Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 5083, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572554

ABSTRACT

Protein-protein interactions (PPIs) are at the core of virtually all biological processes in cells. Consequently, targeting PPIs is emerging at the forefront of drug discovery. Cellular assays which closely recapitulate native conditions in vivo are instrumental to understand how small molecule drugs can modulate such interactions. We have integrated MultiBacMam, a baculovirus-based mammalian gene delivery tool we developed, with bimolecular fluorescence complementation (BiFC), giving rise to a highly efficient system for assay development, identification and characterization of PPI modulators. We used our system to analyze compounds impacting on CDK5-p25 PPI, which is implicated in numerous diseases including Alzheimer's. We evaluated our tool-kit with the known inhibitor p5T, and we established a mini-screen to identify compounds that modulate this PPI in dose-response experiments. Finally, we discovered several compounds disrupting CDK5-p25 PPI, which had not been identified by other screening or structure-based methods before.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Drug Evaluation, Preclinical/methods , Nerve Tissue Proteins/metabolism , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Drug Discovery/methods , Fluorescence , Humans , Molecular Docking Simulation , Nerve Tissue Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry
2.
Annu Rev Genet ; 47: 275-306, 2013.
Article in English | MEDLINE | ID: mdl-24016189

ABSTRACT

Saccharomyces cerevisiae provides a well-studied model system for heritable silent chromatin in which a histone-binding protein complex [the SIR (silent information regulator) complex] represses gene transcription in a sequence-independent manner by spreading along nucleosomes, much like heterochromatin in higher eukaryotes. Recent advances in the biochemistry and structural biology of the SIR-chromatin system bring us much closer to a molecular understanding of yeast silent chromatin. Simultaneously, genome-wide approaches have shed light on the biological importance of this form of epigenetic repression. Here, we integrate genetic, structural, and cell biological data into an updated overview of yeast silent chromatin assembly.


Subject(s)
Chromatin/metabolism , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/physiology , Acetylation , Chromatin/genetics , DNA, Fungal/genetics , Gene Silencing , Genes, Fungal , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , Homeodomain Proteins/physiology , Models, Genetic , Nucleosomes/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Repressor Proteins/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Telomere/physiology , Transcription Factors/physiology , Transcription, Genetic
3.
Gene ; 527(1): 10-25, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23791651

ABSTRACT

Discrete regions of the eukaryotic genome assume a heritable chromatin structure that is refractory to gene expression, referred to as heterochromatin or "silent" chromatin. Constitutively silent chromatin is found in subtelomeric domains in a number of species, ranging from yeast to man. In addition, chromatin-dependent repression of mating type loci occurs in both budding and fission yeasts, to enable sexual reproduction. The silencing of chromatin in budding yeast is characterized by an assembly of Silent Information Regulatory (SIR) proteins-Sir2, Sir3 and Sir4-with unmodified nucleosomes. Silencing requires the lysine deacetylase activity of Sir2, extensive contacts between Sir3 and the nucleosome, as well as interactions among the SIR proteins, to generate the Sir2-3-4 or SIR complex. Results from recent structural and reconstitution studies suggest an updated model for the ordered assembly and organization of SIR-dependent silent chromatin in yeast. Moreover, studies of subtelomeric gene expression reveal the importance of subtelomeric silent chromatin in the regulation of genes other than the silent mating type loci. This review covers recent advances in this field.


Subject(s)
Fungal Proteins/metabolism , Nucleosomes/enzymology , Sirtuins/metabolism , Yeasts/enzymology , Animals , Chromatin/enzymology , Chromatin/metabolism , Gene Expression Regulation, Fungal , Gene Silencing , Histones/metabolism , Humans , Nucleosomes/metabolism , Protein Processing, Post-Translational , Yeasts/genetics
4.
EMBO J ; 32(3): 437-49, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23299941

ABSTRACT

Gene silencing in budding yeast relies on the binding of the Silent Information Regulator (Sir) complex to chromatin, which is mediated by extensive interactions between the Sir proteins and nucleosomes. Sir3, a divergent member of the AAA+ ATPase-like family, contacts both the histone H4 tail and the nucleosome core. Here, we present the structure and function of the conserved C-terminal domain of Sir3, comprising 138 amino acids. This module adopts a variant winged helix-turn-helix (wH) architecture that exists as a stable homodimer in solution. Mutagenesis shows that the self-association mediated by this domain is essential for holo-Sir3 dimerization. Its loss impairs Sir3 loading onto nucleosomes in vitro and eliminates silencing at telomeres and HM loci in vivo. Replacing the Sir3 wH domain with an unrelated bacterial dimerization motif restores both HM and telomeric repression in sir3Δ cells. In contrast, related wH domains of archaeal and human members of the Orc1/Sir3 family are monomeric and have DNA binding activity. We speculate that a dimerization function for the wH evolved with Sir3's ability to facilitate heterochromatin formation.


Subject(s)
Gene Silencing/physiology , Heterochromatin/physiology , Models, Molecular , Protein Conformation , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Chromatin/metabolism , Chromatin Immunoprecipitation , Crystallization , DNA Primers/genetics , Dimerization , Evolution, Molecular , Genetic Complementation Test , Heterochromatin/genetics , Immunoprecipitation , Molecular Sequence Data , Mutagenesis , Nucleosomes/metabolism , Polymerase Chain Reaction , Saccharomyces cerevisiae , Sequence Alignment , Silent Information Regulator Proteins, Saccharomyces cerevisiae/chemistry , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics
5.
PLoS Genet ; 8(5): e1002727, 2012.
Article in English | MEDLINE | ID: mdl-22654676

ABSTRACT

Silent information regulator proteins Sir2, Sir3, and Sir4 form a heterotrimeric complex that represses transcription at subtelomeric regions and homothallic mating type (HM) loci in budding yeast. We have performed a detailed biochemical and genetic analysis of the largest Sir protein, Sir4. The N-terminal half of Sir4 is dispensable for SIR-mediated repression of HM loci in vivo, except in strains that lack Yku70 or have weak silencer elements. For HM silencing in these cells, the C-terminal domain (Sir4C, residues 747-1,358) must be complemented with an N-terminal domain (Sir4N; residues 1-270), expressed either independently or as a fusion with Sir4C. Nonetheless, recombinant Sir4C can form a complex with Sir2 and Sir3 in vitro, is catalytically active, and has sedimentation properties similar to a full-length Sir4-containing SIR complex. Sir4C-containing SIR complexes bind nucleosomal arrays and protect linker DNA from nucleolytic digestion, but less effectively than wild-type SIR complexes. Consistently, full-length Sir4 is required for the complete repression of subtelomeric genes. Supporting the notion that the Sir4 N-terminus is a regulatory domain, we find it extensively phosphorylated on cyclin-dependent kinase consensus sites, some being hyperphosphorylated during mitosis. Mutation of two major phosphoacceptor sites (S63 and S84) derepresses natural subtelomeric genes when combined with a serendipitous mutation (P2A), which alone can enhance the stability of either the repressed or active state. The triple mutation confers resistance to rapamycin-induced stress and a loss of subtelomeric repression. We conclude that the Sir4 N-terminus plays two roles in SIR-mediated silencing: it contributes to epigenetic repression by stabilizing the SIR-mediated protection of linker DNA; and, as a target of phosphorylation, it can destabilize silencing in a regulated manner.


Subject(s)
Genes, Mating Type, Fungal , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Telomere/genetics , Transcription, Genetic , Chromatin/genetics , Cyclin-Dependent Kinases , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Fungal , Gene Silencing , Genes, Mating Type, Fungal/genetics , Mitosis , Phosphorylation , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Transcriptional Activation
6.
Genes Dev ; 25(17): 1835-46, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21896656

ABSTRACT

The silent information regulator 2/3/4 (Sir2/3/4) complex is required for gene silencing at the silent mating-type loci and at telomeres in Saccharomyces cerevisiae. Sir3 is closely related to the origin recognition complex 1 subunit and consists of an N-terminal bromo-adjacent homology (BAH) domain and a C-terminal AAA(+) ATPase-like domain. Here, through a combination of structure biology and exhaustive mutagenesis, we identified unusual, silencing-specific features of the AAA(+) domain of Sir3. Structural analysis of the putative nucleotide-binding pocket in this domain reveals a shallow groove that would preclude nucleotide binding. Mutation of this site has little effect on Sir3 function in vivo. In contrast, several surface regions are shown to be necessary for the Sir3 silencing function. Interestingly, the Sir3 AAA(+) domain is shown here to bind chromatin in vitro in a manner sensitive to histone H3K79 methylation. Moreover, an exposed loop on the surface of this Sir3 domain is found to interact with Sir4. In summary, the unique folding of this conserved Sir3 AAA(+) domain generates novel surface regions that mediate Sir3-Sir4 and Sir3-nucleosome interactions, both being required for the proper assembly of heterochromatin in living cells.


Subject(s)
Gene Silencing , Histones/metabolism , Models, Molecular , Saccharomyces cerevisiae , Silent Information Regulator Proteins, Saccharomyces cerevisiae/chemistry , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Alleles , Chromatin/metabolism , DNA Methylation , Histones/chemistry , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics
7.
EMBO J ; 30(13): 2610-21, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21666601

ABSTRACT

Discrete regions of the eukaryotic genome assume heritable chromatin structure that is refractory to transcription. In budding yeast, silent chromatin is characterized by the binding of the Silent Information Regulatory (Sir) proteins to unmodified nucleosomes. Using an in vitro reconstitution assay, which allows us to load Sir proteins onto arrays of regularly spaced nucleosomes, we have examined the impact of specific histone modifications on Sir protein binding and linker DNA accessibility. Two typical marks for active chromatin, H3K79(me) and H4K16(ac) decrease the affinity of Sir3 for chromatin, yet only H4K16(ac) affects chromatin structure, as measured by nuclease accessibility. Surprisingly, we found that the Sir2-4 subcomplex, unlike Sir3, has higher affinity for chromatin carrying H4K16(ac). NAD-dependent deacetylation of H4K16(ac) promotes binding of the SIR holocomplex but not of the Sir2-4 heterodimer. This function of H4K16(ac) cannot be substituted by H3K56(ac). We conclude that acetylated H4K16 has a dual role in silencing: it recruits Sir2-4 and repels Sir3. Moreover, the deacetylation of H4K16(ac) by Sir2 actively promotes the high-affinity binding of the SIR holocomplex.


Subject(s)
Chromatin/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/physiology , Histones/metabolism , Acetylation , Animals , Cells, Cultured , Chromatin Assembly and Disassembly/physiology , Histones/physiology , Lysine/metabolism , Models, Biological , Models, Molecular , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/physiology , Sirtuin 2/metabolism , Sirtuin 2/physiology , Spodoptera , Yeasts/genetics , Yeasts/metabolism
8.
Mol Cell ; 33(3): 323-34, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19217406

ABSTRACT

At yeast telomeres and silent mating-type loci, chromatin assumes a higher-order structure that represses transcription by means of the histone deacetylase Sir2 and structural proteins Sir3 and Sir4. Here, we present a fully reconstituted system to analyze SIR holocomplex binding to nucleosomal arrays. Purified Sir2-3-4 heterotrimers bind chromatin, cooperatively yielding a stable complex of homogeneous molecular weight. Remarkably, Sir2-3-4 also binds naked DNA, reflecting the strong, albeit nonspecific, DNA-binding activity of Sir4. The binding of Sir3 to nucleosomes is sensitive to histone H4 N-terminal tail removal, while that of Sir2-4 is not. Dot1-mediated methylation of histone H3K79 reduces the binding of both Sir3 and Sir2-3-4. Additionally, a byproduct of Sir2-mediated NAD hydrolysis, O-acetyl-ADP-ribose, increases the efficiency with which Sir3 and Sir2-3-4 bind nucleosomes. Thus, in small cumulative steps, each Sir protein, unmodified histone domains, and contacts with DNA contribute to the stability of the silent chromatin complex.


Subject(s)
Chromatin/metabolism , Nucleosomes/metabolism , O-Acetyl-ADP-Ribose/metabolism , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Binding Sites , Histone Deacetylases/isolation & purification , Histone Deacetylases/metabolism , Models, Biological , Models, Molecular , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/isolation & purification , Sirtuin 2 , Sirtuins/isolation & purification , Sirtuins/metabolism
9.
Chromosoma ; 117(2): 199-210, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18075750

ABSTRACT

Sister chromatids are physically connected by cohesin complexes. This sister chromatid cohesion is essential for the biorientation of chromosomes on the mitotic and meiotic spindle. In many species, cohesion between chromosome arms is partly dissolved in prophase of mitosis, whereas cohesion is protected at centromeres until the onset of anaphase. In vertebrates, the protein Sgo1, protein phosphatase 2A, and several other proteins are required for protection of centromeric cohesin in early mitosis. In fission yeast, the recruitment of heterochromatin protein Swi6/HP1 to centromeres by the histone-methyltransferase Clr4/Suv39h is required for enrichment of cohesin at centromeres already in interphase. We have tested if the Suv39h-HP1 histone methylation pathway is also required for enrichment and mitotic protection of cohesin at centromeres in mammalian cells. We show that cohesin and HP1 proteins partially colocalize at mitotic centromeres but that cohesin localization is not detectably altered in mouse embryonic fibroblasts that lack Suv39h genes and in which HP1 proteins can, therefore, not be properly enriched in pericentric heterochromatin. Our data indicate that the Suv39h-HP1 pathway is not essential for enrichment and mitotic protection of cohesin at centromeres in mammalian cells.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Methyltransferases/physiology , Repressor Proteins/physiology , Animals , Centromere/ultrastructure , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Fibroblasts/metabolism , Meiosis , Methylation , Methyltransferases/genetics , Mice , Microscopy, Fluorescence , Mitosis , Repressor Proteins/genetics , Schizosaccharomyces , Spindle Apparatus , Cohesins
10.
Cell ; 127(5): 955-67, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17113138

ABSTRACT

Cohesin establishes sister-chromatid cohesion from S phase until mitosis or meiosis. To allow chromosome segregation, cohesion has to be dissolved. In vertebrate cells, this process is mediated in part by the protease separase, which destroys a small amount of cohesin, but most cohesin is removed from chromosomes without proteolysis. How this is achieved is poorly understood. Here, we show that the interaction between cohesin and chromatin is controlled by Wapl, a protein implicated in heterochromatin formation and tumorigenesis. Wapl is associated with cohesin throughout the cell cycle, and its depletion blocks cohesin dissociation from chromosomes during the early stages of mitosis and prevents the resolution of sister chromatids until anaphase, which occurs after a delay. Wapl depletion also increases the residence time of cohesin on chromatin in interphase. Our data indicate that Wapl is required to unlock cohesin from a particular state in which it is stably bound to chromatin.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Chromatids/metabolism , Chromosomes, Human/genetics , Conserved Sequence , DNA-Binding Proteins , Evolution, Molecular , Humans , Interphase , Multiprotein Complexes/metabolism , Oncogene Proteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Prophase , Protein Binding , Protein Structure, Secondary , Proto-Oncogene Proteins , Repetitive Sequences, Amino Acid , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Telophase , Cohesins
11.
EMBO Rep ; 7(3): 314-20, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16439995

ABSTRACT

A crucial step in the RNA interference (RNAi) pathway involves the assembly of RISC, the RNA-induced silencing complex. RISC initially recognizes a double-stranded short interfering RNA (siRNA), but only one strand is finally retained in the functional ribonucleoprotein complex. The non-incorporated strand, or 'passenger' strand, is removed during the assembly process and most probably degraded thereafter. In this report, we show that the passenger strand is cleaved during the course of RISC assembly following the same rules established for the siRNA-guided cleavage of a target RNA. Chemical modifications impairing the cleavage of the passenger strand also impair the cleavage of a target RNA in vitro as well as the silencing of a reporter gene in vivo, suggesting that passenger strand removal is facilitated by its cleavage during RISC assembly. Interestingly, target RNA cleavage can be rescued if an otherwise non-cleavable passenger strand shows a nick at the scissile phosphodiester bond, which further indicates that the cleavage event per se is not essential.


Subject(s)
Gene Silencing , RNA Interference , RNA, Small Interfering/metabolism , RNA/metabolism , Genes, Reporter , HeLa Cells , Humans , Multiprotein Complexes , RNA/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...