- -

Different Mechanisms Confer Gradual Control and Memory at Nutrient- and Stress-Regulated Genes in Yeast

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

Different Mechanisms Confer Gradual Control and Memory at Nutrient- and Stress-Regulated Genes in Yeast

Mostrar el registro sencillo del ítem

Ficheros en el ítem

dc.contributor.author Rienzo, Alessandro es_ES
dc.contributor.author Poveda Huertes, Daniel es_ES
dc.contributor.author Aydin, Selcan es_ES
dc.contributor.author Buchler, Nicolas E. es_ES
dc.contributor.author Pascual-Ahuir Giner, María Desamparados es_ES
dc.contributor.author Proft, Markus Hans es_ES
dc.date.accessioned 2017-03-13T10:57:18Z
dc.date.available 2017-03-13T10:57:18Z
dc.date.issued 2015-11
dc.identifier.issn 0270-7306
dc.identifier.uri http://hdl.handle.net/10251/78703
dc.description.abstract Cells respond to environmental stimuli by fine-tuned regulation of gene expression. Here we investigated the dose-dependent modulation of gene expression at high temporal resolution in response to nutrient and stress signals in yeast. The GAL1 activity in cell populations is modulated in a well-defined range of galactose concentrations, correlating with a dynamic change of histone remodeling and RNA polymerase II (RNAPII) association. This behavior is the result of a heterogeneous induction delay caused by decreasing inducer concentrations across the population. Chromatin remodeling appears to be the basis for the dynamic GAL1 expression, because mutants with impaired histone dynamics show severely truncated dose-response profiles. In contrast, the GRE2 promoter operates like a rapid off/on switch in response to increasing osmotic stress, with almost constant expression rates and exclusively temporal regulation of histone remodeling and RNAPII occupancy. The Gal3 inducer and the Hog1 mitogen-activated protein (MAP) kinase seem to determine the different dose-response strategies at the two promoters. Accordingly, GAL1 becomes highly sensitive and dose independent if previously stimulated because of residual Gal3 levels, whereas GRE2 expression diminishes upon repeated stimulation due to acquired stress resistance. Our analysis reveals important differences in the way dynamic signals create dose-sensitive gene expression outputs. es_ES
dc.description.sponsorship This work was supported by grants from Ministerio de Economia y Competitividad (BFU2011-23326), Generalitat de Valencia (ACOMP2011/031), and the NIH Director's New Innovator Award (DP2 OD008654-01). Alessandro Rienzo was a recipient of a predoctoral FPI grant from Ministerio de Economia y Competitividad. en_EN
dc.language Inglés es_ES
dc.publisher American Society for Microbiology es_ES
dc.relation.ispartof Molecular and Cellular Biology es_ES
dc.rights Reserva de todos los derechos es_ES
dc.subject RNA-polymerase-II es_ES
dc.subject Graded transcriptional responses es_ES
dc.subject Saccharomyces cerevisiae es_ES
dc.subject In-vivo es_ES
dc.subject MAP kinase es_ES
dc.subject Expression program es_ES
dc.subject Nuclear periphery es_ES
dc.subject Oxidative stress es_ES
dc.subject Cellular memory es_ES
dc.subject Osmotic stress es_ES
dc.subject.classification BIOQUIMICA Y BIOLOGIA MOLECULAR es_ES
dc.title Different Mechanisms Confer Gradual Control and Memory at Nutrient- and Stress-Regulated Genes in Yeast es_ES
dc.type Artículo es_ES
dc.identifier.doi 10.1128/MCB.00729-15
dc.relation.projectID info:eu-repo/grantAgreement/MICINN//BFU2011-23326/ES/REGULACION DE LA CROMATINA Y DE LA ESTRUCTURA MITOCONDRIAL EN RESPUESTA A ESTRES OSMOTICO/ es_ES
dc.relation.projectID info:eu-repo/grantAgreement/NIH//DP2 OD008654-01/ es_ES
dc.relation.projectID info:eu-repo/grantAgreement/GVA//ACOMP%2F2011%2F031/ es_ES
dc.rights.accessRights Abierto es_ES
dc.contributor.affiliation Universitat Politècnica de València. Instituto Universitario Mixto de Biología Molecular y Celular de Plantas - Institut Universitari Mixt de Biologia Molecular i Cel·lular de Plantes es_ES
dc.contributor.affiliation Universitat Politècnica de València. Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural - Escola Tècnica Superior d'Enginyeria Agronòmica i del Medi Natural es_ES
dc.description.bibliographicCitation Rienzo, A.; Poveda Huertes, D.; Aydin, S.; Buchler, NE.; Pascual-Ahuir Giner, MD.; Proft, MH. (2015). Different Mechanisms Confer Gradual Control and Memory at Nutrient- and Stress-Regulated Genes in Yeast. Molecular and Cellular Biology. 35(21):3669-3683. https://doi.org/10.1128/MCB.00729-15 es_ES
dc.description.accrualMethod S es_ES
dc.relation.publisherversion http://dx.doi.org/10.1128/MCB.00729-15 es_ES
dc.description.upvformatpinicio 3669 es_ES
dc.description.upvformatpfin 3683 es_ES
dc.type.version info:eu-repo/semantics/publishedVersion es_ES
dc.description.volume 35 es_ES
dc.description.issue 21 es_ES
dc.relation.senia 303448 es_ES
dc.identifier.eissn 1098-5549
dc.identifier.pmcid PMC4589597 en_EN
dc.contributor.funder Ministerio de Ciencia e Innovación es_ES
dc.contributor.funder National Institutes of Health, EEUU es_ES
dc.contributor.funder Generalitat Valenciana es_ES
dc.description.references Gasch, A. P., & Werner-Washburne, M. (2002). The genomics of yeast responses to environmental stress and starvation. Functional & Integrative Genomics, 2(4-5), 181-192. doi:10.1007/s10142-002-0058-2 es_ES
dc.description.references Hahn, S., & Young, E. T. (2011). Transcriptional Regulation inSaccharomyces cerevisiae: Transcription Factor Regulation and Function, Mechanisms of Initiation, and Roles of Activators and Coactivators. Genetics, 189(3), 705-736. doi:10.1534/genetics.111.127019 es_ES
dc.description.references Dos Santos, S. C. (2012). Yeast toxicogenomics: genome-wide responses to chemical stresses with impact in environmental health, pharmacology, and biotechnology. Frontiers in Genetics, 3. doi:10.3389/fgene.2012.00063 es_ES
dc.description.references Gasch, A. P., Spellman, P. T., Kao, C. M., Carmel-Harel, O., Eisen, M. B., Storz, G., … Brown, P. O. (2000). Genomic Expression Programs in the Response of Yeast Cells to Environmental Changes. Molecular Biology of the Cell, 11(12), 4241-4257. doi:10.1091/mbc.11.12.4241 es_ES
dc.description.references Martínez-Montañés, F., Pascual-Ahuir, A., & Proft, M. (2010). Toward a Genomic View of the Gene Expression Program Regulated by Osmostress in Yeast. OMICS: A Journal of Integrative Biology, 14(6), 619-627. doi:10.1089/omi.2010.0046 es_ES
dc.description.references Morano, K. A., Grant, C. M., & Moye-Rowley, W. S. (2011). The Response to Heat Shock and Oxidative Stress in Saccharomyces cerevisiae. Genetics, 190(4), 1157-1195. doi:10.1534/genetics.111.128033 es_ES
dc.description.references De Nadal, E., Ammerer, G., & Posas, F. (2011). Controlling gene expression in response to stress. Nature Reviews Genetics, 12(12), 833-845. doi:10.1038/nrg3055 es_ES
dc.description.references Takahashi, S., & Pryciak, P. M. (2008). Membrane Localization of Scaffold Proteins Promotes Graded Signaling in the Yeast MAP Kinase Cascade. Current Biology, 18(16), 1184-1191. doi:10.1016/j.cub.2008.07.050 es_ES
dc.description.references Cai, L., Dalal, C. K., & Elowitz, M. B. (2008). Frequency-modulated nuclear localization bursts coordinate gene regulation. Nature, 455(7212), 485-490. doi:10.1038/nature07292 es_ES
dc.description.references Giorgetti, L., Siggers, T., Tiana, G., Caprara, G., Notarbartolo, S., Corona, T., … Natoli, G. (2010). Noncooperative Interactions between Transcription Factors and Clustered DNA Binding Sites Enable Graded Transcriptional Responses to Environmental Inputs. Molecular Cell, 37(3), 418-428. doi:10.1016/j.molcel.2010.01.016 es_ES
dc.description.references Hao, N., & O’Shea, E. K. (2011). Signal-dependent dynamics of transcription factor translocation controls gene expression. Nature Structural & Molecular Biology, 19(1), 31-39. doi:10.1038/nsmb.2192 es_ES
dc.description.references Stewart-Ornstein, J., Nelson, C., DeRisi, J., Weissman, J. S., & El-Samad, H. (2013). Msn2 Coordinates a Stoichiometric Gene Expression Program. Current Biology, 23(23), 2336-2345. doi:10.1016/j.cub.2013.09.043 es_ES
dc.description.references Hao, N., Budnik, B. A., Gunawardena, J., & O’Shea, E. K. (2013). Tunable Signal Processing Through Modular Control of Transcription Factor Translocation. Science, 339(6118), 460-464. doi:10.1126/science.1227299 es_ES
dc.description.references Lam, F. H., Steger, D. J., & O’Shea, E. K. (2008). Chromatin decouples promoter threshold from dynamic range. Nature, 453(7192), 246-250. doi:10.1038/nature06867 es_ES
dc.description.references Dolz-Edo, L., Rienzo, A., Poveda-Huertes, D., Pascual-Ahuir, A., & Proft, M. (2013). Deciphering Dynamic Dose Responses of Natural Promoters and Single cis Elements upon Osmotic and Oxidative Stress in Yeast. Molecular and Cellular Biology, 33(11), 2228-2240. doi:10.1128/mcb.00240-13 es_ES
dc.description.references Sellick, C. A., Campbell, R. N., & Reece, R. J. (2008). Chapter 3 Galactose Metabolism in Yeast—Structure and Regulation of the Leloir Pathway Enzymes and the Genes Encoding Them. International Review of Cell and Molecular Biology, 111-150. doi:10.1016/s1937-6448(08)01003-4 es_ES
dc.description.references Kumar, P. R., Yu, Y., Sternglanz, R., Johnston, S. A., & Joshua-Tor, L. (2008). NADP Regulates the Yeast GAL Induction System. Science, 319(5866), 1090-1092. doi:10.1126/science.1151903 es_ES
dc.description.references Thoden, J. B., Ryan, L. A., Reece, R. J., & Holden, H. M. (2008). The Interaction between an Acidic Transcriptional Activator and Its Inhibitor. Journal of Biological Chemistry, 283(44), 30266-30272. doi:10.1074/jbc.m805200200 es_ES
dc.description.references Egriboz, O., Jiang, F., & Hopper, J. E. (2011). Rapid GAL Gene Switch of Saccharomyces cerevisiae Depends on Nuclear Gal3, Not Nucleocytoplasmic Trafficking of Gal3 and Gal80. Genetics, 189(3), 825-836. doi:10.1534/genetics.111.131839 es_ES
dc.description.references Jiang, F., Frey, B. R., Evans, M. L., Friel, J. C., & Hopper, J. E. (2009). Gene Activation by Dissociation of an Inhibitor from a Transcriptional Activation Domain. Molecular and Cellular Biology, 29(20), 5604-5610. doi:10.1128/mcb.00632-09 es_ES
dc.description.references Lavy, T., Kumar, P. R., He, H., & Joshua-Tor, L. (2012). The Gal3p transducer of the GAL regulon interacts with the Gal80p repressor in its ligand-induced closed conformation. Genes & Development, 26(3), 294-303. doi:10.1101/gad.182691.111 es_ES
dc.description.references Bhaumik, S. R. (2001). SAGA is an essential in vivo target of the yeast acidic activator Gal4p. Genes & Development, 15(15), 1935-1945. doi:10.1101/gad.911401 es_ES
dc.description.references Brown, C. E. (2001). Recruitment of HAT Complexes by Direct Activator Interactions with the ATM-Related Tra1 Subunit. Science, 292(5525), 2333-2337. doi:10.1126/science.1060214 es_ES
dc.description.references Jeong, C.-J., Yang, S.-H., Xie, Y., Zhang, L., Johnston, S. A., & Kodadek, T. (2001). Evidence That Gal11 Protein Is a Target of the Gal4 Activation Domain in the Mediator†. Biochemistry, 40(31), 9421-9427. doi:10.1021/bi010011k es_ES
dc.description.references Koh, S. S., Ansari, A. Z., Ptashne, M., & Young, R. A. (1998). An Activator Target in the RNA Polymerase II Holoenzyme. Molecular Cell, 1(6), 895-904. doi:10.1016/s1097-2765(00)80088-x es_ES
dc.description.references Larschan, E. (2001). The S. cerevisiae SAGA complex functions in vivo as a coactivator for transcriptional activation by Gal4. Genes & Development, 15(15), 1946-1956. doi:10.1101/gad.911501 es_ES
dc.description.references Lemieux, K., & Gaudreau, L. (2004). Targeting of Swi/Snf to the yeast GAL1 UASG requires the Mediator, TAFIIs, and RNA polymerase II. The EMBO Journal, 23(20), 4040-4050. doi:10.1038/sj.emboj.7600416 es_ES
dc.description.references De Nadal, E., & Posas, F. (2009). Multilayered control of gene expression by stress-activated protein kinases. The EMBO Journal, 29(1), 4-13. doi:10.1038/emboj.2009.346 es_ES
dc.description.references Proft, M. (2001). Regulation of the Sko1 transcriptional repressor by the Hog1 MAP kinase in response to osmotic stress. The EMBO Journal, 20(5), 1123-1133. doi:10.1093/emboj/20.5.1123 es_ES
dc.description.references Proft, M., & Struhl, K. (2002). Hog1 Kinase Converts the Sko1-Cyc8-Tup1 Repressor Complex into an Activator that Recruits SAGA and SWI/SNF in Response to Osmotic Stress. Molecular Cell, 9(6), 1307-1317. doi:10.1016/s1097-2765(02)00557-9 es_ES
dc.description.references Brickner, D. G., Cajigas, I., Fondufe-Mittendorf, Y., Ahmed, S., Lee, P.-C., Widom, J., & Brickner, J. H. (2007). H2A.Z-Mediated Localization of Genes at the Nuclear Periphery Confers Epigenetic Memory of Previous Transcriptional State. PLoS Biology, 5(4), e81. doi:10.1371/journal.pbio.0050081 es_ES
dc.description.references Kundu, S., Horn, P. J., & Peterson, C. L. (2007). SWI/SNF is required for transcriptional memory at the yeast GAL gene cluster. Genes & Development, 21(8), 997-1004. doi:10.1101/gad.1506607 es_ES
dc.description.references Kundu, S., & Peterson, C. L. (2010). Dominant Role for Signal Transduction in the Transcriptional Memory of Yeast GAL Genes. Molecular and Cellular Biology, 30(10), 2330-2340. doi:10.1128/mcb.01675-09 es_ES
dc.description.references Zacharioudakis, I., Gligoris, T., & Tzamarias, D. (2007). A Yeast Catabolic Enzyme Controls Transcriptional Memory. Current Biology, 17(23), 2041-2046. doi:10.1016/j.cub.2007.10.044 es_ES
dc.description.references Winzeler, E. A. (1999). Functional Characterization of the S. cerevisiae Genome by Gene Deletion and Parallel Analysis. Science, 285(5429), 901-906. doi:10.1126/science.285.5429.901 es_ES
dc.description.references Ghaemmaghami, S., Huh, W.-K., Bower, K., Howson, R. W., Belle, A., Dephoure, N., … Weissman, J. S. (2003). Global analysis of protein expression in yeast. Nature, 425(6959), 737-741. doi:10.1038/nature02046 es_ES
dc.description.references Mason, P. B., & Struhl, K. (2003). The FACT Complex Travels with Elongating RNA Polymerase II and Is Important for the Fidelity of Transcriptional Initiation In Vivo. Molecular and Cellular Biology, 23(22), 8323-8333. doi:10.1128/mcb.23.22.8323-8333.2003 es_ES
dc.description.references Rienzo, A., Pascual-Ahuir, A., & Proft, M. (2012). The use of a real-time luciferase assay to quantify gene expression dynamics in the living yeast cell. Yeast, 29(6), 219-231. doi:10.1002/yea.2905 es_ES
dc.description.references Alberti, S., Gitler, A. D., & Lindquist, S. (2007). A suite of Gateway®cloning vectors for high-throughput genetic analysis inSaccharomyces cerevisiae. Yeast, 24(10), 913-919. doi:10.1002/yea.1502 es_ES
dc.description.references Mazo-Vargas, A., Park, H., Aydin, M., & Buchler, N. E. (2014). Measuring fast gene dynamics in single cells with time-lapse luminescence microscopy. Molecular Biology of the Cell, 25(22), 3699-3708. doi:10.1091/mbc.e14-07-1187 es_ES
dc.description.references Kuras, L., & Struhl, K. (1999). Binding of TBP to promoters in vivo is stimulated by activators and requires Pol II holoenzyme. Nature, 399(6736), 609-613. doi:10.1038/21239 es_ES
dc.description.references Kvarnström, M., Logg, K., Diez, A., Bodvard, K., & Käll, M. (2008). Image analysis algorithms for cell contour recognition in budding yeast. Optics Express, 16(17), 12943. doi:10.1364/oe.16.012943 es_ES
dc.description.references Acar, M., Becskei, A., & van Oudenaarden, A. (2005). Enhancement of cellular memory by reducing stochastic transitions. Nature, 435(7039), 228-232. doi:10.1038/nature03524 es_ES
dc.description.references Acar, M., Pando, B. F., Arnold, F. H., Elowitz, M. B., & van Oudenaarden, A. (2010). A General Mechanism for Network-Dosage Compensation in Gene Circuits. Science, 329(5999), 1656-1660. doi:10.1126/science.1190544 es_ES
dc.description.references Biggar, S. R. (2001). Cell signaling can direct either binary or graded transcriptional responses. The EMBO Journal, 20(12), 3167-3176. doi:10.1093/emboj/20.12.3167 es_ES
dc.description.references Gandhi, S. J., Zenklusen, D., Lionnet, T., & Singer, R. H. (2010). Transcription of functionally related constitutive genes is not coordinated. Nature Structural & Molecular Biology, 18(1), 27-34. doi:10.1038/nsmb.1934 es_ES
dc.description.references Abramczyk, D., Holden, S., Page, C. J., & Reece, R. J. (2011). Interplay of a Ligand Sensor and an Enzyme in Controlling Expression of the Saccharomyces cerevisiae GAL Genes. Eukaryotic Cell, 11(3), 334-342. doi:10.1128/ec.05294-11 es_ES
dc.description.references Ahmed, S., & Brickner, J. H. (2007). Regulation and epigenetic control of transcription at the nuclear periphery. Trends in Genetics, 23(8), 396-402. doi:10.1016/j.tig.2007.05.009 es_ES
dc.description.references Babazadeh, R., Adiels, C. B., Smedh, M., Petelenz-Kurdziel, E., Goksör, M., & Hohmann, S. (2013). Osmostress-Induced Cell Volume Loss Delays Yeast Hog1 Signaling by Limiting Diffusion Processes and by Hog1-Specific Effects. PLoS ONE, 8(11), e80901. doi:10.1371/journal.pone.0080901 es_ES
dc.description.references Miermont, A., Waharte, F., Hu, S., McClean, M. N., Bottani, S., Leon, S., & Hersen, P. (2013). Severe osmotic compression triggers a slowdown of intracellular signaling, which can be explained by molecular crowding. Proceedings of the National Academy of Sciences, 110(14), 5725-5730. doi:10.1073/pnas.1215367110 es_ES
dc.description.references Proft, M., & Struhl, K. (2004). MAP Kinase-Mediated Stress Relief that Precedes and Regulates the Timing of Transcriptional Induction. Cell, 118(3), 351-361. doi:10.1016/j.cell.2004.07.016 es_ES
dc.description.references Guan, Q., Haroon, S., Bravo, D. G., Will, J. L., & Gasch, A. P. (2012). Cellular Memory of Acquired Stress Resistance inSaccharomyces cerevisiae. Genetics, 192(2), 495-505. doi:10.1534/genetics.112.143016 es_ES
dc.description.references Pelet, S., Rudolf, F., Nadal-Ribelles, M., de Nadal, E., Posas, F., & Peter, M. (2011). Transient Activation of the HOG MAPK Pathway Regulates Bimodal Gene Expression. Science, 332(6030), 732-735. doi:10.1126/science.1198851 es_ES


Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del ítem