Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37961445

ABSTRACT

Genome differential positioning within interphase nuclei remains poorly explored. We extended and validated TSA-seq to map genomic regions near nucleoli and pericentric heterochromatin in four human cell lines. Our study confirmed that smaller chromosomes localize closer to nucleoli but further deconvolved this by revealing a preference for chromosome arms below 36-46 Mbp in length. We identified two lamina associated domain subsets through their differential nuclear lamina versus nucleolar positioning in different cell lines which showed distinctive patterns of DNA replication timing and gene expression across all cell lines. Unexpectedly, active, nuclear speckle-associated genomic regions were found near typically repressive nuclear compartments, which is attributable to the close proximity of nuclear speckles and nucleoli in some cell types, and association of centromeres with nuclear speckles in hESCs. Our study points to a more complex and variable nuclear genome organization than suggested by current models, as revealed by our TSA-seq methodology.

2.
FASEB J ; 36(11): e22588, 2022 11.
Article in English | MEDLINE | ID: mdl-36287614

ABSTRACT

This conference brought together leaders in the investigation of various bodies that populate the nucleus, a field that complements research on chromosome biology. These nuclear bodies had been receiving increasing attention as hubs of genome activity and the new findings reported at the conference strongly confirmed and significantly expanded this principle.


Subject(s)
Genome , Nuclear Bodies , Nova Scotia , Chromosomes/genetics , Genomics
3.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34353903

ABSTRACT

Vertebrate mammals express a protein called Ki-67 which is most widely known as a clinically useful marker of highly proliferative cells. Previous studies of human cells indicated that acute depletion of Ki-67 can elicit a delay at the G1/S boundary of the cell cycle, dependent on induction of the checkpoint protein p21. Consistent with those observations, we show here that acute Ki-67 depletion causes hallmarks of DNA damage, and the damage occurs even in the absence of checkpoint signaling. This damage is not observed in cells traversing S phase but is instead robustly detected in mitotic cells. The C-terminal chromatin-binding domain of Ki-67 is necessary and sufficient to protect cells from this damage. We also observe synergistic effects when Ki-67 and p53 are simultaneously depleted, resulting in increased levels of chromosome bridges at anaphase, followed by the appearance of micronuclei. Therefore, these studies identify the C terminus of Ki-67 as an important module for genome stability.


Subject(s)
Chromatin/metabolism , Chromosomes, Human , Ki-67 Antigen/metabolism , Tumor Suppressor Protein p53/metabolism , Anaphase , Binding Sites , Cell Line , DNA Damage , Genomic Instability , Humans , Ki-67 Antigen/genetics , Mitosis , Protein Domains , Tumor Suppressor Protein p53/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
4.
Biochim Biophys Acta Gene Regul Mech ; 1864(1): 194666, 2021 01.
Article in English | MEDLINE | ID: mdl-33307247

ABSTRACT

Chromatin is a dynamic structure composed of DNA, RNA, and proteins, regulating storage and expression of the genetic material in the nucleus. Heterochromatin plays a crucial role in driving the three-dimensional arrangement of the interphase genome, and in preserving genome stability by maintaining a subset of the genome in a silent state. Spatial genome organization contributes to normal patterns of gene function and expression, and is therefore of broad interest. Mammalian heterochromatin, the focus of this review, mainly localizes at the nuclear periphery, forming Lamina-associated domains (LADs), and at the nucleolar periphery, forming Nucleolus-associated domains (NADs). Together, these regions comprise approximately one-half of mammalian genomes, and most but not all loci within these domains are stochastically placed at either of these two locations after exit from mitosis at each cell cycle. Excitement about the role of these heterochromatic domains in early development has recently been heightened by the discovery that LADs appear at some loci in the preimplantation mouse embryo prior to other chromosomal features like compartmental identity and topologically-associated domains (TADs). While LADs have been extensively studied and mapped during cellular differentiation and early embryonic development, NADs have been less thoroughly studied. Here, we summarize pioneering studies of NADs and LADs, more recent advances in our understanding of cis/trans-acting factors that mediate these localizations, and discuss the functional significance of these associations.


Subject(s)
Cell Nucleolus/metabolism , Genome, Human , Genomic Instability , Heterochromatin/metabolism , Animals , Cell Nucleolus/genetics , Heterochromatin/genetics , Humans
5.
Chromosoma ; 129(2): 121-139, 2020 06.
Article in English | MEDLINE | ID: mdl-32219510

ABSTRACT

Heterochromatin in eukaryotic interphase cells frequently localizes to the nucleolar periphery (nucleolus-associated domains (NADs)) and the nuclear lamina (lamina-associated domains (LADs)). Gene expression in somatic cell NADs is generally low, but NADs have not been characterized in mammalian stem cells. Here, we generated the first genome-wide map of NADs in mouse embryonic stem cells (mESCs) via deep sequencing of chromatin associated with biochemically purified nucleoli. As we had observed in mouse embryonic fibroblasts (MEFs), the large type I subset of NADs overlaps with constitutive LADs and is enriched for features of constitutive heterochromatin, including late replication timing and low gene density and expression levels. Conversely, the type II NAD subset overlaps with loci that are not lamina-associated, but in mESCs, type II NADs are much less abundant than in MEFs. mESC NADs are also much less enriched in H3K27me3 modified regions than are NADs in MEFs. Additionally, comparision of MEF and mESC NADs revealed enrichment of developmentally regulated genes in cell-type-specific NADs. Together, these data indicate that NADs are a developmentally dynamic component of heterochromatin. These studies implicate association with the nucleolar periphery as a mechanism for developmentally regulated gene expression and will facilitate future studies of NADs during mESC differentiation.


Subject(s)
Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Chromosome Mapping , Computational Biology/methods , Fibroblasts , Gene Expression , Gene Ontology , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , In Situ Hybridization, Fluorescence , Mice , Nuclear Lamina
6.
Genome Res ; 29(8): 1235-1249, 2019 08.
Article in English | MEDLINE | ID: mdl-31201210

ABSTRACT

In interphase eukaryotic cells, almost all heterochromatin is located adjacent to the nucleolus or to the nuclear lamina, thus defining nucleolus-associated domains (NADs) and lamina-associated domains (LADs), respectively. Here, we determined the first genome-scale map of murine NADs in mouse embryonic fibroblasts (MEFs) via deep sequencing of chromatin associated with purified nucleoli. We developed a Bioconductor package called NADfinder and demonstrated that it identifies NADs more accurately than other peak-calling tools, owing to its critical feature of chromosome-level local baseline correction. We detected two distinct classes of NADs. Type I NADs associate frequently with both the nucleolar periphery and the nuclear lamina, and generally display characteristics of constitutive heterochromatin, including late DNA replication, enrichment of H3K9me3, and little gene expression. In contrast, Type II NADs associate with nucleoli but do not overlap with LADs. Type II NADs tend to replicate earlier, display greater gene expression, and are more often enriched in H3K27me3 than Type I NADs. The nucleolar associations of both classes of NADs were confirmed via DNA-FISH, which also detected Type I but not Type II probes enriched at the nuclear lamina. Type II NADs are enriched in distinct gene classes, including factors important for differentiation and development. In keeping with this, we observed that a Type II NAD is developmentally regulated, and present in MEFs but not in undifferentiated embryonic stem (ES) cells.


Subject(s)
Cell Nucleolus/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Genome , Heterochromatin/classification , Animals , Cell Nucleolus/ultrastructure , Cells, Cultured , Chromosome Mapping/methods , DNA Replication , Embryo, Mammalian , Fibroblasts/ultrastructure , Heterochromatin/chemistry , Heterochromatin/ultrastructure , Histones/genetics , Histones/metabolism , In Situ Hybridization, Fluorescence , Mice , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure
7.
Curr Genet ; 65(2): 371-377, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30478690

ABSTRACT

In eukaryotes, genomic DNA is packaged into the nucleus together with histone proteins, forming chromatin. The fundamental repeating unit of chromatin is the nucleosome, a naturally symmetric structure that wraps DNA and is the substrate for numerous regulatory post-translational modifications. However, the biological significance of nucleosomal symmetry until recently had been unexplored. To investigate this issue, we developed an obligate pair of histone H3 heterodimers, a novel genetic tool that allowed us to modulate modification sites on individual H3 molecules within nucleosomes in vivo. We used these constructs for molecular genetic studies, for example demonstrating that H3K36 methylation on a single H3 molecule per nucleosome in vivo is sufficient to restrain cryptic transcription. We also used asymmetric nucleosomes for mass spectrometric analysis of dependency relationships among histone modifications. Furthermore, we extended this system to the centromeric H3 isoform (Cse4/CENP-A), gaining insights into centromeric nucleosomal symmetry and structure. In this review, we summarize our findings and discuss the utility of this novel approach.


Subject(s)
Genomics , Histones/genetics , Histones/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Centromere/genetics , Centromere/metabolism , Gene Expression Regulation , Genomics/methods , Histones/chemistry , Mutation , Nucleosomes/chemistry , Protein Multimerization , Structure-Activity Relationship , Transcription, Genetic
8.
Curr Genet ; 65(2): 379-380, 2019 04.
Article in English | MEDLINE | ID: mdl-30569238

ABSTRACT

In the original publication, Fig. 1 was incorrectly published. The amino acid sequence was shifted to the left relative to the rest of the diagram in the published version and the corrected figure is given here.

9.
Elife ; 72018 08 23.
Article in English | MEDLINE | ID: mdl-30136924

ABSTRACT

Nucleosomes contain two copies of each core histone, held together by a naturally symmetric, homodimeric histone H3-H3 interface. This symmetry has complicated efforts to determine the regulatory potential of this architecture. Through molecular design and in vivo selection, we recently generated obligately heterodimeric H3s, providing a powerful tool for discovery of the degree to which nucleosome symmetry regulates chromosomal functions in living cells (Ichikawa et al., 2017). We now have extended this tool to the centromeric H3 isoform (Cse4/CENP-A) in budding yeast. These studies indicate that a single Cse4 N- or C-terminal extension per pair of Cse4 molecules is sufficient for kinetochore function, and validate previous experiments indicating that an octameric centromeric nucleosome is required for viability in this organism. These data also support the generality of the H3 asymmetric interface for probing general questions in chromatin biology.


Subject(s)
Centromere/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Base Sequence , Microbial Viability , Mutation/genetics , Plasmids/metabolism , Protein Domains , Protein Multimerization , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Static Electricity , Temperature
10.
Bio Protoc ; 8(6)2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29644260

ABSTRACT

Nucleosomes are the fundamental unit of eukaryotic chromosome packaging, comprised of 147 bp of DNA wrapped around two molecules of each of the core histone proteins H2A, H2B, H3, and H4. Nucleosomes are symmetrical, with one axis of symmetry centered on the homodimeric interaction between the C-termini of the H3 molecules. To explore the functional consequences of nucleosome symmetry, we designed an obligate pair of H3 heterodimers, termed H3X and H3Y, allowing us to compare cells with single or double H3 alterations. Our biochemical validation of the heterodimeric X-Y interaction included intra-nucleosomal H3 crosslinking using dimethyl suberimidate (DMS). Here, we provide a detailed protocol for the use of DMS to analyze yeast nucleosomes.

11.
Cytoskeleton (Hoboken) ; 75(3): 87-102, 2018 03.
Article in English | MEDLINE | ID: mdl-29360272

ABSTRACT

Traditionally, we imagine that cell division gives rise to two identical daughter cells. Nevertheless, all cell divisions, to some degree, display asymmetry. Asymmetric cell division is defined as the generation of two daughter cells with different physical content and/or developmental potential. Several organelles and cellular components including the centrosome, non-coding RNA, chromatin, and recycling endosomes are involved in the process of asymmetric cell division. Disruption of this important process is known to induce profound defects in development, the immune response, regeneration of tissues, aging, and cancer. Here, we discuss recent advances that expand our understanding of the mechanisms and consequences of asymmetric cell division in vertebrate organisms.


Subject(s)
Asymmetric Cell Division , Mitosis , Stem Cells/cytology , Animals , Humans , Vertebrates
12.
Chromosoma ; 127(2): 175-186, 2018 06.
Article in English | MEDLINE | ID: mdl-29322240

ABSTRACT

Ki-67 protein has been widely used as a proliferation marker for human tumor cells for decades. In recent studies, multiple molecular functions of this large protein have become better understood. Ki-67 has roles in both interphase and mitotic cells, and its cellular distribution dramatically changes during cell cycle progression. These localizations correlate with distinct functions. For example, during interphase, Ki-67 is required for normal cellular distribution of heterochromatin antigens and for the nucleolar association of heterochromatin. During mitosis, Ki-67 is essential for formation of the perichromosomal layer (PCL), a ribonucleoprotein sheath coating the condensed chromosomes. In this structure, Ki-67 acts to prevent aggregation of mitotic chromosomes. Here, we present an overview of functional roles of Ki-67 across the cell cycle and also describe recent experiments that clarify its role in regulating cell cycle progression in human cells.


Subject(s)
Cell Nucleolus/metabolism , Heterochromatin/metabolism , Ki-67 Antigen/genetics , Mitosis , Ribonucleoproteins/genetics , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation , Heterochromatin/ultrastructure , Humans , Interphase , Ki-67 Antigen/metabolism , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Elife ; 62017 09 12.
Article in English | MEDLINE | ID: mdl-28895528

ABSTRACT

The repeating subunit of chromatin, the nucleosome, includes two copies of each of the four core histones, and several recent studies have reported that asymmetrically-modified nucleosomes occur at regulatory elements in vivo. To probe the mechanisms by which histone modifications are read out, we designed an obligate pair of H3 heterodimers, termed H3X and H3Y, which we extensively validated genetically and biochemically. Comparing the effects of asymmetric histone tail point mutants with those of symmetric double mutants revealed that a single methylated H3K36 per nucleosome was sufficient to silence cryptic transcription in vivo. We also demonstrate the utility of this system for analysis of histone modification crosstalk, using mass spectrometry to separately identify modifications on each H3 molecule within asymmetric nucleosomes. The ability to generate asymmetric nucleosomes in vivo and in vitro provides a powerful and generalizable tool to probe the mechanisms by which H3 tails are read out by effector proteins in the cell.


Subject(s)
Histones/analysis , Nucleosomes/chemistry , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae/chemistry , Histones/genetics , Mass Spectrometry/methods , Protein Processing, Post-Translational , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Synthetic Biology/methods
14.
Mol Cell Biol ; 37(17)2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28630280

ABSTRACT

The Ki-67 protein is widely used as a tumor proliferation marker. However, whether Ki-67 affects cell cycle progression has been controversial. Here we demonstrate that depletion of Ki-67 in human hTERT-RPE1, WI-38, IMR90, and hTERT-BJ cell lines and primary fibroblast cells slowed entry into S phase and coordinately downregulated genes related to DNA replication. Some gene expression changes were partially relieved in Ki-67-depleted hTERT-RPE1 cells by codepletion of the Rb checkpoint protein, but more thorough suppression of the transcriptional and cell cycle defects was observed upon depletion of the cell cycle inhibitor p21. Notably, induction of p21 upon depletion of Ki-67 was a consistent hallmark of cell types in which transcription and cell cycle distribution were sensitive to Ki-67; these responses were absent in cells that did not induce p21. Furthermore, upon Ki-67 depletion, a subset of inactive X (Xi) chromosomes in female hTERT-RPE1 cells displayed several features of compromised heterochromatin maintenance, including decreased H3K27me3 and H4K20me1 labeling. These chromatin alterations were limited to Xi chromosomes localized away from the nuclear lamina and were not observed in checkpoint-deficient 293T cells. Altogether, our results indicate that Ki-67 integrates normal S-phase progression and Xi heterochromatin maintenance in p21 checkpoint-proficient human cells.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Heterochromatin/metabolism , Ki-67 Antigen/metabolism , Cell Division/physiology , Cell Line , DNA Replication/physiology , Humans
15.
Mol Biol Cell ; 28(1): 21-29, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27807046

ABSTRACT

Chromatin assembly factor 1 (CAF-1) deposits histones during DNA synthesis. The p150 subunit of human CAF-1 contains an N-terminal domain (p150N) that is dispensable for histone deposition but promotes the localization of specific loci (nucleolar-associated domains [NADs]) and proteins to the nucleolus during interphase. One of the p150N-regulated proteins is proliferation antigen Ki-67, whose depletion also decreases the nucleolar association of NADs. Ki-67 is also a fundamental component of the perichromosomal layer (PCL), a sheath of proteins surrounding condensed chromosomes during mitosis. We show here that a subset of p150 localizes to the PCL during mitosis and that p150N is required for normal levels of Ki-67 accumulation on the PCL. This activity requires the sumoylation-interacting motif within p150N, which is also required for the nucleolar localization of NADs and Ki-67 during interphase. In this manner, p150N coordinates both interphase and mitotic nuclear structures via Ki67.


Subject(s)
Chromatin Assembly Factor-1/metabolism , Ki-67 Antigen/metabolism , Cell Cycle/physiology , Cell Nucleolus/metabolism , Chromatin/metabolism , Chromatin Assembly Factor-1/genetics , Chromosome Structures/metabolism , Chromosomes/metabolism , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Interphase/physiology , Ki-67 Antigen/genetics , Mitosis/physiology , Transcription Factors
16.
Chromosoma ; 125(3): 361-71, 2016 06.
Article in English | MEDLINE | ID: mdl-26174338

ABSTRACT

The regions of the genome that interact frequently with the nucleolus have been termed nucleolar-associated domains (NADs). Deep sequencing and DNA-fluorescence in situ hybridization (FISH) experiments have revealed that these domains are enriched for repetitive elements, regions of the inactive X chromosome (Xi), and several RNA polymerase III-transcribed genes. NADs are often marked by chromatin modifications characteristic of heterochromatin, including H3K27me3, H3K9me3, and H4K20me3, and artificial targeting of genes to this area is correlated with reduced expression. It has therefore been hypothesized that NAD localization to the nucleolar periphery contributes to the establishment and/or maintenance of heterochromatic silencing. Recently published studies from several multicellular eukaryotes have begun to reveal the trans-acting factors involved in NAD localization, including the insulator protein CCCTC-binding factor (CTCF), chromatin assembly factor (CAF)-1 subunit p150, several nucleolar proteins, and two long non-coding RNAs (lncRNAs). The mechanisms by which these factors coordinate with one another in regulating NAD localization and/or silencing are still unknown. This review will summarize recently published studies, discuss where additional research is required, and speculate about the mechanistic and functional implications of genome organization around the nucleolus.


Subject(s)
Cell Nucleolus/metabolism , Chromatin Assembly and Disassembly/physiology , Chromosomes, Human, X/metabolism , Genome, Human/physiology , Heterochromatin/metabolism , X Chromosome Inactivation/physiology , Animals , Cell Nucleolus/genetics , Chromosomes, Human, X/genetics , Heterochromatin/genetics , Humans
18.
Mol Biol Cell ; 25(18): 2866-81, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25057015

ABSTRACT

Chromatin assembly factor-1 (CAF-1) is a three-subunit protein complex conserved throughout eukaryotes that deposits histones during DNA synthesis. Here we present a novel role for the human p150 subunit in regulating nucleolar macromolecular interactions. Acute depletion of p150 causes redistribution of multiple nucleolar proteins and reduces nucleolar association with several repetitive element-containing loci. Of note, a point mutation in a SUMO-interacting motif (SIM) within p150 abolishes nucleolar associations, whereas PCNA or HP1 interaction sites within p150 are not required for these interactions. In addition, acute depletion of SUMO-2 or the SUMO E2 ligase Ubc9 reduces α-satellite DNA association with nucleoli. The nucleolar functions of p150 are separable from its interactions with the other subunits of the CAF-1 complex because an N-terminal fragment of p150 (p150N) that cannot interact with other CAF-1 subunits is sufficient for maintaining nucleolar chromosome and protein associations. Therefore these data define novel functions for a separable domain of the p150 protein, regulating protein and DNA interactions at the nucleolus.


Subject(s)
Cell Nucleolus/metabolism , Chromatin Assembly Factor-1/physiology , Chromosomes, Human/metabolism , HeLa Cells , Humans , Ki-67 Antigen/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Transcription Factors
19.
Proc Natl Acad Sci U S A ; 110(33): 13594-9, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23904484

ABSTRACT

Infection by pathogenic fungi, such as Candida albicans, begins with adhesion to host cells or implanted medical devices followed by biofilm formation. By high-throughput phenotypic screening of small molecules, we identified compounds that inhibit adhesion of C. albicans to polystyrene. Our lead candidate compound also inhibits binding of C. albicans to cultured human epithelial cells, the yeast-to-hyphal morphological transition, induction of the hyphal-specific HWP1 promoter, biofilm formation on silicone elastomers, and pathogenesis in a nematode infection model as well as alters fungal morphology in a mouse mucosal infection assay. We term this compound filastatin based on its strong inhibition of filamentation, and we use chemical genetic experiments to show that it acts downstream of multiple signaling pathways. These studies show that high-throughput functional assays targeting fungal adhesion can provide chemical probes for study of multiple aspects of fungal pathogenesis.


Subject(s)
Candida albicans/drug effects , Cell Adhesion/drug effects , High-Throughput Screening Assays/methods , Hyphae/drug effects , Morphogenesis/drug effects , Piperazines/pharmacology , Small Molecule Libraries/analysis , Animals , Candida albicans/physiology , Cells, Cultured , Epithelial Cells/metabolism , Humans , Hyphae/growth & development , Mice , Nematoda , Piperazines/chemistry , Polystyrenes/chemistry
20.
Bioorg Med Chem Lett ; 23(10): 2853-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23587423

ABSTRACT

The histone acetyltransferase Rtt109 is the sole enzyme responsible for acetylation of histone H3 lysine 56 (H3K56) in fungal organisms. Loss of Rtt109 renders fungal cells extremely sensitive to genotoxic agents, and prevents pathogenesis in several clinically important species. Here, via a high throughput chemical screen of >300,000 compounds, we discovered a chemical inhibitor of Rtt109 that does not inhibit other acetyltransferase enzymes. This compound inhibits Rtt109 regardless of which histone chaperone cofactor protein (Asf1 or Vps75) is present, and appears to inhibit Rtt109 via a tight-binding, uncompetitive mechanism.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces/enzymology , Small Molecule Libraries/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Acetyltransferases/metabolism , Molecular Structure , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...