or 1 extra gene, 3 instead of 2), and these are all transcription factors.

Our current circadian work has three major goals: (1) to understand in mechanistic detail how the Drosophila circadian timing occurs; (2) to understand how circadian gene expression regulation takes place; (2) to understand the neural circuit(s) relevant to circadian timekeeping within the fruit fly brain and the functions of individual circadian neurons. There is also some non-circadian work that is focused more on basic aspects of gene expression regulation.

We are using a variety of biochemical and genetic approaches to further dissect the transcriptional and post-transcriptional regulatory mechanisms that have an impact on the clock. Many of these new strategies are designed to identify new clock genes and simultaneously to identify new regulatory mechanisms of interest. One current approach is that of ChIP-tiling arrays or ChIP-seq, i.e., chromatin immunoprecipitation followed by tiling or by high-throughput sequencing. We have carried out circadian time course experiments, 6 time points/day, with antibodies against CLK, PER and RNA Pol II. The results indicate cyclical binding of CLK and PER to an enormous number of potential direct target genes. Some of these encode proteins that likely contribute to transcriptional regulation, whereas others have potential roles in the post-transcriptional regulation of circadian function. This suggests that the core transcriptional feedback loop may be upstream of some modes of post-transcriptional regulation. One attractive explanation for the large number of direct target genes is the heterogeneity of the head tissue used for these kinds of experiments in Drosophila. Indeed, we have some evidence that the circadian machinery accesses different direct target genes in different cells, and uses different transcriptional start sites in these different locations.

We are also using high-throughput sequencing to address old as well as new circadian issues. For example, how extensive are mRNA circadian oscillations in fly heads? This question has been addressed in many labs with Affymetrix arrays, and we are now using high-throughput sequencing to assess mRNA cycling. This technology is also being used to address how much of this is truly transcriptional in origin, by comparing the mRNA cycling with nascent RNA cycling, as assessed by high throughput sequencing. Another new question is, are there microRNAs (miRNAs) that undergo circadian oscillations in fly heads? The answer is yes, and some of these have impressive cycling amplitudes. This indicates that they must have short half-lives. This has implications for the metabolism or miRNAs, which are usually very stable; it’s not known how their degradation is regulated. The observation also suggests that translational regulation makes important contributions to rhythms. Indeed, recent results show that the abundant miRNA bantam regulates the translation of the important circadian transcription factor CLK, and we have evidence that the cycling miRNAs we have recently identified also impact in a circadian manner the translation of specific target genes of physiological interest. More generally, the circadian regulation of translation is an emerging interest.

Our lab has also focused these past few years on various brain-neuronal aspects of Drosophila rhythms. There are seven anatomically distinct neuronal groups, comprising in total about 75 cells on each side of the adult fly brain; they express high levels of clock genes and usually undergo robust circadian oscillations of clock gene mRNA and protein in synchrony. One neuronal group, the small ventral-lateral neurons (s-LNvs), controls the characteristic morning activity peak of the insect activity pattern, and another controls the evening peak. In addition, the morning group is the master pacemaker in constant darkness. We are interested in generating a more sophisticated view of how these circadian neurons interact with each other and what is the relationship between the gene expression machinery and neuronal firing. For example, do the morning cells fire at a different time than the evening cells, or is the distinction between the two cell groups made at the level of output gene expression? Although the core clock genes are highly enriched in all brain circadian neurons, many mRNAs appear to be enriched in a circadian neuron-specific manner. As noted above, this distinction may provide a link to the distinct functions of different neuronal subgroups. mRNAs differentially expressed within specific neuron subsets should also provide invaluable tools for manipulating specific circadian neurons without affecting others. We are therefore interested in extending neuronal profiling, both to other neuronal groups within the fly brain and to high throughput sequencing analysis.

A related interest is how gene expression responds to different environmental stimuli, and the one of most interest currently is light. In addition to an important and general role in circadian entrainment and phase resetting, light appears to have specific effects on different circadian neuronal subsets: for example, one set of circadian neurons is directly light-sensitive to lights-on (dawn), whereas another appears light-sensitive to lights-off (dusk). Intriguingly, the dawn cells are arousal cells, whereas the lights-off cells appear to promote sleep, as one might expect for a diurnal animal. Sleep is yet another area of interest, and we are trying to identify additional groups of brain neurons with a major impact on sleep. Current results focus attention outside of the circadian network. By combining these different sleep-relevant neuronal groups with different in vivo approaches to manipulate neuronal function, we are trying to gain insight into the circuitry and rules that underlie sleep regulation.

Lastly, high-throughput sequencing is being used to address the issue of splicing and post-transcriptional regulation more generally. For example, we have been interested for many years in the extent to which splicing takes place prior to 3'-end formation. Although we have traditionally studied this question in yeast, we are now addressing it in Drosophila. To this end, we are trying to measure at a genomic level how much nascent splicing takes place, by sequencing nascent RNA of Drosophila tissue culture cells. Are there some general rules, and are there then exceptional genes? We are using similar approaches to address the circadian regulation of splicing in flies: how much nascent splicing takes place in flies, and is there a relationship to circadian rhythms and alternate splicing? We are currently in a descriptive phase but hope to move on shortly to more mechanistic questions: for example, how does the circadian regulation of splicing take place, and do any of these splicing events make a contribution to core circadian timekeeping? Are there other aspects of post-transcriptional regulation under circadian control?

Selected Publications

  • Rosbash M. Circadian Rhythms and the Transcriptional Feedback Loop (Nobel Lecture)*. Angew Chem Int Ed Engl. 2021 Feb 26. doi: 10.1002/anie.202015199. Epub ahead of print. PMID: 33634573.
  • Ma D, Przybylski D, Abruzzi KC, Schlichting M, Li Q, Long X, Rosbash M. A transcriptomic taxonomy of Drosophila circadian neurons around the clock. Elife. 2021 Jan 13;10:e63056. doi: 10.7554/eLife.63056. PMID: 33438579; PMCID: PMC7837698.
  • Kula-Eversole E, Lee DH, Samba I, Yildirim E, Levine DC, Hong HK, Lear BC, Bass J, Rosbash M, Allada R. Phosphatase of Regenerating Liver-1 Selectively Times Circadian Behavior in Darkness via Function in PDF Neurons and Dephosphorylation of TIMELESS. Curr Biol. 2021 Jan 11;31(1):138-149.e5. doi: 10.1016/j.cub.2020.10.013. 
  • Vogt K, Zimmerman DM, Schlichting M, Hernandez-Nunez L, Qin S, Malacon K, Rosbash M, Pehlevan C, Cardona A, Samuel ADT. Internal state configures olfactory behavior and early sensory processing in Drosophila larvae. Sci Adv. 2021 Jan 1;7(1):eabd6900. doi: 10.1126/sciadv.abd6900. 
  • Rosbash M. Metformin treatment of the C9orf72 ALS/FTD mouse: Almost too good for words. Proc Natl Acad Sci U S A. 2020 Aug 18;117(33):19627-19628. doi: 10.1073/pnas.2012363117. 
  • Jin H, Xu W, Rahman R, Na D, Fieldsend A, Song W, Liu S, Li C, Rosbash M. TRIBE editing reveals specific mRNA targets of eIF4E-BP in Drosophila and in mammals. Sci Adv. 2020 Aug 12;6(33):eabb8771. doi: 10.1126/sciadv.abb8771. 
  • MS2-TRIBE Evaluates Both Protein-RNA Interactions and Nuclear Organization of Transcription by RNA Editing. Biswas J, Rahman R, Gupta V, Rosbash M, Singer RH. iScience. 2020 Jun 28;23(7):101318. doi: 10.1016/j.isci.2020.101318. 
  • TDP-43 dysfunction restricts dendritic complexity by inhibiting CREB activation and altering gene expression. Herzog JJ, Xu W, Deshpande M, Rahman R, Suib H, Rodal AA, Rosbash M, Paradis S. Proc Natl Acad Sci U S A. 2020 May 26;117(21):11760-11769. doi: 10.1073/pnas.1917038117. Epub 2020 May 11. 
  • Video Recording Can Conveniently Assay Mosquito Locomotor Activity. Araujo MDS, Guo F, Rosbash M. Sci Rep. 2020 Mar 19;10(1):4994. doi: 10.1038/s41598-020-61733-5. 
  • A Serotonin-Modulated Circuit Controls Sleep Architecture to Regulate Cognitive Function Independent of Total Sleep in Drosophila. Liu C, Meng Z, Wiggin TD, Yu J, Reed ML, Guo F, Zhang Y, Rosbash M, Griffith LC. Curr Biol. 2019 Nov 4;29(21):3635-3646.e5. doi: 10.1016/j.cub.2019.08.079. Epub 2019 Oct 24. PMID: 31668619
  • Neuron-specific knockouts indicate the importance of network communication to Drosophila rhythmicity. Schlichting M, Díaz MM, Xin J, Rosbash M. Elife. 2019 Oct 15;8:e48301. doi: 0.7554/eLife.48301. PMID: 31613223
  • Light-Mediated Circuit Switching in the Drosophila Neuronal Clock Network. Schlichting M, Weidner P, Diaz M, Menegazzi P, Dalla Benetta E, Helfrich-Förster C, Rosbash M. Curr Biol. 2019 Oct 7;29(19):3266-3276.e3. doi: 10.1016/j.cub.2019.08.033. Epub 2019 Sep 26. PMID: 31564496
  • Medicine in the Fourth Dimension. Cederroth CR, Albrecht U, Bass J, Brown SA, Dyhrfjeld-Johnsen J, Gachon F, Green CB, Hastings MH, Helfrich-Förster C, Hogenesch JB, Lévi F, Loudon A, Lundkvist GB, Meijer JH, Rosbash M, Takahashi JS, Young M, Canlon B. Cell Metab. 2019 Aug 6;30(2):238-250. doi: 10.1016/j.cmet.2019.06.019. PMID: 31390550
  • A Distinct Visual Pathway Mediates High-Intensity Light Adaptation of the Circadian Clock in Drosophila. Schlichting M, Menegazzi P, Rosbash M, Helfrich-Förster C. J Neurosci. 2019 Feb 27;39(9):1621-1630.
  • Allatostatin-C/AstC-R2 Is a Novel Pathway to Modulate the Circadian Activity Pattern in Drosophila. Díaz MM, Schlichting M, Abruzzi KC, Long X, Rosbash M. Curr Biol. 2019 Jan 7;29(1):13-22.e3. doi: 10.1016/j.cub.2018.11.005.
  • NonA and CPX Link the Circadian Clockwork to Locomotor Activity in Drosophila. Luo W, Guo F, McMahon A, Couvertier S, Jin H, Diaz M, Fieldsend A, Weerapana E, Rosbash M. Neuron. 2018 Jul 24. pii: S0896-6273(18)30541-5. doi: 10.1016/j.neuron.2018.07.001.
  • Identification of RNA-binding protein targets with HyperTRIBE. Rahman R, Xu W, Jin H, Rosbash M. Nat Protoc. 2018 Aug;13(8):1829-1849. doi: 10.1038/s41596-018-0020-y.
  • Striking circadian neuron diversity and cycling of Drosophila alternative splicing. Wang Q, Abruzzi KC, Rosbash M, Rio DC. Elife. 2018 Jun 4;7. pii: e35618. doi: 10.7554/eLife.35618.
  • Mechanistic implications of enhanced editing by a HyperTRIBE RNA-binding protein. Xu W, Rahman R, Rosbash M. RNA. 2018 Feb;24(2):173-182. doi: 10.1261/rna.064691.117.
  • A Circadian Output Circuit Controls Sleep-Wake Arousal in Drosophila. Guo F, Holla M, Díaz MM, Rosbash M. Neuron. 2018 Nov 7;100(3):624-635.e4. doi: 10.1016/j.neuron.2018.09.002.
  • Life Is an N of 1. Rosbash M. Cell. 2017 Nov 30;171(6):1241-1245.
  • Guidelines for Genome-Scale Analysis of Biological Rhythms. Hughes ME, Abruzzi KC, Allada R, Anafi R, Arpat AB, Asher G, Baldi P, de Bekker C, Bell-Pedersen D, Blau J, Brown S, Ceriani MF, Chen Z, Chiu JC, Cox J, Crowell AM, DeBruyne JP, Dijk DJ, DiTacchio L, Doyle FJ, Duffield GE, Dunlap JC, Eckel-Mahan K, Esser KA, FitzGerald GA, Forger DB, Francey LJ, Fu YH, Gachon F, Gatfield D, de Goede P, Golden SS, Green C, Harer J, Harmer S, Haspel J, Hastings MH, Herzel H, Herzog ED, Hoffmann C, Hong C, Hughey JJ, Hurley JM, de la Iglesia HO, Johnson C, Kay SA, Koike N, Kornacker K, Kramer A, Lamia K, Leise T, Lewis SA, Li J, Li X, Liu AC, Loros JJ, Martino TA, Menet JS, Merrow M, Millar AJ, Mockler T, Naef F, Nagoshi E, Nitabach MN, Olmedo M, Nusinow DA, Ptáček LJ, Rand D, Reddy AB, Robles MS, Roenneberg T, Rosbash M, Ruben MD, Rund SSC, Sancar A, Sassone-Corsi P, Sehgal A, Sherrill-Mix S, Skene DJ, Storch KF, Takahashi JS, Ueda HR, Wang H, Weitz C, Westermark PO, Wijnen H, Xu Y, Wu G, Yoo SH, Young M, Zhang EE, Zielinski T, Hogenesch JB. J Biol Rhythms. 2017 Oct;32(5):380-393.
  • Temporal calcium profiling of specific circadian neurons in freely moving flies. Guo F, Chen X, Rosbash M. Proc Natl Acad Sci U S A. 2017 Oct 10;114(41):E8780-E8787.
  • A 50-Year Personal Journey: Location, Gene Expression, and Circadian Rhythms. Rosbash M. Cold Spring Harb Perspect Biol. 2017 Jun 9. pii: a032516. doi: 10.1101/cshperspect.a032516.
  • MicroRNA-92a is a circadian modulator of neuronal excitability in Drosophila. Chen X, Rosbash M. Nature Communications 2017 Mar 9;8:14707.
  • Kim EZ, Vienne J, Rosbash M and Griffith LC (2017). "Non-reciprocal homeostatic compensation in Drosophila potassium channel mutants." J Neurophysiol: jn 00002 02017.
  • Richhariya S, Jayakumar S, Abruzzi K, Rosbash M and Hasan G (2017). "A pupal transcriptomic screen identifies Ral as a target of store-operated calcium entry in Drosophila neurons." Sci Rep 7: 42586.
  • Sabado V, Vienne L, Nunes JM , Rosbash M and Nagoshi E. (2017). "Fluorescence circadian imaging reveals a PDF-dependent transcriptional regulation of the Drosophila molecular clock." Sci Rep 7: 41560.
  • Abruzzi KC, Zadina A, Luo W, Wiyanto E, Rahman R, Guo F, Shafer O and Rosbash M (2017). "RNA-seq analysis of Drosophila clock and non-clock neurons reveals neuron-specific cycling and novel candidate neuropeptides." PLoS Genet 13(2): e1006613.
  • Chen, X. and M. Rosbash (2017). "MicroRNA-92a is a circadian modulator of neuronal excitability in Drosophila." Nat Commun 8: 14707.
  • Guo, F., J. Yu, H. J. Jung, K. C. Abruzzi, W. Luo, L. C. Griffith and M. Rosbash (2016). "Circadian neuron feedback controls the Drosophila sleep--activity profile." Nature 536(7616): 292-297.
  • Chen X, Rahman R, Guo F and Rosbash M (2016). "Genome-wide identification of neuronal activity-regulated genes in Drosophila." Elife 5.
  • McMahon, A. C. and M. Rosbash (2016). "Promiscuous or discriminating: Has the favored mRNA target of Fragile X Mental Retardation Protein been overlooked?" Proc Natl Acad Sci U S A 113(26): 7009-7011.
  • Vienne J, Spann R, Guo F, Rosbash M (2016). "Age-Related Reduction of Recovery Sleep and Arousal Threshold in Drosophila." Sleep. 2016 Jun 9. pii: sp-00707-15.
  • McMahon AC, Rahman R, Jin H, Shen JL, Fieldsend A, Luo W and Rosbash M (2016). "TRIBE: Hijacking an RNA-Editing Enzyme to Identify Cell-Specific Targets of RNA-Binding Proteins." Cell 2016 Apr 21;165(3):742-53.
  • Chen X and Rosbash M (2016). "mir-276a strengthens Drosophila circadian rhythms by regulating timeless expression." Proc Natl Acad Sci U S A. 2016 May 24;113(21):E2965-72.
  • Abruzzi K, Chen X, Nagoshi E, Zadina A and Rosbash M (2015). "RNA-seq profiling of small numbers of Drosophila neurons." Methods Enzymol551: 369-386.
  • Hadzic T., Park D, Abruzzi KC, Yang L, Trigg JS, Rohs R, Rosbash M and Taghert PH (2015). "Genome-wide features of neuroendocrine regulation in Drosophila by the basic helix-loop-helix transcription factor DIMMED." Nucleic Acids Res. 2015 Feb 27;43(4):2199-215.
  • Lerner I, Bartok O, Wolfson V, Menet JS, Weissbein U, Afik S, Haimovich D, Gafni C, Friedman N, Rosbash M and Kadener S (2015). "Clk post-transcriptional control denoises circadian transcription both temporally and spatially." Nat Commun 015 May 8;6:7056.
  • Rosbash M. (2015). "We'll always have RNA." RNA 2015 Apr;21(4):546-7.
  • Guo F, Cerullo I, Chen X, Rosbash M (2014). "PDF neuron firing phase-shifts key circadian activity neurons in Drosophila." Elife. June 17:e02780.
  • Li Y, Guo F, Shen J, Rosbash M (2014). "PDF and cAMP enhance PER stability in Drosophila clock neurons." Proc.Natl.Acad.Sci. USA 111:E1284-90.
  • Menet JS, Pescatore S, Rosbash M (2014). "CLOCK:BMAL1 is a pioneer-like transcription factor." Genes Dev. 28:8-13.
  • Salisbury JP, Boggio KJ, Hsu YW, Quijada J, Sivachenko A, Gloeckner G, Kowalski PJ, Easterling ML, Rosbash M, Agar JN (2013) "A rapid MALDI-TOF mass spectrometry workflow for Drosophila melanogaster differential neuropeptidomics." Mol. Brain. 6:60.
  • Shang Y, Donelson NC, Vecsey CG, Guo F, Rosbash M, Griffith LC (2013). "Short neuropeptide F is a sleep-promoting inhibitory modulator." Neuron 80:171-83.
  • Sivachenko A, Li Y, Abruzzi KC, Rosbash M (2013). "The transcription factor Mef2 links the Drosophila core clock to Fas2, neuronal morphology, and circadian behavior." Neuron 79:281092.
  • Li Y, Rosbash M (2013). "Accelerated degradation of perS protein provides insight into light-mediated phase shifting." J. Biol. Rhythms. 28:171-82.
  • Perrat PN, DasGupta S, Wang J, Theurkauf W, Weng Z, Rosbash M, Waddell S (2013). "Transposition-driven genomic heterogeneit in the Drosophila brain." Science. 340:91-5.
  • Rodriguez J, Tang CH, Khodor YL, Vodala S, Menet JS, Rosbash M (2013). "Nascent-Seq analysis of Drosophila cycling gene expression." Proc. Natl. Acad. Sci. U.S.A. 110:E275-84.
  • Luo W, Tang CH, Abruzzi KC, Rodriguez J, Pescatore S, Rosbash M (2012). "CLOCK deubiquitylation by USP8 inhibits CLK/CYC transcription in Drosophila." Genes Dev. 26:2536-2549.
  • Menet JS, Rodriguez J, Abruzzi KC, Rosbash M (2012). "Nascent-Seq reveals novel features of mouse circadian transcriptional regulation." eLife. 1:e00011.
  • Vodala S, Pescatore S, Rodriguez J, Buescher M, Chen YW, Weng R, Cohen SM, Rosbash M (2012). "The Oscillating miRNA 959-964 Cluster Impacts Drosophila Feeding Time and Other Circadian Outputs." Cell Metab. 16:601-612.
  • Khodor YL, Menet JS, Tolan M, Rosbash M (2012). "Cotranscriptional splicing efficiency differs dramatically between Drosophila and mouse." RNA. 18:2174-2186.
  • Bradley S, Narayanan S, Rosbash M (2012). "NAT1/DAP5/p97 and Atypical Translational Control in the Drosophila Circadian Oscillator." Genetics. 192:943-957.
  • Choi C, Cao G, Tanenhaus AK, McCarthy EV, Jung M, Schleyer W, Shang Y, Rosbash M, Yin JC, Nitabach MN (2012). "Autoreceptor Control of Peptide/Neurotransmitter Corelease from PDF Neurons Determines Allocation of Circadian Activity in Drosophila." Cell Rep. 2:332-344.
  • Rodriguez J, Menet JS, Rosbash M (2012). "Nascent-seq indicates widespread cotranscriptional RNA editing in Drosophila." Mol Cell. 47:27-37.