Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.393
Filter
Add more filters

Publication year range
1.
Cell ; 187(18): 4824-4826, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241742

ABSTRACT

Epigenetic inheritance of heterochromatin requires transfer of parental H3-H4 tetramers to both daughter duplexes during replication. Three recent papers exploit yeast genetics coupled to inheritance assays and AlphaFold2-multimer predictions coupled to biochemistry to reveal that a replisome component (Mrc1/CLASPIN) is an H3-H4 tetramer chaperone important for parental histone transfer to daughters.


Subject(s)
DNA Replication , Deep Learning , Histones , Saccharomyces cerevisiae , Histones/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Heterochromatin/metabolism , Epigenesis, Genetic
2.
Cell ; 187(18): 5010-5028.e24, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39094570

ABSTRACT

Faithful transfer of parental histones to newly replicated daughter DNA strands is critical for inheritance of epigenetic states. Although replication proteins that facilitate parental histone transfer have been identified, how intact histone H3-H4 tetramers travel from the front to the back of the replication fork remains unknown. Here, we use AlphaFold-Multimer structural predictions combined with biochemical and genetic approaches to identify the Mrc1/CLASPIN subunit of the replisome as a histone chaperone. Mrc1 contains a conserved histone-binding domain that forms a brace around the H3-H4 tetramer mimicking nucleosomal DNA and H2A-H2B histones, is required for heterochromatin inheritance, and promotes parental histone recycling during replication. We further identify binding sites for the FACT histone chaperone in Swi1/TIMELESS and DNA polymerase α that are required for heterochromatin inheritance. We propose that Mrc1, in concert with FACT acting as a mobile co-chaperone, coordinates the distribution of parental histones to newly replicated DNA.


Subject(s)
DNA Replication , Epigenesis, Genetic , Heterochromatin , Histones , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Histones/metabolism , Heterochromatin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Histone Chaperones/metabolism , Molecular Chaperones/metabolism , DNA Polymerase I/metabolism , DNA Polymerase I/genetics
3.
Cell ; 186(4): 837-849.e11, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36693376

ABSTRACT

Concomitant with DNA replication, the chromosomal cohesin complex establishes cohesion between newly replicated sister chromatids. Cohesion establishment requires acetylation of conserved cohesin lysine residues by Eco1 acetyltransferase. Here, we explore how cohesin acetylation is linked to DNA replication. Biochemical reconstitution of replication-coupled cohesin acetylation reveals that transient DNA structures, which form during DNA replication, control the acetylation reaction. As polymerases complete lagging strand replication, strand displacement synthesis produces DNA flaps that are trimmed to result in nicked double-stranded DNA. Both flaps and nicks stimulate cohesin acetylation, while subsequent nick ligation to complete Okazaki fragment maturation terminates the acetylation reaction. A flapped or nicked DNA substrate constitutes a transient molecular clue that directs cohesin acetylation to a window behind the replication fork, next to where cohesin likely entraps both sister chromatids. Our results provide an explanation for how DNA replication is linked to sister chromatid cohesion establishment.


Subject(s)
Chromatids , Saccharomyces cerevisiae Proteins , Chromatids/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DNA Replication , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA , Acetyltransferases/genetics , Acetyltransferases/metabolism
4.
Cell ; 186(4): 748-763.e15, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36758548

ABSTRACT

Although many prokaryotes have glycolysis alternatives, it's considered as the only energy-generating glucose catabolic pathway in eukaryotes. Here, we managed to create a hybrid-glycolysis yeast. Subsequently, we identified an inositol pyrophosphatase encoded by OCA5 that could regulate glycolysis and respiration by adjusting 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) levels. 5-InsP7 levels could regulate the expression of genes involved in glycolysis and respiration, representing a global mechanism that could sense ATP levels and regulate central carbon metabolism. The hybrid-glycolysis yeast did not produce ethanol during growth under excess glucose and could produce 2.68 g/L free fatty acids, which is the highest reported production in shake flask of Saccharomyces cerevisiae. This study demonstrated the significance of hybrid-glycolysis yeast and determined Oca5 as an inositol pyrophosphatase controlling the balance between glycolysis and respiration, which may shed light on the role of inositol pyrophosphates in regulating eukaryotic metabolism.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Diphosphates/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Inositol Phosphates/genetics , Inositol Phosphates/metabolism , Glycolysis/genetics , Respiration , Pyrophosphatases/metabolism , Glucose/metabolism
5.
Annu Rev Biochem ; 89: 359-388, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31794245

ABSTRACT

The spliceosome removes introns from messenger RNA precursors (pre-mRNA). Decades of biochemistry and genetics combined with recent structural studies of the spliceosome have produced a detailed view of the mechanism of splicing. In this review, we aim to make this mechanism understandable and provide several videos of the spliceosome in action to illustrate the intricate choreography of splicing. The U1 and U2 small nuclear ribonucleoproteins (snRNPs) mark an intron and recruit the U4/U6.U5 tri-snRNP. Transfer of the 5' splice site (5'SS) from U1 to U6 snRNA triggers unwinding of U6 snRNA from U4 snRNA. U6 folds with U2 snRNA into an RNA-based active site that positions the 5'SS at two catalytic metal ions. The branch point (BP) adenosine attacks the 5'SS, producing a free 5' exon. Removal of the BP adenosine from the active site allows the 3'SS to bind, so that the 5' exon attacks the 3'SS to produce mature mRNA and an excised lariat intron.


Subject(s)
DEAD-box RNA Helicases/genetics , RNA Splicing Factors/genetics , RNA Splicing , RNA, Small Nuclear/genetics , Saccharomyces cerevisiae/genetics , Spliceosomes/metabolism , Catalytic Domain , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/metabolism , Exons , Humans , Introns , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Structure, Secondary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/ultrastructure
6.
Annu Rev Biochem ; 89: 103-133, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32176524

ABSTRACT

Cells confront DNA damage in every cell cycle. Among the most deleterious types of DNA damage are DNA double-strand breaks (DSBs), which can cause cell lethality if unrepaired or cancers if improperly repaired. In response to DNA DSBs, cells activate a complex DNA damage checkpoint (DDC) response that arrests the cell cycle, reprograms gene expression, and mobilizes DNA repair factors to prevent the inheritance of unrepaired and broken chromosomes. Here we examine the DDC, induced by DNA DSBs, in the budding yeast model system and in mammals.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA End-Joining Repair , DNA/genetics , Recombinational DNA Repair , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , Humans , Models, Molecular , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Annu Rev Biochem ; 89: 333-358, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31815536

ABSTRACT

Splicing of the precursor messenger RNA, involving intron removal and exon ligation, is mediated by the spliceosome. Together with biochemical and genetic investigations of the past four decades, structural studies of the intact spliceosome at atomic resolution since 2015 have led to mechanistic delineation of RNA splicing with remarkable insights. The spliceosome is proven to be a protein-orchestrated metalloribozyme. Conserved elements of small nuclear RNA (snRNA) constitute the splicing active site with two catalytic metal ions and recognize three conserved intron elements through duplex formation, which are delivered into the splicing active site for branching and exon ligation. The protein components of the spliceosome stabilize the conformation of the snRNA, drive spliceosome remodeling, orchestrate the movement of the RNA elements, and facilitate the splicing reaction. The overall organization of the spliceosome and the configuration of the splicing active site are strictly conserved between human and yeast.


Subject(s)
RNA Splicing Factors/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Spliceosomes/metabolism , Catalytic Domain , Conserved Sequence , Exons , Humans , Introns , Models, Molecular , Nucleic Acid Conformation , Protein Structure, Secondary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/ultrastructure
8.
Annu Rev Biochem ; 89: 637-666, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569522

ABSTRACT

The evolution of eukaryotic genomes has been propelled by a series of gene duplication events, leading to an expansion in new functions and pathways. While duplicate genes may retain some functional redundancy, it is clear that to survive selection they cannot simply serve as a backup but rather must acquire distinct functions required for cellular processes to work accurately and efficiently. Understanding these differences and characterizing gene-specific functions is complex. Here we explore different gene pairs and families within the context of the endoplasmic reticulum (ER), the main cellular hub of lipid biosynthesis and the entry site for the secretory pathway. Focusing on each of the ER functions, we highlight specificities of related proteins and the capabilities conferred to cells through their conservation. More generally, these examples suggest why related genes have been maintained by evolutionary forces and provide a conceptual framework to experimentally determine why they have survived selection.


Subject(s)
Endoplasmic Reticulum/metabolism , Evolution, Molecular , Gene Duplication , Saccharomyces cerevisiae/metabolism , Selection, Genetic , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Antiporters/genetics , Antiporters/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Endoplasmic Reticulum/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
9.
Cell ; 180(5): 928-940.e14, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32109413

ABSTRACT

Covalent modifications to histones are essential for development, establishing distinct and functional chromatin domains from a common genetic sequence. Whereas repressed chromatin is robustly inherited, no mechanism that facilitates inheritance of an activated domain has been described. Here, we report that the Set3C histone deacetylase scaffold Snt1 can act as a prion that drives the emergence and transgenerational inheritance of an activated chromatin state. This prion, which we term [ESI+] for expressed sub-telomeric information, is triggered by transient Snt1 phosphorylation upon cell cycle arrest. Once engaged, the prion reshapes the activity of Snt1 and the Set3C complex, recruiting RNA pol II and interfering with Rap1 binding to activate genes in otherwise repressed sub-telomeric domains. This transcriptional state confers broad resistance to environmental stress, including antifungal drugs. Altogether, our results establish a robust means by which a prion can facilitate inheritance of an activated chromatin state to provide adaptive benefit.


Subject(s)
Chromatin/genetics , Histone Deacetylases/genetics , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Telomere-Binding Proteins/genetics , Transcription Factors/genetics , Cell Cycle Checkpoints/genetics , Histone Code/genetics , Histones/genetics , Phosphorylation/genetics , Prions/genetics , RNA Polymerase II/genetics , Saccharomyces cerevisiae , Shelterin Complex , Telomere/genetics , Transcription, Genetic
10.
Cell ; 181(6): 1380-1394.e18, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32502392

ABSTRACT

Homologous recombination (HR) helps maintain genome integrity, and HR defects give rise to disease, especially cancer. During HR, damaged DNA must be aligned with an undamaged template through a process referred to as the homology search. Despite decades of study, key aspects of this search remain undefined. Here, we use single-molecule imaging to demonstrate that Rad54, a conserved Snf2-like protein found in all eukaryotes, switches the search from the diffusion-based pathways characteristic of the basal HR machinery to an active process in which DNA sequences are aligned via an ATP-dependent molecular motor-driven mechanism. We further demonstrate that Rad54 disrupts the donor template strands, enabling the search to take place within a migrating DNA bubble-like structure that is bound by replication protein A (RPA). Our results reveal that Rad54, working together with RPA, fundamentally alters how DNA sequences are aligned during HR.


Subject(s)
Adenosine Triphosphate/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , DNA/genetics , Homologous Recombination/genetics , Saccharomyces cerevisiae Proteins/genetics , Adenosine Triphosphatases/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Hydrolysis , Saccharomyces cerevisiae/genetics , Sequence Alignment/methods
11.
Cell ; 176(3): 520-534.e25, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661750

ABSTRACT

Elucidating the global and local rules that govern genome-wide, hierarchical chromatin architecture remains a critical challenge. Current high-throughput chromosome conformation capture (Hi-C) technologies have identified large-scale chromatin structural motifs, such as topologically associating domains and looping. However, structural rules at the smallest or nucleosome scale remain poorly understood. Here, we coupled nucleosome-resolved Hi-C technology with simulated annealing-molecular dynamics (SA-MD) simulation to reveal 3D spatial distributions of nucleosomes and their genome-wide orientation in chromatin. Our method, called Hi-CO, revealed distinct nucleosome folding motifs across the yeast genome. Our results uncovered two types of basic secondary structural motifs in nucleosome folding: α-tetrahedron and ß-rhombus analogous to α helix and ß sheet motifs in protein folding. Using mutants and cell-cycle-synchronized cells, we further uncovered motifs with specific nucleosome positioning and orientation coupled to epigenetic features at individual loci. By illuminating molecular-level structure-function relationships in eukaryotic chromatin, our findings establish organizational principles of nucleosome folding.


Subject(s)
Chromatin/ultrastructure , Nucleosomes/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosomes/metabolism , Chromosomes/ultrastructure , Nucleosomes/genetics , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Initiation Site
12.
Cell ; 177(3): 782-796.e27, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955892

ABSTRACT

G protein-coupled receptor (GPCR) signaling is the primary method eukaryotes use to respond to specific cues in their environment. However, the relationship between stimulus and response for each GPCR is difficult to predict due to diversity in natural signal transduction architecture and expression. Using genome engineering in yeast, we constructed an insulated, modular GPCR signal transduction system to study how the response to stimuli can be predictably tuned using synthetic tools. We delineated the contributions of a minimal set of key components via computational and experimental refactoring, identifying simple design principles for rationally tuning the dose response. Using five different GPCRs, we demonstrate how this enables cells and consortia to be engineered to respond to desired concentrations of peptides, metabolites, and hormones relevant to human health. This work enables rational tuning of cell sensing while providing a framework to guide reprogramming of GPCR-based signaling in other systems.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Gene Expression/drug effects , Genetic Engineering , Humans , Pheromones/pharmacology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cell ; 177(3): 697-710.e17, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30982600

ABSTRACT

Yeast ataxin-2, also known as Pbp1 (polyA binding protein-binding protein 1), is an intrinsically disordered protein implicated in stress granule formation, RNA biology, and neurodegenerative disease. To understand the endogenous function of this protein, we identify Pbp1 as a dedicated regulator of TORC1 signaling and autophagy under conditions that require mitochondrial respiration. Pbp1 binds to TORC1 specifically during respiratory growth, but utilizes an additional methionine-rich, low complexity (LC) region to inhibit TORC1. This LC region causes phase separation, forms reversible fibrils, and enables self-association into assemblies required for TORC1 inhibition. Mutants that weaken phase separation in vitro exhibit reduced capacity to inhibit TORC1 and induce autophagy. Loss of Pbp1 leads to mitochondrial dysfunction and reduced fitness during nutritional stress. Thus, Pbp1 forms a condensate in response to respiratory status to regulate TORC1 signaling.


Subject(s)
Carrier Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Amino Acid Sequence , Autophagy/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Methionine/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Domains , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology
14.
Cell ; 177(3): 711-721.e8, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30982603

ABSTRACT

Yeast ataxin-2, also known as Pbp1, senses the activity state of mitochondria in order to regulate TORC1. A domain of Pbp1 required to adapt cells to mitochondrial activity is of low sequence complexity. The low-complexity (LC) domain of Pbp1 forms labile, cross-ß polymers that facilitate phase transition of the protein into liquid-like or gel-like states. Phase transition for other LC domains is reliant upon widely distributed aromatic amino acids. In place of tyrosine or phenylalanine residues prototypically used for phase separation, Pbp1 contains 24 similarly disposed methionine residues. Here, we show that the Pbp1 methionine residues are sensitive to hydrogen peroxide (H2O2)-mediated oxidation in vitro and in living cells. Methionine oxidation melts Pbp1 liquid-like droplets in a manner reversed by methionine sulfoxide reductase enzymes. These observations explain how reversible formation of labile polymers by the Pbp1 LC domain enables the protein to function as a sensor of cellular redox state.


Subject(s)
Carrier Proteins/metabolism , Methionine/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Hydrogen Peroxide/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Methionine/metabolism , Methionine Sulfoxide Reductases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidative Stress/drug effects , Phase Transition , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
15.
Annu Rev Biochem ; 87: 75-100, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29328783

ABSTRACT

RNA polymerase (Pol) III has a specialized role in transcribing the most abundant RNAs in eukaryotic cells, transfer RNAs (tRNAs), along with other ubiquitous small noncoding RNAs, many of which have functions related to the ribosome and protein synthesis. The high energetic cost of producing these RNAs and their central role in protein synthesis underlie the robust regulation of Pol III transcription in response to nutrients and stress by growth regulatory pathways. Downstream of Pol III, signaling impacts posttranscriptional processes affecting tRNA function in translation and tRNA cleavage into smaller fragments that are increasingly attributed with novel cellular activities. In this review, we consider how nutrients and stress control Pol III transcription via its factors and its negative regulator, Maf1. We highlight recent work showing that the composition of the tRNA population and the function of individual tRNAs is dynamically controlled and that unrestrained Pol III transcription can reprogram central metabolic pathways.


Subject(s)
RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Animals , Humans , Models, Biological , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein Conformation , RNA Polymerase III/chemistry , RNA Processing, Post-Transcriptional , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Stress, Physiological , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factor TFIIIB/genetics , Transcription Factor TFIIIB/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
16.
Cell ; 175(7): 1946-1957.e13, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30415839

ABSTRACT

Directed evolution is a powerful approach for engineering biomolecules and understanding adaptation. However, experimental strategies for directed evolution are notoriously labor intensive and low throughput, limiting access to demanding functions, multiple functions in parallel, and the study of molecular evolution in replicate. We report OrthoRep, an orthogonal DNA polymerase-plasmid pair in yeast that stably mutates ∼100,000-fold faster than the host genome in vivo, exceeding the error threshold of genomic replication that causes single-generation extinction. User-defined genes in OrthoRep continuously and rapidly evolve through serial passaging, a highly straightforward and scalable process. Using OrthoRep, we evolved drug-resistant malarial dihydrofolate reductases (DHFRs) in 90 independent replicates. We uncovered a more complex fitness landscape than previously realized, including common adaptive trajectories constrained by epistasis, rare outcomes that avoid a frequent early adaptive mutation, and a suboptimal fitness peak that occasionally traps evolving populations. OrthoRep enables a new paradigm of routine, high-throughput evolution of biomolecular and cellular function.


Subject(s)
Adaptation, Physiological/genetics , Genome, Fungal , Models, Genetic , Mutation Rate , Saccharomyces cerevisiae/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
17.
Cell ; 173(6): 1508-1519.e18, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29754816

ABSTRACT

As predicted by the notion that sister chromatid cohesion is mediated by entrapment of sister DNAs inside cohesin rings, there is perfect correlation between co-entrapment of circular minichromosomes and sister chromatid cohesion. In most cells where cohesin loads without conferring cohesion, it does so by entrapment of individual DNAs. However, cohesin with a hinge domain whose positively charged lumen is neutralized loads and moves along chromatin despite failing to entrap DNAs. Thus, cohesin engages chromatin in non-topological, as well as topological, manners. Since hinge mutations, but not Smc-kleisin fusions, abolish entrapment, DNAs may enter cohesin rings through hinge opening. Mutation of three highly conserved lysine residues inside the Smc1 moiety of Smc1/3 hinges abolishes all loading without affecting cohesin's recruitment to CEN loading sites or its ability to hydrolyze ATP. We suggest that loading and translocation are mediated by conformational changes in cohesin's hinge driven by cycles of ATP hydrolysis.


Subject(s)
Cell Cycle Proteins/chemistry , Chromatids/chemistry , Chromosomal Proteins, Non-Histone/chemistry , DNA/chemistry , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Chromatin/chemistry , Humans , Hydrolysis , Lysine/chemistry , Mice , Mutation , Nuclear Proteins/genetics , Protein Conformation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Cohesins
18.
Cell ; 174(6): 1549-1558.e14, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100189

ABSTRACT

Engineering microorganisms for production of fuels and chemicals often requires major re-programming of metabolism to ensure high flux toward the product of interest. This is challenging, as millions of years of evolution have resulted in establishment of tight regulation of metabolism for optimal growth in the organism's natural habitat. Here, we show through metabolic engineering that it is possible to alter the metabolism of Saccharomyces cerevisiae from traditional ethanol fermentation to a pure lipogenesis metabolism, resulting in high-level production of free fatty acids. Through metabolic engineering and process design, we altered subcellular metabolic trafficking, fine-tuned NADPH and ATP supply, and decreased carbon flux to biomass, enabling production of 33.4 g/L extracellular free fatty acids. We further demonstrate that lipogenesis metabolism can replace ethanol fermentation by deletion of pyruvate decarboxylase enzymes followed by adaptive laboratory evolution. Genome sequencing of evolved strains showed that pyruvate kinase mutations were essential for this phenotype.


Subject(s)
Fatty Acids, Nonesterified/biosynthesis , Metabolic Engineering , Saccharomyces cerevisiae/metabolism , Acetyl Coenzyme A/metabolism , Glucose/metabolism , Glycolysis , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lipogenesis , NADP/metabolism , Pentose Phosphate Pathway/genetics , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
19.
Cell ; 172(5): 910-923.e16, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474919

ABSTRACT

To better understand the gene regulatory mechanisms that program developmental processes, we carried out simultaneous genome-wide measurements of mRNA, translation, and protein through meiotic differentiation in budding yeast. Surprisingly, we observed that the levels of several hundred mRNAs are anti-correlated with their corresponding protein products. We show that rather than arising from canonical forms of gene regulatory control, the regulation of at least 380 such cases, or over 8% of all measured genes, involves temporally regulated switching between production of a canonical, translatable transcript and a 5' extended isoform that is not efficiently translated into protein. By this pervasive mechanism for the modulation of protein levels through a natural developmental program, a single transcription factor can coordinately activate and repress protein synthesis for distinct sets of genes. The distinction is not based on whether or not an mRNA is induced but rather on the type of transcript produced.


Subject(s)
Meiosis/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Gene Expression Regulation, Fungal , Genes, Fungal , Models, Biological , Molecular Sequence Annotation , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/metabolism
20.
Cell ; 175(3): 780-795.e15, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30318142

ABSTRACT

During mitosis, chromatin condensation shapes chromosomes as separate, rigid, and compact sister chromatids to facilitate their segregation. Here, we show that, unlike wild-type yeast chromosomes, non-chromosomal DNA circles and chromosomes lacking a centromere fail to condense during mitosis. The centromere promotes chromosome condensation strictly in cis through recruiting the kinases Aurora B and Bub1, which trigger the autonomous condensation of the entire chromosome. Shugoshin and the deacetylase Hst2 facilitated spreading the condensation signal to the chromosome arms. Targeting Aurora B to DNA circles or centromere-ablated chromosomes or releasing Shugoshin from PP2A-dependent inhibition bypassed the centromere requirement for condensation and enhanced the mitotic stability of DNA circles. Our data indicate that yeast cells license the chromosome-autonomous condensation of their chromatin in a centromere-dependent manner, excluding from this process non-centromeric DNA and thereby inhibiting their propagation.


Subject(s)
Centromere/genetics , Chromosomes, Fungal/genetics , Mitosis , Saccharomyces cerevisiae/genetics , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL