1
|
Hoffmann SA. YeastFab Cloning of Toxic Genes and Protein Expression Optimization in Yeast. Methods Mol Biol 2025; 2850:435-450. [PMID: 39363086 DOI: 10.1007/978-1-0716-4220-7_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
YeastFab is a Golden Gate-based cloning standard and parts repository. It is designed for modular, hierarchical assembly of transcription units and multi-gene assemblies for expression in Saccharomyces cerevisiae. This makes it a suitable toolbox to optimize the expression strength of heterologous genes in yeast. When cloning heterologous coding sequences into YeastFab vectors, in several cases we have observed toxicity to the cloning host Escherichia coli. The provided protocol details how to clone such toxic genes from multiple synthetic DNA fragments while adhering to the YeastFab standard. The presented cloning strategy includes a C-terminal FLAG tag that allows screening for constructs with a desired protein expression in yeast by western blot. The design allows scarlessly removing the tag through a Golden Gate reaction to facilitate cloning of expression constructs with the native, untagged transgene.
Collapse
Affiliation(s)
- Stefan A Hoffmann
- Systems and Synthetic Biology, Wageningen University & Research, Wageningen, Netherlands.
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Marchando P, Hu G, Sun Y, Drubin DG. Polarized exocytosis and anionic phospholipid species implicated in the initiation of clathrin-mediated endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617284. [PMID: 39416225 PMCID: PMC11482806 DOI: 10.1101/2024.10.08.617284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Understanding of the mechanisms that initiate clathrin-mediated endocytosis (CME) is incomplete. Recent studies in budding yeast identified the endocytic adaptor protein Yap1801/Yap1802 (budding yeast AP180) as a key CME factor that promotes CME initiation in daughter cells during polarized growth, but how Yap1801/2 is recruited preferentially to the plasma membrane of daughter cells is not clear. The only known cargos for Yap1801/2 in yeast are the synaptobrevins Snc1 and Snc2, which act as v-SNARES for exocytic vesicles arriving at the plasma membrane and are essential for polarized cell growth. In this study, we analyze the spatiotemporal dynamics of functional, fluorescently-tagged Snc1/2 expressed from their endogenous loci and provide evidence that, in concert with anionic phospholipids, Snc1/2 recruit Yap1801/2 preferentially to growing daughter cells. These findings suggest that the coincidence of anionic phospholipids and Snc1/2 facilitates CME initiation in growing daughter cells and directly links polarized CME to polarized secretion.
Collapse
|
3
|
Xu M, Vallières C, Finnis C, Winzer K, Avery SV. Exploiting phenotypic heterogeneity to improve production of glutathione by yeast. Microb Cell Fact 2024; 23:267. [PMID: 39375675 PMCID: PMC11457410 DOI: 10.1186/s12934-024-02536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Gene expression noise (variation in gene expression among individual cells of a genetically uniform cell population) can result in heterogenous metabolite production by industrial microorganisms, with cultures containing both low- and high-producing cells. The presence of low-producing individuals may be a factor limiting the potential for high yields. This study tested the hypothesis that low-producing variants in yeast cell populations can be continuously counter-selected, to increase net production of glutathione (GSH) as an exemplar product. RESULTS A counter-selection system was engineered in Saccharomyces cerevisiae based on the known feedback inhibition of gamma-glutamylcysteine synthetase (GSH1) gene expression, which is rate limiting for GSH synthesis: the GSH1 ORF and the counter-selectable marker GAP1 were expressed under control of the TEF1 and GSH-regulated GSH1 promoters, respectively. An 18% increase in the mean cellular GSH level was achieved in cultures of the engineered strain supplemented with D-histidine to counter-select cells with high GAP1 expression (i.e. low GSH-producing cells). The phenotype was non-heritable and did not arise from a generic response to D-histidine, unlike that with certain other test-constructs prepared with alternative markers. CONCLUSIONS The results corroborate that the system developed here improves GSH production by targeting low-producing cells. This supports the potential for exploiting end-product/promoter interactions to enrich high-producing cells in phenotypically heterogeneous populations, in order to improve metabolite production by yeast.
Collapse
Affiliation(s)
- Mingzhi Xu
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Cindy Vallières
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Chris Finnis
- Phenotypeca, BioCity Nottingham, Nottingham, NG1 1GF, UK
| | - Klaus Winzer
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Simon V Avery
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
4
|
Sejour R, Leatherwood J, Yurovsky A, Futcher B. Enrichment of rare codons at 5' ends of genes is a spandrel caused by evolutionary sequence turnover and does not improve translation. eLife 2024; 12:RP89656. [PMID: 39008347 PMCID: PMC11249729 DOI: 10.7554/elife.89656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Previously, Tuller et al. found that the first 30-50 codons of the genes of yeast and other eukaryotes are slightly enriched for rare codons. They argued that this slowed translation, and was adaptive because it queued ribosomes to prevent collisions. Today, the translational speeds of different codons are known, and indeed rare codons are translated slowly. We re-examined this 5' slow translation 'ramp.' We confirm that 5' regions are slightly enriched for rare codons; in addition, they are depleted for downstream Start codons (which are fast), with both effects contributing to slow 5' translation. However, we also find that the 5' (and 3') ends of yeast genes are poorly conserved in evolution, suggesting that they are unstable and turnover relatively rapidly. When a new 5' end forms de novo, it is likely to include codons that would otherwise be rare. Because evolution has had a relatively short time to select against these codons, 5' ends are typically slightly enriched for rare, slow codons. Opposite to the expectation of Tuller et al., we show by direct experiment that genes with slowly translated codons at the 5' end are expressed relatively poorly, and that substituting faster synonymous codons improves expression. Direct experiment shows that slow codons do not prevent downstream ribosome collisions. Further informatic studies suggest that for natural genes, slow 5' ends are correlated with poor gene expression, opposite to the expectation of Tuller et al. Thus, we conclude that slow 5' translation is a 'spandrel'--a non-adaptive consequence of something else, in this case, the turnover of 5' ends in evolution, and it does not improve translation.
Collapse
Affiliation(s)
- Richard Sejour
- Department of Pharmacological Sciences, Stony Brook UniversityStony BrookUnited States
| | - Janet Leatherwood
- Department of Microbiology and Immunology, Stony Brook UniversityStony BrookUnited States
| | - Alisa Yurovsky
- Department of Biomedical Informatics, Stony Brook UniversityStony BrookUnited States
| | - Bruce Futcher
- Department of Microbiology and Immunology, Stony Brook UniversityStony BrookUnited States
| |
Collapse
|
5
|
Pons C, van Leeuwen J. Meta-analysis of dispensable essential genes and their interactions with bypass suppressors. Life Sci Alliance 2024; 7:e202302192. [PMID: 37918966 PMCID: PMC10622647 DOI: 10.26508/lsa.202302192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023] Open
Abstract
Genes have been historically classified as essential or non-essential based on their requirement for viability. However, genomic mutations can sometimes bypass the requirement for an essential gene, challenging the binary classification of gene essentiality. Such dispensable essential genes represent a valuable model for understanding the incomplete penetrance of loss-of-function mutations often observed in natural populations. Here, we compiled data from multiple studies on essential gene dispensability in Saccharomyces cerevisiae to comprehensively characterize these genes. In analyses spanning different evolutionary timescales, dispensable essential genes exhibited distinct phylogenetic properties compared with other essential and non-essential genes. Integration of interactions with suppressor genes that can bypass the gene essentiality revealed the high functional modularity of the bypass suppression network. Furthermore, dispensable essential and bypass suppressor gene pairs reflected simultaneous changes in the mutational landscape of S. cerevisiae strains. Importantly, species in which dispensable essential genes were non-essential tended to carry bypass suppressor mutations in their genomes. Overall, our study offers a comprehensive view of dispensable essential genes and illustrates how their interactions with bypass suppressors reflect evolutionary outcomes.
Collapse
Affiliation(s)
- Carles Pons
- https://ror.org/01z1gye03 Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Jolanda van Leeuwen
- Center for Integrative Genomics, Bâtiment Génopode, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Scimeca RC, Reichard MV. Differential gene expression response to acute and chronic Cytauzxoon felis infection in domestic cats (Felis catus). Ticks Tick Borne Dis 2023; 14:102242. [PMID: 37651848 DOI: 10.1016/j.ttbdis.2023.102242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/25/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Cytauxzoonosis is a severe tick transmitted protozoan disease of domestic cats, caused by Cytauxzoon felis. The disease is characterized by acute onset of high fever, depression, lethargy, inappentence, anorexia, icterus, dehydration, hemolytic anemia, and alteration of immune response. The aim of our study was to further detail the immune response of domestic cats to C. felis infection by comparing the differential expression of feline immune transcriptional elements during acute and chronic cytauxzoonosis. True single molecule sequencing (tSMS) was used to analyze the whole genome of acutely and chronically infected C. felis cats, focusing on the analysis of genes involved on the immune response. Two C. felis donor cats were infested with Amblyomma americanum nymphs, which after repletion were collected and kept in humidity chambers until they molted. The resulting A. americanum were randomly selected to infest three C. felis naïve principal cats. Infection of these cats was confirmed by nested PCR of the 18S rRNA C. felis gene and clinical signs. RNA was extracted from whole blood at different time points and used for tSMS analyses, the results revealed overexpression in transcripts involved in type I interferon signaling, cellular and cytokine responses during the acute stage of infection, while cell cycle, and metabolic processes were downregulated. Genes involved in cell adhesion increased their expression in the chronic infected cats, whereas inflammatory and apoptotic related genes were downregulated. This study provided information on the host immune response to C. felis in domestic cats, demonstrating that inflammatory, apoptotic, and cell adhesion are some of the pathways altered during acute and chronic infection.
Collapse
Affiliation(s)
- Ruth C Scimeca
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078.
| | - Mason V Reichard
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078
| |
Collapse
|
7
|
Williams JD, Zhu D, García-Rubio M, Shaltz S, Aguilera A, Jinks-Robertson S. Spontaneous deamination of cytosine to uracil is biased to the non-transcribed DNA strand in yeast. DNA Repair (Amst) 2023; 126:103489. [PMID: 37018983 PMCID: PMC10494324 DOI: 10.1016/j.dnarep.2023.103489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Transcription in Saccharomyces cerevisiae is associated with elevated mutation and this partially reflects enhanced damage of the corresponding DNA. Spontaneous deamination of cytosine to uracil leads to CG>TA mutations that provide a strand-specific read-out of damage in strains that lack the ability to remove uracil from DNA. Using the CAN1 forward mutation reporter, we found that C>T and G>A mutations, which reflect deamination of the non-transcribed and transcribed DNA strands, respectively, occurred at similar rates under low-transcription conditions. By contrast, the rate of C>T mutations was 3-fold higher than G>A mutations under high-transcription conditions, demonstrating biased deamination of the non-transcribed strand (NTS). The NTS is transiently single-stranded within the ∼15 bp transcription bubble, or a more extensive region of the NTS can be exposed as part of an R-loop that can form behind RNA polymerase. Neither the deletion of genes whose products restrain R-loop formation nor the over-expression of RNase H1, which degrades R-loops, reduced the biased deamination of the NTS, and no transcription-associated R-loop formation at CAN1 was detected. These results suggest that the NTS within the transcription bubble is a target for spontaneous deamination and likely other types of DNA damage.
Collapse
Affiliation(s)
- Jonathan D Williams
- Department of Molecular Genetics and Microbiology, 213 Research Dr., Duke University Medical Center, Durham, NC 27710, USA
| | - Demi Zhu
- Department of Molecular Genetics and Microbiology, 213 Research Dr., Duke University Medical Center, Durham, NC 27710, USA
| | - María García-Rubio
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla-CSIC, Seville, Spain
| | - Samantha Shaltz
- Department of Molecular Genetics and Microbiology, 213 Research Dr., Duke University Medical Center, Durham, NC 27710, USA
| | - Andrés Aguilera
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla-CSIC, Seville, Spain
| | - Sue Jinks-Robertson
- Department of Molecular Genetics and Microbiology, 213 Research Dr., Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Roth MG, Westrick NM, Baldwin TT. Fungal biotechnology: From yesterday to tomorrow. FRONTIERS IN FUNGAL BIOLOGY 2023; 4:1135263. [PMID: 37746125 PMCID: PMC10512358 DOI: 10.3389/ffunb.2023.1135263] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/07/2023] [Indexed: 09/26/2023]
Abstract
Fungi have been used to better the lives of everyday people and unravel the mysteries of higher eukaryotic organisms for decades. However, comparing progress and development stemming from fungal research to that of human, plant, and bacterial research, fungi remain largely understudied and underutilized. Recent commercial ventures have begun to gain popularity in society, providing a new surge of interest in fungi, mycelia, and potential new applications of these organisms to various aspects of research. Biotechnological advancements in fungal research cannot occur without intensive amounts of time, investments, and research tool development. In this review, we highlight past breakthroughs in fungal biotechnology, discuss requirements to advance fungal biotechnology even further, and touch on the horizon of new breakthroughs with the highest potential to positively impact both research and society.
Collapse
Affiliation(s)
- Mitchell G. Roth
- Department of Plant Pathology, The Ohio State University, Wooster, OH, United States
| | - Nathaniel M. Westrick
- Department of Plant Pathology, University of Wisconsin-Madison, Madison, WI, United States
| | - Thomas T. Baldwin
- Department of Plant Pathology, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
9
|
A dynamical stochastic model of yeast translation across the cell cycle. Heliyon 2023; 9:e13101. [PMID: 36793957 PMCID: PMC9922973 DOI: 10.1016/j.heliyon.2023.e13101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 01/04/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Translation is a central step in gene expression, however its quantitative and time-resolved regulation is poorly understood. We developed a discrete, stochastic model for protein translation in S. cerevisiae in a whole-transcriptome, single-cell context. A "base case" scenario representing an average cell highlights translation initiation rates as the main co-translational regulatory parameters. Codon usage bias emerges as a secondary regulatory mechanism through ribosome stalling. Demand for anticodons with low abundancy is shown to cause above-average ribosome dwelling times. Codon usage bias correlates strongly both with protein synthesis rates and elongation rates. Applying the model to a time-resolved transcriptome estimated by combining data from FISH and RNA-Seq experiments, it could be shown that increased total transcript abundance during the cell cycle decreases translation efficiency at single transcript level. Translation efficiency grouped by gene function shows highest values for ribosomal and glycolytic genes. Ribosomal proteins peak in S phase while glycolytic proteins rank highest in later cell cycle phases.
Collapse
|
10
|
St Laurent G, Toma I, Seilheimer B, Cesnulevicius K, Schultz M, Tackett M, Zhou J, Ri M, Shtokalo D, Antonets D, Jepson T, McCaffrey TA. RNAseq analysis of treatment-dependent signaling changes during inflammation in a mouse cutaneous wound healing model. BMC Genomics 2021; 22:854. [PMID: 34823472 PMCID: PMC8614049 DOI: 10.1186/s12864-021-08083-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite proven therapeutic effects in inflammatory conditions, the specific mechanisms of phytochemical therapies are not well understood. The transcriptome effects of Traumeel (Tr14), a multicomponent natural product, and diclofenac, a non-selective cyclooxygenase (COX) inhibitor, were compared in a mouse cutaneous wound healing model to identify both known and novel pathways for the anti-inflammatory effect of plant-derived natural products. METHODS Skin samples from abraded mice were analyzed by single-molecule, amplification-free RNAseq transcript profiling at 7 points between 12 and 192 h after injury. Immediately after injury, the wounds were treated with either diclofenac, Tr14, or placebo control (n = 7 per group/time). RNAseq levels were compared between treatment and control at each time point using a systems biology approach. RESULTS At early time points (12-36 h), both control and Tr14-treated wounds showed marked increase in the inducible COX2 enzyme mRNA, while diclofenac-treated wounds did not. Tr14, in contrast, modulated lipoxygenase transcripts, especially ALOX12/15, and phospholipases involved in arachidonate metabolism. Notably, Tr14 modulated a group of cell-type specific markers, including the T cell receptor, that could be explained by an overarching effect on the type of cells that were recruited into the wound tissue. CONCLUSIONS Tr14 and diclofenac had very different effects on the COX/LOX synthetic pathway after cutaneous wounding. Tr14 allowed normal autoinduction of COX2 mRNA, but suppressed mRNA levels for key enzymes in the leukotriene synthetic pathway. Tr14 appeared to have a broad 'phytocellular' effect on the wound transcriptome by altering the balance of cell types present in the wound.
Collapse
Affiliation(s)
- Georges St Laurent
- The St. Laurent Institute, Vancouver, WA, USA.,SeqLL, Inc., Woburn, MA, USA
| | - Ian Toma
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 Eye St, Washington D.C, 20037, USA
| | | | | | | | - Michael Tackett
- The St. Laurent Institute, Vancouver, WA, USA.,SeqLL, Inc., Woburn, MA, USA
| | | | - Maxim Ri
- The St. Laurent Institute, Vancouver, WA, USA.,AcademGene, LLC, Novosibirsk, Russia
| | - Dmitry Shtokalo
- The St. Laurent Institute, Vancouver, WA, USA.,AcademGene, LLC, Novosibirsk, Russia.,A.P. Ershov Institute of Informatics Systems, Novosibirsk, Russia
| | - Denis Antonets
- AcademGene, LLC, Novosibirsk, Russia.,A.P. Ershov Institute of Informatics Systems, Novosibirsk, Russia
| | - Tisha Jepson
- The St. Laurent Institute, Vancouver, WA, USA.,SeqLL, Inc., Woburn, MA, USA
| | - Timothy A McCaffrey
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 Eye St, Washington D.C, 20037, USA.
| |
Collapse
|
11
|
Decourty L, Malabat C, Frachon E, Jacquier A, Saveanu C. Investigation of RNA metabolism through large-scale genetic interaction profiling in yeast. Nucleic Acids Res 2021; 49:8535-8555. [PMID: 34358317 PMCID: PMC8421204 DOI: 10.1093/nar/gkab680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 07/19/2021] [Accepted: 08/02/2021] [Indexed: 11/15/2022] Open
Abstract
Gene deletion and gene expression alteration can lead to growth defects that are amplified or reduced when a second mutation is present in the same cells. We performed 154 genetic interaction mapping (GIM) screens with query mutants related with RNA metabolism and estimated the growth rates of about 700 000 double mutant Saccharomyces cerevisiae strains. The tested targets included the gene deletion collection and 900 strains in which essential genes were affected by mRNA destabilization (DAmP). To analyze the results, we developed RECAP, a strategy that validates genetic interaction profiles by comparison with gene co-citation frequency, and identified links between 1471 genes and 117 biological processes. In addition to these large-scale results, we validated both enhancement and suppression of slow growth measured for specific RNA-related pathways. Thus, negative genetic interactions identified a role for the OCA inositol polyphosphate hydrolase complex in mRNA translation initiation. By analysis of suppressors, we found that Puf4, a Pumilio family RNA binding protein, inhibits ribosomal protein Rpl9 function, by acting on a conserved UGUAcauUA motif located downstream the stop codon of the RPL9B mRNA. Altogether, the results and their analysis should represent a useful resource for discovery of gene function in yeast.
Collapse
Affiliation(s)
- Laurence Decourty
- Unité de Génétique des Interactions Macromoléculaires, Département Génomes et Génétique, Institut Pasteur, 75015 Paris, France.,UMR3525, Centre national de la recherche scientifique (CNRS), 75015 Paris, France
| | - Christophe Malabat
- Hub Bioinformatique et Biostatistique, Département de Biologie Computationnelle, Institut Pasteur, 75015 Paris, France
| | - Emmanuel Frachon
- Plate-forme Technologique Biomatériaux et Microfluidique, Centre des ressources et recherches technologiques, Institut Pasteur, 75015 Paris, France
| | - Alain Jacquier
- Unité de Génétique des Interactions Macromoléculaires, Département Génomes et Génétique, Institut Pasteur, 75015 Paris, France.,UMR3525, Centre national de la recherche scientifique (CNRS), 75015 Paris, France
| | - Cosmin Saveanu
- Unité de Génétique des Interactions Macromoléculaires, Département Génomes et Génétique, Institut Pasteur, 75015 Paris, France.,UMR3525, Centre national de la recherche scientifique (CNRS), 75015 Paris, France
| |
Collapse
|
12
|
van Leeuwen J, Pons C, Tan G, Wang JZ, Hou J, Weile J, Gebbia M, Liang W, Shuteriqi E, Li Z, Lopes M, Ušaj M, Dos Santos Lopes A, van Lieshout N, Myers CL, Roth FP, Aloy P, Andrews BJ, Boone C. Systematic analysis of bypass suppression of essential genes. Mol Syst Biol 2021; 16:e9828. [PMID: 32939983 PMCID: PMC7507402 DOI: 10.15252/msb.20209828] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Essential genes tend to be highly conserved across eukaryotes, but, in some cases, their critical roles can be bypassed through genetic rewiring. From a systematic analysis of 728 different essential yeast genes, we discovered that 124 (17%) were dispensable essential genes. Through whole-genome sequencing and detailed genetic analysis, we investigated the genetic interactions and genome alterations underlying bypass suppression. Dispensable essential genes often had paralogs, were enriched for genes encoding membrane-associated proteins, and were depleted for members of protein complexes. Functionally related genes frequently drove the bypass suppression interactions. These gene properties were predictive of essential gene dispensability and of specific suppressors among hundreds of genes on aneuploid chromosomes. Our findings identify yeast's core essential gene set and reveal that the properties of dispensable essential genes are conserved from yeast to human cells, correlating with human genes that display cell line-specific essentiality in the Cancer Dependency Map (DepMap) project.
Collapse
Affiliation(s)
- Jolanda van Leeuwen
- Center for Integrative Genomics, Bâtiment Génopode, University of Lausanne, Lausanne, Switzerland.,Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Carles Pons
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Guihong Tan
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Jason Zi Wang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Jing Hou
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Jochen Weile
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Marinella Gebbia
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Wendy Liang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Ermira Shuteriqi
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Zhijian Li
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Maykel Lopes
- Center for Integrative Genomics, Bâtiment Génopode, University of Lausanne, Lausanne, Switzerland
| | - Matej Ušaj
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Andreia Dos Santos Lopes
- Center for Integrative Genomics, Bâtiment Génopode, University of Lausanne, Lausanne, Switzerland
| | - Natascha van Lieshout
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Frederick P Roth
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Brenda J Andrews
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Charles Boone
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Arita Y, Kim G, Li Z, Friesen H, Turco G, Wang RY, Climie D, Usaj M, Hotz M, Stoops EH, Baryshnikova A, Boone C, Botstein D, Andrews BJ, McIsaac RS. A genome-scale yeast library with inducible expression of individual genes. Mol Syst Biol 2021; 17:e10207. [PMID: 34096681 PMCID: PMC8182650 DOI: 10.15252/msb.202110207] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
The ability to switch a gene from off to on and monitor dynamic changes provides a powerful approach for probing gene function and elucidating causal regulatory relationships. Here, we developed and characterized YETI (Yeast Estradiol strains with Titratable Induction), a collection in which > 5,600 yeast genes are engineered for transcriptional inducibility with single-gene precision at their native loci and without plasmids. Each strain contains SGA screening markers and a unique barcode, enabling high-throughput genetics. We characterized YETI using growth phenotyping and BAR-seq screens, and we used a YETI allele to identify the regulon of Rof1, showing that it acts to repress transcription. We observed that strains with inducible essential genes that have low native expression can often grow without inducer. Analysis of data from eukaryotic and prokaryotic systems shows that native expression is a variable that can bias promoter-perturbing screens, including CRISPRi. We engineered a second expression system, Z3 EB42, that gives lower expression than Z3 EV, a feature enabling conditional activation and repression of lowly expressed essential genes that grow without inducer in the YETI library.
Collapse
Affiliation(s)
- Yuko Arita
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- RIKEN Centre for Sustainable Resource ScienceWakoSaitamaJapan
| | - Griffin Kim
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | - Zhijian Li
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Helena Friesen
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Gina Turco
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | | | - Dale Climie
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Matej Usaj
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Manuel Hotz
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | | | | | - Charles Boone
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- RIKEN Centre for Sustainable Resource ScienceWakoSaitamaJapan
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | | | - Brenda J Andrews
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | | |
Collapse
|
14
|
Petibon C, Malik Ghulam M, Catala M, Abou Elela S. Regulation of ribosomal protein genes: An ordered anarchy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 12:e1632. [PMID: 33038057 PMCID: PMC8047918 DOI: 10.1002/wrna.1632] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023]
Abstract
Ribosomal protein genes are among the most highly expressed genes in most cell types. Their products are generally essential for ribosome synthesis, which is the cornerstone for cell growth and proliferation. Many cellular resources are dedicated to producing ribosomal proteins and thus this process needs to be regulated in ways that carefully balance the supply of nascent ribosomal proteins with the demand for new ribosomes. Ribosomal protein genes have classically been viewed as a uniform interconnected regulon regulated in eukaryotic cells by target of rapamycin and protein kinase A pathway in response to changes in growth conditions and/or cellular status. However, recent literature depicts a more complex picture in which the amount of ribosomal proteins produced varies between genes in response to two overlapping regulatory circuits. The first includes the classical general ribosome‐producing program and the second is a gene‐specific feature responsible for fine‐tuning the amount of ribosomal proteins produced from each individual ribosomal gene. Unlike the general pathway that is mainly controlled at the level of transcription and translation, this specific regulation of ribosomal protein genes is largely achieved through changes in pre‐mRNA splicing efficiency and mRNA stability. By combining general and specific regulation, the cell can coordinate ribosome production, while allowing functional specialization and diversity. Here we review the many ways ribosomal protein genes are regulated, with special focus on the emerging role of posttranscriptional regulatory events in fine‐tuning the expression of ribosomal protein genes and its role in controlling the potential variation in ribosome functions. This article is categorized under:Translation > Ribosome Biogenesis Translation > Ribosome Structure/Function Translation > Translation Regulation
Collapse
Affiliation(s)
- Cyrielle Petibon
- Département de microbiologie et d'infectiologie, Universite de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Sherbrooke, Quebec, Canada
| | - Mustafa Malik Ghulam
- Département de microbiologie et d'infectiologie, Universite de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Sherbrooke, Quebec, Canada
| | - Mathieu Catala
- Département de microbiologie et d'infectiologie, Universite de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Sherbrooke, Quebec, Canada
| | - Sherif Abou Elela
- Département de microbiologie et d'infectiologie, Universite de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Sherbrooke, Quebec, Canada
| |
Collapse
|
15
|
Tang H, Wu Y, Deng J, Chen N, Zheng Z, Wei Y, Luo X, Keasling JD. Promoter Architecture and Promoter Engineering in Saccharomyces cerevisiae. Metabolites 2020; 10:metabo10080320. [PMID: 32781665 PMCID: PMC7466126 DOI: 10.3390/metabo10080320] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022] Open
Abstract
Promoters play an essential role in the regulation of gene expression for fine-tuning genetic circuits and metabolic pathways in Saccharomyces cerevisiae (S. cerevisiae). However, native promoters in S. cerevisiae have several limitations which hinder their applications in metabolic engineering. These limitations include an inadequate number of well-characterized promoters, poor dynamic range, and insufficient orthogonality to endogenous regulations. Therefore, it is necessary to perform promoter engineering to create synthetic promoters with better properties. Here, we review recent advances related to promoter architecture, promoter engineering and synthetic promoter applications in S. cerevisiae. We also provide a perspective of future directions in this field with an emphasis on the recent advances of machine learning based promoter designs.
Collapse
Affiliation(s)
- Hongting Tang
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
| | - Yanling Wu
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
| | - Jiliang Deng
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
| | - Nanzhu Chen
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
| | - Zhaohui Zheng
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
| | - Yongjun Wei
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China;
| | - Xiaozhou Luo
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
- Correspondence: (X.L.); (J.D.K.)
| | - Jay D. Keasling
- Center for Synthetic Biochemistry, Shenzhen Institutes for Advanced Technologies, Chinese Academy of Sciences, Shenzhen 518055, China; (H.T.); (Y.W.); (J.D.); (N.C.); (Z.Z.)
- Joint BioEnergy Institute, Emeryville, CA 94608, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering & Department of Bioengineering, University of California, Berkeley, CA 94720, USA
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Correspondence: (X.L.); (J.D.K.)
| |
Collapse
|
16
|
Mattiazzi Usaj M, Sahin N, Friesen H, Pons C, Usaj M, Masinas MPD, Shuteriqi E, Shkurin A, Aloy P, Morris Q, Boone C, Andrews BJ. Systematic genetics and single-cell imaging reveal widespread morphological pleiotropy and cell-to-cell variability. Mol Syst Biol 2020; 16:e9243. [PMID: 32064787 PMCID: PMC7025093 DOI: 10.15252/msb.20199243] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Our ability to understand the genotype-to-phenotype relationship is hindered by the lack of detailed understanding of phenotypes at a single-cell level. To systematically assess cell-to-cell phenotypic variability, we combined automated yeast genetics, high-content screening and neural network-based image analysis of single cells, focussing on genes that influence the architecture of four subcellular compartments of the endocytic pathway as a model system. Our unbiased assessment of the morphology of these compartments-endocytic patch, actin patch, late endosome and vacuole-identified 17 distinct mutant phenotypes associated with ~1,600 genes (~30% of all yeast genes). Approximately half of these mutants exhibited multiple phenotypes, highlighting the extent of morphological pleiotropy. Quantitative analysis also revealed that incomplete penetrance was prevalent, with the majority of mutants exhibiting substantial variability in phenotype at the single-cell level. Our single-cell analysis enabled exploration of factors that contribute to incomplete penetrance and cellular heterogeneity, including replicative age, organelle inheritance and response to stress.
Collapse
Affiliation(s)
| | - Nil Sahin
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | | | - Carles Pons
- Institute for Research in Biomedicine (IRB Barcelona)The Barcelona Institute for Science and TechnologyBarcelona, CataloniaSpain
| | - Matej Usaj
- The Donnelly CentreUniversity of TorontoTorontoONCanada
| | | | | | - Aleksei Shkurin
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona)The Barcelona Institute for Science and TechnologyBarcelona, CataloniaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona, CataloniaSpain
| | - Quaid Morris
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Computational and Systems Biology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Charles Boone
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- RIKEN Centre for Sustainable Resource ScienceWakoSaitamaJapan
| | - Brenda J Andrews
- The Donnelly CentreUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| |
Collapse
|
17
|
Cao H, Salazar-García L, Gao F, Wahlestedt T, Wu CL, Han X, Cai Y, Xu D, Wang F, Tang L, Ricciardi N, Cai D, Wang H, Chin MPS, Timmons JA, Wahlestedt C, Kapranov P. Novel approach reveals genomic landscapes of single-strand DNA breaks with nucleotide resolution in human cells. Nat Commun 2019; 10:5799. [PMID: 31862872 PMCID: PMC6925131 DOI: 10.1038/s41467-019-13602-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
Single-strand breaks (SSBs) represent the major form of DNA damage, yet techniques to map these lesions genome-wide with nucleotide-level precision are limited. Here, we present a method, termed SSiNGLe, and demonstrate its utility to explore the distribution and dynamic changes in genome-wide SSBs in response to different biological and environmental stimuli. We validate SSiNGLe using two very distinct sequencing techniques and apply it to derive global profiles of SSBs in different biological states. Strikingly, we show that patterns of SSBs in the genome are non-random, specific to different biological states, enriched in regulatory elements, exons, introns, specific types of repeats and exhibit differential preference for the template strand between exons and introns. Furthermore, we show that breaks likely contribute to naturally occurring sequence variants. Finally, we demonstrate strong links between SSB patterns and age. Overall, SSiNGLe provides access to unexplored realms of cellular biology, not obtainable with current approaches.
Collapse
Affiliation(s)
- Huifen Cao
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Lorena Salazar-García
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Fan Gao
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Thor Wahlestedt
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Chun-Lin Wu
- Department of Pathology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xueer Han
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Ye Cai
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Dongyang Xu
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Fang Wang
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Lu Tang
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Natalie Ricciardi
- Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Ave, Miami, FL, 33136, USA
| | - DingDing Cai
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Huifang Wang
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - Mario P S Chin
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China
| | - James A Timmons
- Augur Precision Medicine LTD, Scion House, Stirling University Innovation Park, Stirling, FK9 4NF, UK
| | - Claes Wahlestedt
- Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Ave, Miami, FL, 33136, USA.
| | - Philipp Kapranov
- Institute of Genomics, School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China.
| |
Collapse
|
18
|
Tan C, Liu H, Ren J, Ye X, Feng H, Liu Z. Single-molecule real-time sequencing facilitates the analysis of transcripts and splice isoforms of anthers in Chinese cabbage (Brassica rapa L. ssp. pekinensis). BMC PLANT BIOLOGY 2019; 19:517. [PMID: 31771515 PMCID: PMC6880451 DOI: 10.1186/s12870-019-2133-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/12/2019] [Indexed: 05/06/2023]
Abstract
BACKGROUND Anther development has been extensively studied at the transcriptional level, but a systematic analysis of full-length transcripts on a genome-wide scale has not yet been published. Here, the Pacific Biosciences (PacBio) Sequel platform and next-generation sequencing (NGS) technology were combined to generate full-length sequences and completed structures of transcripts in anthers of Chinese cabbage. RESULTS Using single-molecule real-time sequencing (SMRT), a total of 1,098,119 circular consensus sequences (CCSs) were generated with a mean length of 2664 bp. More than 75% of the CCSs were considered full-length non-chimeric (FLNC) reads. After error correction, 725,731 high-quality FLNC reads were estimated to carry 51,501 isoforms from 19,503 loci, consisting of 38,992 novel isoforms from known genes and 3691 novel isoforms from novel genes. Of the novel isoforms, we identified 407 long non-coding RNAs (lncRNAs) and 37,549 open reading frames (ORFs). Furthermore, a total of 453,270 alternative splicing (AS) events were identified and the majority of AS models in anther were determined to be approximate exon skipping (XSKIP) events. Of the key genes regulated during anther development, AS events were mainly identified in the genes SERK1, CALS5, NEF1, and CESA1/3. Additionally, we identified 104 fusion transcripts and 5806 genes that had alternative polyadenylation (APA). CONCLUSIONS Our work demonstrated the transcriptome diversity and complexity of anther development in Chinese cabbage. The findings provide a basis for further genome annotation and transcriptome research in Chinese cabbage.
Collapse
Affiliation(s)
- Chong Tan
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China
| | - Hongxin Liu
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China
| | - Jie Ren
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China
| | - Xueling Ye
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China
| | - Hui Feng
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China
| | - Zhiyong Liu
- College of Horticulture, Shenyang Agricultural University, Shenyang, Liaoning, 110866, People's Republic of China.
| |
Collapse
|
19
|
Sessegolo C, Cruaud C, Da Silva C, Cologne A, Dubarry M, Derrien T, Lacroix V, Aury JM. Transcriptome profiling of mouse samples using nanopore sequencing of cDNA and RNA molecules. Sci Rep 2019; 9:14908. [PMID: 31624302 PMCID: PMC6797730 DOI: 10.1038/s41598-019-51470-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/28/2019] [Indexed: 01/27/2023] Open
Abstract
Our vision of DNA transcription and splicing has changed dramatically with the introduction of short-read sequencing. These high-throughput sequencing technologies promised to unravel the complexity of any transcriptome. Generally gene expression levels are well-captured using these technologies, but there are still remaining caveats due to the limited read length and the fact that RNA molecules had to be reverse transcribed before sequencing. Oxford Nanopore Technologies has recently launched a portable sequencer which offers the possibility of sequencing long reads and most importantly RNA molecules. Here we generated a full mouse transcriptome from brain and liver using the Oxford Nanopore device. As a comparison, we sequenced RNA (RNA-Seq) and cDNA (cDNA-Seq) molecules using both long and short reads technologies and tested the TeloPrime preparation kit, dedicated to the enrichment of full-length transcripts. Using spike-in data, we confirmed that expression levels are efficiently captured by cDNA-Seq using short reads. More importantly, Oxford Nanopore RNA-Seq tends to be more efficient, while cDNA-Seq appears to be more biased. We further show that the cDNA library preparation of the Nanopore protocol induces read truncation for transcripts containing internal runs of T's. This bias is marked for runs of at least 15 T's, but is already detectable for runs of at least 9 T's and therefore concerns more than 20% of expressed transcripts in mouse brain and liver. Finally, we outline that bioinformatics challenges remain ahead for quantifying at the transcript level, especially when reads are not full-length. Accurate quantification of repeat-associated genes such as processed pseudogenes also remains difficult, and we show that current mapping protocols which map reads to the genome largely over-estimate their expression, at the expense of their parent gene.
Collapse
Affiliation(s)
- Camille Sessegolo
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622, Villeurbanne, France
- EPI ERABLE - Inria Grenoble, Rhône-Alpes, France
| | - Corinne Cruaud
- Genoscope, Institut de biologie François-Jacob, Commissariat a l'Energie Atomique (CEA), Université Paris-Saclay, F-91057, Evry, France
| | - Corinne Da Silva
- Genoscope, Institut de biologie François-Jacob, Commissariat a l'Energie Atomique (CEA), Université Paris-Saclay, F-91057, Evry, France
| | - Audric Cologne
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622, Villeurbanne, France
- EPI ERABLE - Inria Grenoble, Rhône-Alpes, France
| | - Marion Dubarry
- Genoscope, Institut de biologie François-Jacob, Commissariat a l'Energie Atomique (CEA), Université Paris-Saclay, F-91057, Evry, France
| | - Thomas Derrien
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Rennes, France
| | - Vincent Lacroix
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622, Villeurbanne, France
- EPI ERABLE - Inria Grenoble, Rhône-Alpes, France
| | - Jean-Marc Aury
- Genoscope, Institut de biologie François-Jacob, Commissariat a l'Energie Atomique (CEA), Université Paris-Saclay, F-91057, Evry, France.
| |
Collapse
|
20
|
Catala M, Abou Elela S. Promoter-dependent nuclear RNA degradation ensures cell cycle-specific gene expression. Commun Biol 2019; 2:211. [PMID: 31240249 PMCID: PMC6572803 DOI: 10.1038/s42003-019-0441-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/29/2019] [Indexed: 11/09/2022] Open
Abstract
Cell cycle progression depends on phase-specific gene expression. Here we show that the nuclear RNA degradation machinery plays a lead role in promoting cell cycle-dependent gene expression by triggering promoter-dependent co-transcriptional RNA degradation. Single molecule quantification of RNA abundance in different phases of the cell cycle indicates that relative curtailment of gene expression in certain phases is attained even when transcription is not completely inhibited. When nuclear ribonucleases are deleted, transcription of the Saccharomyces cerevisiae G1-specific axial budding gene AXL2 is detected throughout the cell cycle and its phase-specific expression is lost. Promoter replacement abolished cell cycle-dependent RNA degradation and rendered the RNA insensitive to the deletion of nuclear ribonucleases. Together the data reveal a model of gene regulation whereby RNA abundance is controlled by promoter-dependent induction of RNA degradation.
Collapse
Affiliation(s)
- Mathieu Catala
- Département de microbiologie et d’infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8 Canada
| | - Sherif Abou Elela
- Département de microbiologie et d’infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8 Canada
| |
Collapse
|
21
|
Assessment of Bioleaching Microbial Community Structure and Function Based on Next-Generation Sequencing Technologies. MINERALS 2018. [DOI: 10.3390/min8120596] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
It is widely known that bioleaching microorganisms have to cope with the complex extreme environment in which microbial ecology relating to community structure and function varies across environmental types. However, analyses of microbial ecology of bioleaching bacteria is still a challenge. To address this challenge, numerous technologies have been developed. In recent years, high-throughput sequencing technologies enabling comprehensive sequencing analysis of cellular RNA and DNA within the reach of most laboratories have been added to the toolbox of microbial ecology. The next-generation sequencing technology allowing processing DNA sequences can produce available draft genomic sequences of more bioleaching bacteria, which provides the opportunity to predict models of genetic and metabolic potential of bioleaching bacteria and ultimately deepens our understanding of bioleaching microorganism. High-throughput sequencing that focuses on targeted phylogenetic marker 16S rRNA has been effectively applied to characterize the community diversity in an ore leaching environment. RNA-seq, another application of high-throughput sequencing to profile RNA, can be for both mapping and quantifying transcriptome and has demonstrated a high efficiency in quantifying the changing expression level of each transcript under different conditions. It has been demonstrated as a powerful tool for dissecting the relationship between genotype and phenotype, leading to interpreting functional elements of the genome and revealing molecular mechanisms of adaption. This review aims to describe the high-throughput sequencing approach for bioleaching environmental microorganisms, particularly focusing on its application associated with challenges.
Collapse
|
22
|
Ho YH, Shishkova E, Hose J, Coon JJ, Gasch AP. Decoupling Yeast Cell Division and Stress Defense Implicates mRNA Repression in Translational Reallocation during Stress. Curr Biol 2018; 28:2673-2680.e4. [PMID: 30078561 DOI: 10.1016/j.cub.2018.06.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/24/2018] [Accepted: 06/19/2018] [Indexed: 12/29/2022]
Abstract
Stress tolerance and rapid growth are often competing interests in cells. Upon severe environmental stress, many organisms activate defense systems concurrent with growth arrest. There has been debate as to whether aspects of the stress-activated transcriptome are regulated by stress or an indirect byproduct of reduced proliferation. For example, stressed Saccharomyces cerevisiae cells mount a common gene expression program called the environmental stress response (ESR) [1] comprised of ∼300 induced (iESR) transcripts involved in stress defense and ∼600 reduced (rESR) mRNAs encoding ribosomal proteins (RPs) and ribosome biogenesis factors (RiBi) important for division. Because ESR activation also correlates with reduced growth rate in nutrient-restricted chemostats and prolonged G1 in slow-growing mutants, an alternate proposal is that the ESR is simply a consequence of reduced division [2-5]. A major challenge is that past studies did not separate effects of division arrest and stress defense; thus, the true responsiveness of the ESR-and the purpose of stress-dependent rESR repression in particular-remains unclear. Here, we decoupled cell division from the stress response by following transcriptome, proteome, and polysome changes in arrested cells responding to acute stress. We show that the ESR cannot be explained by changes in growth rate or cell-cycle phase during stress acclimation. Instead, failure to repress rESR transcripts reduces polysome association of induced transcripts, delaying production of their proteins. Our results suggest that stressed cells alleviate competition for translation factors by removing mRNAs and ribosomes from the translating pool, directing translational capacity toward induced transcripts to accelerate protein production.
Collapse
Affiliation(s)
- Yi-Hsuan Ho
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Evgenia Shishkova
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James Hose
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Joshua J Coon
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - Audrey P Gasch
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
23
|
Wilson D, Daly NL. Venomics: A Mini-Review. High Throughput 2018; 7:E19. [PMID: 30041430 PMCID: PMC6164461 DOI: 10.3390/ht7030019] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/23/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023] Open
Abstract
Venomics is the integration of proteomic, genomic and transcriptomic approaches to study venoms. Advances in these approaches have enabled increasingly more comprehensive analyses of venoms to be carried out, overcoming to some extent the limitations imposed by the complexity of the venoms and the small quantities that are often available. Advances in bioinformatics and high-throughput functional assay screening approaches have also had a significant impact on venomics. A combination of all these techniques is critical for enhancing our knowledge on the complexity of venoms and their potential therapeutic and agricultural applications. Here we highlight recent advances in these fields and their impact on venom analyses.
Collapse
Affiliation(s)
- David Wilson
- Centre for Biodiscovery and Molecular, Development of Therapeutics, AITHM, James Cook University, Cairns, QLD 4878, Australia.
| | - Norelle L Daly
- Centre for Biodiscovery and Molecular, Development of Therapeutics, AITHM, James Cook University, Cairns, QLD 4878, Australia.
| |
Collapse
|
24
|
Abstract
Throughout the past nearly a decade, the application of high-throughput sequencing to RNA molecules in the form of RNA sequencing (RNA-seq) and its many variations has revolutionized transcriptomic studies by enabling researchers to take a simultaneously deep and truly global look into the transcriptome. However, there is still considerable scope for improvement on RNA-seq data in its current form, primarily because of the short-read nature of the dominant sequencing technologies, which prevents the completely reliable reconstruction and quantification of full-length transcripts, and the sequencing library building protocols used, which introduce various distortions in the final data sets. The ideal approach toward resolving these remaining issues would involve the direct amplification-free sequencing of full-length RNA molecules. This has recently become practical with the advent of nanopore sequencing, which raises the possibility of yet another revolution in transcriptomics. I discuss the design considerations to be taken into account, the technical challenges that need to be addressed and the biological questions these advances can be expected to resolve.
Collapse
|
25
|
Garalde DR, Snell EA, Jachimowicz D, Sipos B, Lloyd JH, Bruce M, Pantic N, Admassu T, James P, Warland A, Jordan M, Ciccone J, Serra S, Keenan J, Martin S, McNeill L, Wallace EJ, Jayasinghe L, Wright C, Blasco J, Young S, Brocklebank D, Juul S, Clarke J, Heron AJ, Turner DJ. Highly parallel direct RNA sequencing on an array of nanopores. Nat Methods 2018; 15:201-206. [PMID: 29334379 DOI: 10.1101/068809] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/21/2017] [Indexed: 05/23/2023]
Abstract
Sequencing the RNA in a biological sample can unlock a wealth of information, including the identity of bacteria and viruses, the nuances of alternative splicing or the transcriptional state of organisms. However, current methods have limitations due to short read lengths and reverse transcription or amplification biases. Here we demonstrate nanopore direct RNA-seq, a highly parallel, real-time, single-molecule method that circumvents reverse transcription or amplification steps. This method yields full-length, strand-specific RNA sequences and enables the direct detection of nucleotide analogs in RNA.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark Bruce
- Oxford Nanopore Technologies Ltd., Oxford, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sissel Juul
- Oxford Nanopore Technologies Inc., New York, New York, USA
| | | | | | | |
Collapse
|
26
|
Ho B, Baryshnikova A, Brown GW. Unification of Protein Abundance Datasets Yields a Quantitative Saccharomyces cerevisiae Proteome. Cell Syst 2018; 6:192-205.e3. [PMID: 29361465 DOI: 10.1016/j.cels.2017.12.004] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/10/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022]
Abstract
Protein activity is the ultimate arbiter of function in most cellular pathways, and protein concentration is fundamentally connected to protein action. While the proteome of yeast has been subjected to the most comprehensive analysis of any eukaryote, existing datasets are difficult to compare, and there is no consensus abundance value for each protein. We evaluated 21 quantitative analyses of the S. cerevisiae proteome, normalizing and converting all measurements of protein abundance into the intuitive measurement of absolute molecules per cell. We estimate the cellular abundance of 92% of the proteins in the yeast proteome and assess the variation in each abundance measurement. Using our protein abundance dataset, we find that a global response to diverse environmental stresses is not detected at the level of protein abundance, we find that protein tags have only a modest effect on protein abundance, and we identify proteins that are differentially regulated at the mRNA abundance, mRNA translation, and protein abundance levels.
Collapse
Affiliation(s)
- Brandon Ho
- Department of Biochemistry and Donnelly Center, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anastasia Baryshnikova
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Grant W Brown
- Department of Biochemistry and Donnelly Center, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
27
|
Garalde DR, Snell EA, Jachimowicz D, Sipos B, Lloyd JH, Bruce M, Pantic N, Admassu T, James P, Warland A, Jordan M, Ciccone J, Serra S, Keenan J, Martin S, McNeill L, Wallace EJ, Jayasinghe L, Wright C, Blasco J, Young S, Brocklebank D, Juul S, Clarke J, Heron AJ, Turner DJ. Highly parallel direct RNA sequencing on an array of nanopores. Nat Methods 2018; 15:201-206. [DOI: 10.1038/nmeth.4577] [Citation(s) in RCA: 568] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
|
28
|
Gasch AP, Yu FB, Hose J, Escalante LE, Place M, Bacher R, Kanbar J, Ciobanu D, Sandor L, Grigoriev IV, Kendziorski C, Quake SR, McClean MN. Single-cell RNA sequencing reveals intrinsic and extrinsic regulatory heterogeneity in yeast responding to stress. PLoS Biol 2017; 15:e2004050. [PMID: 29240790 PMCID: PMC5746276 DOI: 10.1371/journal.pbio.2004050] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/28/2017] [Accepted: 11/17/2017] [Indexed: 02/01/2023] Open
Abstract
From bacteria to humans, individual cells within isogenic populations can show significant variation in stress tolerance, but the nature of this heterogeneity is not clear. To investigate this, we used single-cell RNA sequencing to quantify transcript heterogeneity in single Saccharomyces cerevisiae cells treated with and without salt stress to explore population variation and identify cellular covariates that influence the stress-responsive transcriptome. Leveraging the extensive knowledge of yeast transcriptional regulation, we uncovered significant regulatory variation in individual yeast cells, both before and after stress. We also discovered that a subset of cells appears to decouple expression of ribosomal protein genes from the environmental stress response in a manner partly correlated with the cell cycle but unrelated to the yeast ultradian metabolic cycle. Live-cell imaging of cells expressing pairs of fluorescent regulators, including the transcription factor Msn2 with Dot6, Sfp1, or MAP kinase Hog1, revealed both coordinated and decoupled nucleocytoplasmic shuttling. Together with transcriptomic analysis, our results suggest that cells maintain a cellular filter against decoupled bursts of transcription factor activation but mount a stress response upon coordinated regulation, even in a subset of unstressed cells. Genetically identical cells growing in the same environment can vary in their cellular state and behavior. Such heterogeneity may explain why some cells in an isogenic population can survive sudden severe environmental stress whereas other cells succumb. Cell-to-cell variation in gene expression has been linked to variable stress survival, but how and why transcript levels vary across the transcriptome in single cells is only beginning to emerge. Here, we used single-cell RNA sequencing (scRNA-seq) to measure cell-to-cell heterogeneity in the transcriptome of budding yeast (Saccharomyces cerevisiae). We find surprising patterns of variation across known sets of transcription factor targets, indicating that cells vary in their transcriptome profile both before and after stress exposure. scRNA-seq analysis combined with live-cell imaging of transcription factor activation dynamics revealed some cells in which the stress response was coordinately activated and other cells in which the traditional response was decoupled, suggesting unrecognized regulatory nuances that expand our understanding of stress response and survival.
Collapse
Affiliation(s)
- Audrey P. Gasch
- Laboratory of Genetics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Great Lakes Bioenergy Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- * E-mail:
| | - Feiqiao Brian Yu
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - James Hose
- Laboratory of Genetics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Leah E. Escalante
- Laboratory of Genetics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mike Place
- Great Lakes Bioenergy Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Rhonda Bacher
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Jad Kanbar
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Doina Ciobanu
- Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Laura Sandor
- Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Igor V. Grigoriev
- Department of Energy Joint Genome Institute, Walnut Creek, California, United States of America
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Megan N. McClean
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
29
|
Li JJ, Chew GL, Biggin MD. Quantitating translational control: mRNA abundance-dependent and independent contributions and the mRNA sequences that specify them. Nucleic Acids Res 2017; 45:11821-11836. [PMID: 29040683 PMCID: PMC5714229 DOI: 10.1093/nar/gkx898] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/25/2017] [Indexed: 11/17/2022] Open
Abstract
Translation rate per mRNA molecule correlates positively with mRNA abundance. As a result, protein levels do not scale linearly with mRNA levels, but instead scale with the abundance of mRNA raised to the power of an ‘amplification exponent’. Here we show that to quantitate translational control, the translation rate must be decomposed into two components. One, TRmD, depends on the mRNA level and defines the amplification exponent. The other, TRmIND, is independent of mRNA amount and impacts the correlation coefficient between protein and mRNA levels. We show that in Saccharomyces cerevisiae TRmD represents ∼20% of the variance in translation and directs an amplification exponent of 1.20 with a 95% confidence interval [1.14, 1.26]. TRmIND constitutes the remaining ∼80% of the variance in translation and explains ∼5% of the variance in protein expression. We also find that TRmD and TRmIND are preferentially determined by different mRNA sequence features: TRmIND by the length of the open reading frame and TRmD both by a ∼60 nucleotide element that spans the initiating AUG and by codon and amino acid frequency. Our work provides more appropriate estimates of translational control and implies that TRmIND is under different evolutionary selective pressures than TRmD.
Collapse
Affiliation(s)
- Jingyi Jessica Li
- Department of Statistics and Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Guo-Liang Chew
- Computational Biology Program, Public Health Sciences and Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mark D Biggin
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94708, USA
| |
Collapse
|
30
|
Gliddon HD, Herberg JA, Levin M, Kaforou M. Genome-wide host RNA signatures of infectious diseases: discovery and clinical translation. Immunology 2017; 153:171-178. [PMID: 28921535 PMCID: PMC5765383 DOI: 10.1111/imm.12841] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 12/31/2022] Open
Abstract
The use of whole blood gene expression to derive diagnostic biomarkers capable of distinguishing between phenotypically similar diseases holds great promise but remains a challenge. Differential gene expression analysis is used to identify the key genes that undergo changes in expression relative to healthy individuals, as well as to patients with other diseases. These key genes can act as diagnostic, prognostic and predictive markers of disease. Gene expression ‘signatures’ in the blood hold the potential to be used for the diagnosis of infectious diseases, where current diagnostics are unreliable, ineffective or of limited potential. For diagnostic tests based on RNA signatures to be useful clinically, the first step is to identify the minimum set of gene transcripts that accurately identify the disease in question. The second requirement is rapid and cost‐effective detection of the gene expression levels. Signatures have been described for a number of infectious diseases, but ‘clinic‐ready’ technologies for RNA detection from clinical samples are limited, though existing methods such as RT‐PCR are likely to be superseded by a number of emerging technologies, which may form the basis of the translation of gene expression signatures into routine diagnostic tests for a range of disease states.
Collapse
Affiliation(s)
- Harriet D Gliddon
- London Centre for Nanotechnology, University College London, London, UK
| | | | - Michael Levin
- Department of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
31
|
St Laurent G, Seilheimer B, Tackett M, Zhou J, Shtokalo D, Vyatkin Y, Ri M, Toma I, Jones D, McCaffrey TA. Deep Sequencing Transcriptome Analysis of Murine Wound Healing: Effects of a Multicomponent, Multitarget Natural Product Therapy-Tr14. Front Mol Biosci 2017; 4:57. [PMID: 28879183 PMCID: PMC5572416 DOI: 10.3389/fmolb.2017.00057] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023] Open
Abstract
Wound healing involves an orchestrated response that engages multiple processes, such as hemostasis, cellular migration, extracellular matrix synthesis, and in particular, inflammation. Using a murine model of cutaneous wound repair, the transcriptome was mapped from 12 h to 8 days post-injury, and in response to a multicomponent, multi-target natural product, Tr14. Using single-molecule RNA sequencing (RNA-seq), there were clear temporal changes in known transcripts related to wound healing pathways, and additional novel transcripts of both coding and non-coding genes. Tr14 treatment modulated >100 transcripts related to key wound repair pathways, such as response to wounding, wound contraction, and cytokine response. The results provide the most precise and comprehensive characterization to date of the transcriptome's response to skin damage, repair, and multicomponent natural product therapy. By understanding the wound repair process, and the effects of natural products, it should be possible to intervene more effectively in diseases involving aberrant repair.
Collapse
Affiliation(s)
- Georges St Laurent
- St. Laurent InstituteVancouver, WA, United States.,SeqLL, Inc.Woburn, MA, United States
| | | | | | - Jianhua Zhou
- St. Laurent InstituteVancouver, WA, United States.,Nantong UniversityNantong, China
| | - Dmitry Shtokalo
- St. Laurent InstituteVancouver, WA, United States.,A.P.Ershov Institute of Informatics SystemsNovosibirsk, Russia.,AcademGene LLCNovosibirsk, Russia
| | - Yuri Vyatkin
- St. Laurent InstituteVancouver, WA, United States.,AcademGene LLCNovosibirsk, Russia
| | - Maxim Ri
- St. Laurent InstituteVancouver, WA, United States.,AcademGene LLCNovosibirsk, Russia
| | - Ian Toma
- Nantong UniversityNantong, China
| | - Dan Jones
- Biologische Heilmittel Heel GmbHBaden-Baden, Germany
| | - Timothy A McCaffrey
- Division of Genomic Medicine, The George Washington UniversityWashington, DC, United States
| |
Collapse
|
32
|
Rogers DW, Böttcher MA, Traulsen A, Greig D. Ribosome reinitiation can explain length-dependent translation of messenger RNA. PLoS Comput Biol 2017; 13:e1005592. [PMID: 28598992 PMCID: PMC5482490 DOI: 10.1371/journal.pcbi.1005592] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/23/2017] [Accepted: 05/25/2017] [Indexed: 12/21/2022] Open
Abstract
Models of mRNA translation usually presume that transcripts are linear; upon reaching the end of a transcript each terminating ribosome returns to the cytoplasmic pool before initiating anew on a different transcript. A consequence of linear models is that faster translation of a given mRNA is unlikely to generate more of the encoded protein, particularly at low ribosome availability. Recent evidence indicates that eukaryotic mRNAs are circularized, potentially allowing terminating ribosomes to preferentially reinitiate on the same transcript. Here we model the effect of ribosome reinitiation on translation and show that, at high levels of reinitiation, protein synthesis rates are dominated by the time required to translate a given transcript. Our model provides a simple mechanistic explanation for many previously enigmatic features of eukaryotic translation, including the negative correlation of both ribosome densities and protein abundance on transcript length, the importance of codon usage in determining protein synthesis rates, and the negative correlation between transcript length and both codon adaptation and 5' mRNA folding energies. In contrast to linear models where translation is largely limited by initiation rates, our model reveals that all three stages of translation-initiation, elongation, and termination/reinitiation-determine protein synthesis rates even at low ribosome availability.
Collapse
Affiliation(s)
- David W. Rogers
- Experimental Evolution Research Group, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany
- * E-mail:
| | - Marvin A. Böttcher
- Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Arne Traulsen
- Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Duncan Greig
- Experimental Evolution Research Group, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Department of Genetics, Evolution, and Environment, University College London, London, United Kingdom
| |
Collapse
|
33
|
Nicholson G, Holmes C. A note on statistical repeatability and study design for high-throughput assays. Stat Med 2017; 36:790-798. [PMID: 27882571 PMCID: PMC5299465 DOI: 10.1002/sim.7175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 09/30/2016] [Accepted: 10/28/2016] [Indexed: 01/10/2023]
Abstract
Characterizing the technical precision of measurements is a necessary stage in the planning of experiments and in the formal sample size calculation for optimal design. Instruments that measure multiple analytes simultaneously, such as in high-throughput assays arising in biomedical research, pose particular challenges from a statistical perspective. The current most popular method for assessing precision of high-throughput assays is by scatterplotting data from technical replicates. Here, we question the statistical rationale of this approach from both an empirical and theoretical perspective, illustrating our discussion using four example data sets from different genomic platforms. We demonstrate that such scatterplots convey little statistical information of relevance and are potentially highly misleading. We present an alternative framework for assessing the precision of high-throughput assays and planning biomedical experiments. Our methods are based on repeatability-a long-established statistical quantity also known as the intraclass correlation coefficient. We provide guidance and software for estimation and visualization of repeatability of high-throughput assays, and for its incorporation into study design. © 2016 The Authors. Statistics in Medicine Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- George Nicholson
- Department of StatisticsUniversity of Oxford24‐29 St GilesOxfordOX1 3LBU.K.
| | - Chris Holmes
- Department of StatisticsUniversity of Oxford24‐29 St GilesOxfordOX1 3LBU.K.
| |
Collapse
|
34
|
Multiple Transcript Properties Related to Translation Affect mRNA Degradation Rates in Saccharomyces cerevisiae. G3-GENES GENOMES GENETICS 2016; 6:3475-3483. [PMID: 27633789 PMCID: PMC5100846 DOI: 10.1534/g3.116.032276] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Degradation of mRNA contributes to variation in transcript abundance. Studies of individual mRNAs have shown that both cis and trans factors affect mRNA degradation rates. However, the factors underlying transcriptome-wide variation in mRNA degradation rates are poorly understood. We investigated the contribution of different transcript properties to transcriptome-wide degradation rate variation in the budding yeast, Saccharomyces cerevisiae, using multiple regression analysis. We find that multiple transcript properties are significantly associated with variation in mRNA degradation rates, and that a model incorporating these properties explains ∼50% of the genome-wide variance. Predictors of mRNA degradation rates include transcript length, ribosome density, biased codon usage, and GC content of the third position in codons. To experimentally validate these factors, we studied individual transcripts expressed from identical promoters. We find that decreasing ribosome density by mutating the first translational start site of a transcript increases its degradation rate. Using coding sequence variants of green fluorescent protein (GFP) that differ only at synonymous sites, we show that increased GC content of the third position of codons results in decreased rates of mRNA degradation. Thus, in steady-state conditions, a large fraction of genome-wide variation in mRNA degradation rates is determined by inherent properties of transcripts, many of which are related to translation, rather than specific regulatory mechanisms.
Collapse
|
35
|
Dasgupta S, Yang C, Castro LM, Tashima AK, Ferro ES, Moir RD, Willis IM, Fricker LD. Analysis of the Yeast Peptidome and Comparison with the Human Peptidome. PLoS One 2016; 11:e0163312. [PMID: 27685651 PMCID: PMC5042401 DOI: 10.1371/journal.pone.0163312] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/07/2016] [Indexed: 12/22/2022] Open
Abstract
Peptides function as signaling molecules in species as diverse as humans and yeast. Mass spectrometry-based peptidomics techniques provide a relatively unbiased method to assess the peptidome of biological samples. In the present study, we used a quantitative peptidomic technique to characterize the peptidome of the yeast Saccharomyces cerevisiae and compare it to the peptidomes of mammalian cell lines and tissues. Altogether, 297 yeast peptides derived from 75 proteins were identified. The yeast peptides are similar to those of the human peptidome in average size and amino acid composition. Inhibition of proteasome activity with either bortezomib or epoxomicin led to decreased levels of some yeast peptides, suggesting that these peptides are generated by the proteasome. Approximately 30% of the yeast peptides correspond to the N- or C-terminus of the protein; the human peptidome is also highly represented in N- or C-terminal protein fragments. Most yeast and humans peptides are derived from a subset of abundant proteins, many with functions involving cellular metabolism or protein synthesis and folding. Of the 75 yeast proteins that give rise to peptides, 24 have orthologs that give rise to human and/or mouse peptides and for some, the same region of the proteins are found in the human, mouse, and yeast peptidomes. Taken together, these results support the hypothesis that intracellular peptides may have specific and conserved biological functions.
Collapse
Affiliation(s)
- Sayani Dasgupta
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
| | - Ciyu Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, United States of America
| | - Leandro M. Castro
- Biomedical Science Institute, Campus on the São Paulo Coast, São Paulo State University, São Vicente, 11330–900, SP, Brazil
| | - Alexandre K. Tashima
- Department of Biochemistry, Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, SP, 04023–901, SP, Brazil
| | - Emer S. Ferro
- Department of Pharmacology, Biomedical Science Institute, University of São Paulo, São Paulo, 05508–000, SP, Brazil
| | - Robyn D. Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
- Department of Systems & Computational Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
| | - Lloyd D. Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America
- * E-mail:
| |
Collapse
|
36
|
Tramantano M, Sun L, Au C, Labuz D, Liu Z, Chou M, Shen C, Luk E. Constitutive turnover of histone H2A.Z at yeast promoters requires the preinitiation complex. eLife 2016; 5. [PMID: 27438412 PMCID: PMC4995100 DOI: 10.7554/elife.14243] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/12/2016] [Indexed: 12/17/2022] Open
Abstract
The assembly of the preinitiation complex (PIC) occurs upstream of the +1 nucleosome which, in yeast, obstructs the transcription start site and is frequently assembled with the histone variant H2A.Z. To understand the contribution of the transcription machinery in the disassembly of the +1 H2A.Z nucleosome, conditional mutants were used to block PIC assembly. A quantitative ChIP-seq approach, which allows detection of global occupancy change, was employed to measure H2A.Z occupancy. Blocking PIC assembly resulted in promoter-specific H2A.Z accumulation, indicating that the PIC is required to evict H2A.Z. By contrast, H2A.Z eviction was unaffected upon depletion of INO80, a remodeler previously reported to displace nucleosomal H2A.Z. Robust PIC-dependent H2A.Z eviction was observed at active and infrequently transcribed genes, indicating that constitutive H2A.Z turnover is a general phenomenon. Finally, sites with strong H2A.Z turnover precisely mark transcript starts, providing a new metric for identifying cryptic and alternative sites of initiation. DOI:http://dx.doi.org/10.7554/eLife.14243.001 To fit the genetic information of an animal, yeast or other eukaryote into cells, DNA is tightly wound around proteins called histones to form repeating units known as nucleosomes. However, this tight winding prevents proteins from accessing the DNA, and so prevents gene transcription – the first stage of producing the molecules encoded by a gene. For transcription to take place, nucleosomes at DNA sequences called promoters must be reorganized and disassembled, thereby allowing proteins to bind to and engage these sequences and to turn nearby genes on. H2A is a histone protein that is found in the majority of nucleosomes in yeast cells. A different form of this histone – called H2A.Z – is found in nucleosomes near the promoter of almost every gene. It is thought that nucleosomes that contain H2A.Z are recognized and disassembled as the gene turns on, but it is unclear how this happens. To investigate how H2A.Z nucleosomes are disassembled, Tramantano et al. depleted yeast cells of various proteins thought to play a role in the disassembly process. This indicated that the proteins that transcribe genes play crucial roles in the process of disassembling the H2A.Z nucleosomes, because H2A.Z accumulated at promoters in cells that are depleted of these proteins. Further investigation revealed that disassembled H2A.Z nucleosomes are reassembled with H2A histones, before being converted back to the H2A.Z form by an enzyme called SWR1. This turnover of H2A.Z was seen at active genes and those that are infrequently transcribed, suggesting that it is a general phenomenon. Tramantano et al. also found that the turnover rate of H2A.Z can be used to accurately predict the sites in the DNA where transcription starts. This observation could therefore help to identify previously unknown transcription start sites. Future work could address further questions about how H2A.Z nucleosomes are disassembled. For example, what is the mechanical force that drives this process? And at what step of the transcription process does it occur? DOI:http://dx.doi.org/10.7554/eLife.14243.002
Collapse
Affiliation(s)
- Michael Tramantano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Lu Sun
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Christy Au
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Daniel Labuz
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Zhimin Liu
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Mindy Chou
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Chen Shen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Ed Luk
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| |
Collapse
|
37
|
Jellyfish Bioactive Compounds: Methods for Wet-Lab Work. Mar Drugs 2016; 14:md14040075. [PMID: 27077869 PMCID: PMC4849079 DOI: 10.3390/md14040075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 02/03/2016] [Accepted: 02/18/2016] [Indexed: 11/17/2022] Open
Abstract
The study of bioactive compounds from marine animals has provided, over time, an endless source of interesting molecules. Jellyfish are commonly targets of study due to their toxic proteins. However, there is a gap in reviewing successful wet-lab methods employed in these animals, which compromises the fast progress in the detection of related biomolecules. Here, we provide a compilation of the most effective wet-lab methodologies for jellyfish venom extraction prior to proteomic analysis-separation, identification and toxicity assays. This includes SDS-PAGE, 2DE, gel chromatography, HPLC, DEAE, LC-MS, MALDI, Western blot, hemolytic assay, antimicrobial assay and protease activity assay. For a more comprehensive approach, jellyfish toxicity studies should further consider transcriptome sequencing. We reviewed such methodologies and other genomic techniques used prior to the deep sequencing of transcripts, including RNA extraction, construction of cDNA libraries and RACE. Overall, we provide an overview of the most promising methods and their successful implementation for optimizing time and effort when studying jellyfish.
Collapse
|
38
|
Petibon C, Parenteau J, Catala M, Elela SA. Introns regulate the production of ribosomal proteins by modulating splicing of duplicated ribosomal protein genes. Nucleic Acids Res 2016; 44:3878-91. [PMID: 26945043 PMCID: PMC4856989 DOI: 10.1093/nar/gkw140] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/25/2016] [Indexed: 01/30/2023] Open
Abstract
Most budding yeast introns exist in the many duplicated ribosomal protein genes (RPGs) and it has been posited that they remain there to modulate the expression of RPGs and cell growth in response to stress. However, the mechanism by which introns regulate the expression of RPGs and their impact on the synthesis of ribosomal proteins remain unclear. In this study, we show that introns determine the ratio of ribosomal protein isoforms through asymmetric paralog-specific regulation of splicing. Exchanging the introns and 3′ untranslated regions of the duplicated RPS9 genes altered the splicing efficiency and changed the ratio of the ribosomal protein isoforms. Mutational analysis of the RPS9 genes indicated that splicing is regulated by variations in the intron structure and the 3′ untranslated region. Together these data suggest that preferential splicing of duplicated RPGs provides a means for adjusting the ratio of different ribosomal protein isoforms, while maintaining the overall expression level of each ribosomal protein.
Collapse
Affiliation(s)
- Cyrielle Petibon
- Université de Sherbrooke Centre of Excellence in RNA Biology, Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Julie Parenteau
- Université de Sherbrooke Centre of Excellence in RNA Biology, Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Mathieu Catala
- Université de Sherbrooke Centre of Excellence in RNA Biology, Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| | - Sherif Abou Elela
- Université de Sherbrooke Centre of Excellence in RNA Biology, Département de microbiologie et d'infectiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec J1E 4K8, Canada
| |
Collapse
|
39
|
Święciło A. Cross-stress resistance in Saccharomyces cerevisiae yeast--new insight into an old phenomenon. Cell Stress Chaperones 2016; 21:187-200. [PMID: 26825800 PMCID: PMC4786536 DOI: 10.1007/s12192-016-0667-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/27/2015] [Accepted: 01/04/2016] [Indexed: 12/20/2022] Open
Abstract
Acquired stress resistance is the result of mild stress causing the acquisition of resistance to severe stress of the same or a different type. The mechanism of "same-stress" resistance (resistance to a second, strong stress after mild primary stress of the same type) probably depends on the activation of defense and repair mechanisms specific for a particular type of stress, while cross-stress resistance (i.e., resistance to a second, strong stress after a different type of mild primary stress) is the effect of activation of both a specific and general stress response program, which in Saccharomyces cerevisiae yeast is known as the environmental stress response (ESR). Advancements in research techniques have made it possible to study the mechanism of cross-stress resistance at various levels of cellular organization: stress signal transduction pathways, regulation of gene expression, and transcription or translation processes. As a result of this type of research, views on the cross-stress protection mechanism have been reconsidered. It was originally thought that cross-stress resistance, irrespective of the nature of the two stresses, was determined by universal mechanisms, i.e., the same mechanisms within the general stress response. They are now believed to be more specific and strictly dependent on the features of the first stress.
Collapse
Affiliation(s)
- Agata Święciło
- Faculty of Agrobioengineering, Department of Environmental Microbiology, University of Life Sciences in Lublin, Leszczynskiego 7, 20-069, Lublin, Poland.
| |
Collapse
|
40
|
Smith C, Lari A, Derrer CP, Ouwehand A, Rossouw A, Huisman M, Dange T, Hopman M, Joseph A, Zenklusen D, Weis K, Grunwald D, Montpetit B. In vivo single-particle imaging of nuclear mRNA export in budding yeast demonstrates an essential role for Mex67p. J Cell Biol 2015; 211:1121-30. [PMID: 26694837 PMCID: PMC4687877 DOI: 10.1083/jcb.201503135] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 09/17/2015] [Indexed: 11/22/2022] Open
Abstract
Many messenger RNA export proteins have been identified; yet the spatial and temporal activities of these proteins and how they determine directionality of messenger ribonucleoprotein (mRNP) complex export from the nucleus remain largely undefined. Here, the bacteriophage PP7 RNA-labeling system was used in Saccharomyces cerevisiae to follow single-particle mRNP export events with high spatial precision and temporal resolution. These data reveal that mRNP export, consisting of nuclear docking, transport, and cytoplasmic release from a nuclear pore complex (NPC), is fast (∼ 200 ms) and that upon arrival in the cytoplasm, mRNPs are frequently confined near the nuclear envelope. Mex67p functions as the principal mRNP export receptor in budding yeast. In a mex67-5 mutant, delayed cytoplasmic release from NPCs and retrograde transport of mRNPs was observed. This proves an essential role for Mex67p in cytoplasmic mRNP release and directionality of transport.
Collapse
Affiliation(s)
- Carlas Smith
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Azra Lari
- Department of Cell Biology, University of Alberta, T6G 2H7 Edmonton, Alberta, Canada
| | | | - Anette Ouwehand
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605 Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Ammeret Rossouw
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605 Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Maximiliaan Huisman
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605 Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Thomas Dange
- Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Mark Hopman
- Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Aviva Joseph
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Daniel Zenklusen
- Departement de Biochimie et Medecine Moleculaire, Universite de Montreal, H3T 1J4 Montreal, Quebec, Canada
| | - Karsten Weis
- Department of Biology, Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland Department of Cell and Developmental Biology, University of California, Berkeley, Berkeley, CA 94720
| | - David Grunwald
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605 Department Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605 Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, Netherlands
| | - Ben Montpetit
- Department of Cell Biology, University of Alberta, T6G 2H7 Edmonton, Alberta, Canada
| |
Collapse
|
41
|
Bersani F, Lee E, Kharchenko PV, Xu AW, Liu M, Xega K, MacKenzie OC, Brannigan BW, Wittner BS, Jung H, Ramaswamy S, Park PJ, Maheswaran S, Ting DT, Haber DA. Pericentromeric satellite repeat expansions through RNA-derived DNA intermediates in cancer. Proc Natl Acad Sci U S A 2015; 112:15148-53. [PMID: 26575630 PMCID: PMC4679016 DOI: 10.1073/pnas.1518008112] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aberrant transcription of the pericentromeric human satellite II (HSATII) repeat is present in a wide variety of epithelial cancers. In deriving experimental systems to study its deregulation, we observed that HSATII expression is induced in colon cancer cells cultured as xenografts or under nonadherent conditions in vitro, but it is rapidly lost in standard 2D cultures. Unexpectedly, physiological induction of endogenous HSATII RNA, as well as introduction of synthetic HSATII transcripts, generated cDNA intermediates in the form of DNA/RNA hybrids. Single molecule sequencing of tumor xenografts showed that HSATII RNA-derived DNA (rdDNA) molecules are stably incorporated within pericentromeric loci. Suppression of RT activity using small molecule inhibitors reduced HSATII copy gain. Analysis of whole-genome sequencing data revealed that HSATII copy number gain is a common feature in primary human colon tumors and is associated with a lower overall survival. Together, our observations suggest that cancer-associated derepression of specific repetitive sequences can promote their RNA-driven genomic expansion, with potential implications on pericentromeric architecture.
Collapse
Affiliation(s)
- Francesca Bersani
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129
| | - Eunjung Lee
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115; Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115
| | - Peter V Kharchenko
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115; Hematology/Oncology Program, Children's Hospital, Boston, MA 02115
| | - Andrew W Xu
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115
| | - Mingzhu Liu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129; Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Kristina Xega
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129
| | - Olivia C MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129
| | - Brian W Brannigan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129
| | | | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Peter J Park
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115; Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115; Informatics Program, Children's Hospital, Boston, MA 02115
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114
| | - David T Ting
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114;
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129; Howard Hughes Medical Institute, Chevy Chase, MD 20815; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114;
| |
Collapse
|
42
|
Jonsson P, Coarfa C, Mesmar F, Raz T, Rajapakshe K, Thompson JF, Gunaratne PH, Williams C. Single-Molecule Sequencing Reveals Estrogen-Regulated Clinically Relevant lncRNAs in Breast Cancer. Mol Endocrinol 2015; 29:1634-45. [PMID: 26426411 DOI: 10.1210/me.2015-1153] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Estrogen receptor (ER)α-positive tumors are commonly treated with ERα antagonists or inhibitors of estrogen synthesis, but most tumors develop resistance, and we need to better understand the pathways that underlie the proliferative and tumorigenic role of this estrogen-activated transcription factor. We here present the first single-molecule sequencing of the estradiol-induced ERα transcriptome in the luminal A-type human breast cancer cell lines MCF7 and T47D. Sequencing libraries were prepared from the polyadenylated RNA fraction after 8 hours of estrogen or vehicle treatment. Single-molecule sequencing was carried out in biological and technical replicates and differentially expressed genes were defined and analyzed for enriched processes. Correlation analysis with clinical expression and survival were performed, and follow-up experiments carried out using time series, chromatin immunoprecipitation and quantitative real-time PCR. We uncovered that ERα in addition to regulating approximately 2000 protein-coding genes, also regulated up to 1000 long noncoding RNAs (lncRNAs). Most of these were up-regulated, and 178 lncRNAs were regulated in both cell lines. We demonstrate that Long Intergenic Non-protein Coding RNA 1016 (LINC01016) and LINC00160 are direct transcriptional targets of ERα, correlate with ERα expression in clinical samples, and show prognostic significance in relation to breast cancer survival. We show that silencing of LINC00160 results in reduced proliferation, demonstrating that lncRNA expression have functional consequences. Our findings suggest that ERα regulation of lncRNAs is clinically relevant and that their functions and potential use as biomarkers for endocrine response are important to explore.
Collapse
Affiliation(s)
- Philip Jonsson
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Cristian Coarfa
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Fahmi Mesmar
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Tal Raz
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Kimal Rajapakshe
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - John F Thompson
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Preethi H Gunaratne
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| | - Cecilia Williams
- Center for Nuclear Receptors and Cell Signaling (P.J., F.M., C.W.), Department of Biology and Biochemistry, and Department of Biology and Biochemistry (P.H.G.), University of Houston, Houston, Texas 77204; Molecular and Human Genetics (C.C., K.R.) and Human Genome Sequencing Center (P.H.G.), Baylor College of Medicine, Houston, Texas 77030; Helicos Biosciences (T.R., J.F.T.), Cambridge, Massachusetts 02139; SciLifeLab, School of Biotechnology (C.W.), The Royal Institute of Technology-KTH, 17121 Solna, Sweden; and Department of Biosciences and Nutrition (C.W.), Novum, Karolinska Institutet, 14183 Stockholm, Sweden
| |
Collapse
|
43
|
Affiliation(s)
- Hu Zhang
- School of Chemical Engineering; University of Adelaide; Adelaide Australia
| | - Xiaolin Cui
- School of Chemical Engineering; University of Adelaide; Adelaide Australia
| | - Jingxiu Bi
- School of Chemical Engineering; University of Adelaide; Adelaide Australia
| | - Sheng Dai
- School of Chemical Engineering; University of Adelaide; Adelaide Australia
| | - Haitao Ye
- School of Engineering and Applied Science; Aston University; Birmingham United Kingdom
| |
Collapse
|
44
|
de Las Heras-Rubio A, Perucho L, Paciucci R, Vilardell J, LLeonart ME. Ribosomal proteins as novel players in tumorigenesis. Cancer Metastasis Rev 2015; 33:115-41. [PMID: 24375388 DOI: 10.1007/s10555-013-9460-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ribosome biogenesis is the most demanding energetic and metabolic expenditure of the cell. The nucleolus, a nuclear compartment, coordinates rRNA transcription, maturation, and assembly into ribosome subunits. The transcription process is highly coordinated with ribosome biogenesis. In this context, ribosomal proteins (RPs) play a crucial role. In the last decade, an increasing number of studies have associated RPs with extraribosomal functions related to proliferation. Importantly, the expression of RPs appears to be deregulated in several human disorders due, at least in part, to genetic mutations. Although the deregulation of RPs in human malignancies is commonly observed, a more complex mechanism is believed to be involved, favoring the tumorigenic process, its progression and metastasis. This review explores the roles of the most frequently mutated oncogenes and tumor suppressor genes in human cancer that modulate ribosome biogenesis, including their interaction with RPs. In this regard, we propose a new focus for novel therapies.
Collapse
Affiliation(s)
- A de Las Heras-Rubio
- Oncology and Pathology Group, Institut de Recerca Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | | | | | | |
Collapse
|
45
|
Csárdi G, Franks A, Choi DS, Airoldi EM, Drummond DA. Accounting for experimental noise reveals that mRNA levels, amplified by post-transcriptional processes, largely determine steady-state protein levels in yeast. PLoS Genet 2015; 11:e1005206. [PMID: 25950722 PMCID: PMC4423881 DOI: 10.1371/journal.pgen.1005206] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 04/10/2015] [Indexed: 11/25/2022] Open
Abstract
Cells respond to their environment by modulating protein levels through mRNA transcription and post-transcriptional control. Modest observed correlations between global steady-state mRNA and protein measurements have been interpreted as evidence that mRNA levels determine roughly 40% of the variation in protein levels, indicating dominant post-transcriptional effects. However, the techniques underlying these conclusions, such as correlation and regression, yield biased results when data are noisy, missing systematically, and collinear---properties of mRNA and protein measurements---which motivated us to revisit this subject. Noise-robust analyses of 24 studies of budding yeast reveal that mRNA levels explain more than 85% of the variation in steady-state protein levels. Protein levels are not proportional to mRNA levels, but rise much more rapidly. Regulation of translation suffices to explain this nonlinear effect, revealing post-transcriptional amplification of, rather than competition with, transcriptional signals. These results substantially revise widely credited models of protein-level regulation, and introduce multiple noise-aware approaches essential for proper analysis of many biological phenomena.
Collapse
Affiliation(s)
- Gábor Csárdi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America,
| | - Alexander Franks
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America,
| | - David S. Choi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America,
| | - Edoardo M. Airoldi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America,
- The Broad Institute of Harvard & MIT, Cambridge, Massachusetts, United States of America,
| | - D. Allan Drummond
- Dept. of Biochemistry & Molecular Biology, University of Chicago, Chicago, Illinois, United States of America,
- Dept. of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
46
|
Csárdi G, Franks A, Choi DS, Airoldi EM, Drummond DA. Accounting for experimental noise reveals that mRNA levels, amplified by post-transcriptional processes, largely determine steady-state protein levels in yeast. PLoS Genet 2015. [PMID: 25950722 DOI: 10.5061/dryad.d644f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023] Open
Abstract
Cells respond to their environment by modulating protein levels through mRNA transcription and post-transcriptional control. Modest observed correlations between global steady-state mRNA and protein measurements have been interpreted as evidence that mRNA levels determine roughly 40% of the variation in protein levels, indicating dominant post-transcriptional effects. However, the techniques underlying these conclusions, such as correlation and regression, yield biased results when data are noisy, missing systematically, and collinear---properties of mRNA and protein measurements---which motivated us to revisit this subject. Noise-robust analyses of 24 studies of budding yeast reveal that mRNA levels explain more than 85% of the variation in steady-state protein levels. Protein levels are not proportional to mRNA levels, but rise much more rapidly. Regulation of translation suffices to explain this nonlinear effect, revealing post-transcriptional amplification of, rather than competition with, transcriptional signals. These results substantially revise widely credited models of protein-level regulation, and introduce multiple noise-aware approaches essential for proper analysis of many biological phenomena.
Collapse
Affiliation(s)
- Gábor Csárdi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
| | - Alexander Franks
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
| | - David S Choi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America
| | - Edoardo M Airoldi
- Dept. of Statistics, Harvard University, Cambridge, Massachusetts, United States of America,; The Broad Institute of Harvard & MIT, Cambridge, Massachusetts, United States of America
| | - D Allan Drummond
- Dept. of Biochemistry & Molecular Biology, University of Chicago, Chicago, Illinois, United States of America,; Dept. of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
47
|
Kim WJ, Wittner BS, Amzallag A, Brannigan BW, Ting DT, Ramaswamy S, Maheswaran S, Haber DA. The WTX Tumor Suppressor Interacts with the Transcriptional Corepressor TRIM28. J Biol Chem 2015; 290:14381-90. [PMID: 25882849 DOI: 10.1074/jbc.m114.631945] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Indexed: 02/05/2023] Open
Abstract
WTX encodes a tumor suppressor implicated in the pediatric kidney cancer Wilms tumor and in mesenchymal differentiation with potentially distinct functions in the cytoplasm, at the plasma membrane, and in the nucleus. Although modulating components of the WNT signaling pathway is a proposed function for cytoplasmic and membrane-bound WTX, its nuclear properties are not well understood. Here we report that the transcriptional corepressor TRIM28 is the major binding partner for nuclear WTX. WTX interacted with the coiled coil domain of TRIM28 required for its binding to Krüppel-associated box domains of transcription factors and for its chromatin recruitment through its own coiled coil and proline-rich domains. Knockdown of endogenous WTX reduced the recruitment of TRIM28 to a chromatinized reporter sequence and its ability to repress a target transcript. In mouse embryonic stem cells where TRIM28 plays a major role in repressing endogenous retroviruses and long interspersed elements, knockdown of either TRIM28 or WTX combined with single molecule RNA sequencing revealed a highly significant shared set of differentially regulated transcripts, including derepression of non-coding repetitive sequences and their neighboring protein encoding genes (p < 1e-20). In mesenchymal precursor cells, depletion of WTX and TRIM28 resulted in analogous β-catenin-independent defects in adipogenic and osteogenic differentiation, and knockdown of WTX reduced TRIM28 binding to Pparγ promoter. Together, the physical and functional interaction between WTX and TRIM28 suggests that the nuclear fraction of WTX plays a role in epigenetic silencing, an effect that may contribute to its function as a regulator of cellular differentiation and tumorigenesis.
Collapse
Affiliation(s)
- Woo Jae Kim
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Arnaud Amzallag
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Brian W Brannigan
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - David T Ting
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and Surgery
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| |
Collapse
|
48
|
Franks AM, Csárdi G, Drummond DA, Airoldi EM. Estimating a structured covariance matrix from multi-lab measurements in high-throughput biology. J Am Stat Assoc 2015; 110:27-44. [PMID: 25954056 PMCID: PMC4418505 DOI: 10.1080/01621459.2014.964404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We consider the problem of quantifying the degree of coordination between transcription and translation, in yeast. Several studies have reported a surprising lack of coordination over the years, in organisms as different as yeast and human, using diverse technologies. However, a close look at this literature suggests that the lack of reported correlation may not reflect the biology of regulation. These reports do not control for between-study biases and structure in the measurement errors, ignore key aspects of how the data connect to the estimand, and systematically underestimate the correlation as a consequence. Here, we design a careful meta-analysis of 27 yeast data sets, supported by a multilevel model, full uncertainty quantification, a suite of sensitivity analyses and novel theory, to produce a more accurate estimate of the correlation between mRNA and protein levels-a proxy for coordination. From a statistical perspective, this problem motivates new theory on the impact of noise, model mis-specifications and non-ignorable missing data on estimates of the correlation between high dimensional responses. We find that the correlation between mRNA and protein levels is quite high under the studied conditions, in yeast, suggesting that post-transcriptional regulation plays a less prominent role than previously thought.
Collapse
|
49
|
Biagioli M, Ferrari F, Mendenhall EM, Zhang Y, Erdin S, Vijayvargia R, Vallabh SM, Solomos N, Manavalan P, Ragavendran A, Ozsolak F, Lee JM, Talkowski ME, Gusella JF, Macdonald ME, Park PJ, Seong IS. Htt CAG repeat expansion confers pleiotropic gains of mutant huntingtin function in chromatin regulation. Hum Mol Genet 2015; 24:2442-57. [PMID: 25574027 DOI: 10.1093/hmg/ddv006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022] Open
Abstract
The CAG repeat expansion in the Huntington's disease gene HTT extends a polyglutamine tract in mutant huntingtin that enhances its ability to facilitate polycomb repressive complex 2 (PRC2). To gain insight into this dominant gain of function, we mapped histone modifications genome-wide across an isogenic panel of mouse embryonic stem cell (ESC) and neuronal progenitor cell (NPC) lines, comparing the effects of Htt null and different size Htt CAG mutations. We found that Htt is required in ESC for the proper deposition of histone H3K27me3 at a subset of 'bivalent' loci but in NPC it is needed at 'bivalent' loci for both the proper maintenance and the appropriate removal of this mark. In contrast, Htt CAG size, though changing histone H3K27me3, is prominently associated with altered histone H3K4me3 at 'active' loci. The sets of ESC and NPC genes with altered histone marks delineated by the lack of huntingtin or the presence of mutant huntingtin, though distinct, are enriched in similar pathways with apoptosis specifically highlighted for the CAG mutation. Thus, the manner by which huntingtin function facilitates PRC2 may afford mutant huntingtin with multiple opportunities to impinge upon the broader machinery that orchestrates developmentally appropriate chromatin status.
Collapse
Affiliation(s)
- Marta Biagioli
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Yijing Zhang
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Serkan Erdin
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ravi Vijayvargia
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Sonia M Vallabh
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nicole Solomos
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Poornima Manavalan
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ashok Ragavendran
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Fatih Ozsolak
- RaNA Therapeutics, 790 Memorial Drive, Cambridge, MA 02139, USA
| | - Jong Min Lee
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Michael E Talkowski
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - James F Gusella
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA and
| | - Marcy E Macdonald
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA and
| | - Peter J Park
- Center for Biomedical Informatics, Boston, MA 02114, USA Division of Genetics, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ihn Sik Seong
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA, Department of Neurology, Harvard Medical School, Boston, MA 02114, USA,
| |
Collapse
|
50
|
Kondratova VN, Botezatu IV, Shelepov VP, Lichtenstein AV. Satellite DNA transcripts in blood plasma as potential markers of tumor growth. Mol Biol 2014. [DOI: 10.1134/s0026893314060089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|