Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Proc Natl Acad Sci U S A ; 116(13): 6111-6119, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850548

ABSTRACT

Microrchidia 3 (MORC3) is a human protein linked to autoimmune disorders, Down syndrome, and cancer. It is a member of a newly identified family of human ATPases with an uncharacterized mechanism of action. Here, we elucidate the molecular basis for inhibition and activation of MORC3. The crystal structure of the MORC3 region encompassing the ATPase and CW domains in complex with a nonhydrolyzable ATP analog demonstrates that the two domains are directly coupled. The extensive ATPase:CW interface stabilizes the protein fold but inhibits the catalytic activity of MORC3. Enzymatic, NMR, mutational, and biochemical analyses show that in the autoinhibited, off state, the CW domain sterically impedes binding of the ATPase domain to DNA, which in turn is required for the catalytic activity. MORC3 autoinhibition is released by disrupting the intramolecular ATPase:CW coupling through the competitive interaction of CW with histone H3 tail or by mutating the interfacial residues. Binding of CW to H3 leads to a marked rearrangement in the ATPase-CW cassette, which frees the DNA-binding site in active MORC3 (on state). We show that ATP-induced dimerization of the ATPase domain is strictly required for the catalytic activity and that the dimeric form of ATPase-CW might cooperatively bind to dsDNA. Together, our findings uncovered a mechanism underlying the fine-tuned regulation of the catalytic domain of MORC3 by the epigenetic reader, CW.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Catalysis , Catalytic Domain , Crystallography, X-Ray , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Enzyme Activation , Fluorescence Polarization , Histones/metabolism , Humans , Magnetic Resonance Spectroscopy
2.
J Mol Cell Cardiol ; 153: 44-59, 2021 04.
Article in English | MEDLINE | ID: mdl-33359755

ABSTRACT

Direct reprogramming of fibroblasts into cardiomyocytes (CMs) represents a promising strategy to regenerate CMs lost after ischemic heart injury. Overexpression of GATA4, HAND2, MEF2C, TBX5, miR-1, and miR-133 (GHMT2m) along with transforming growth factor beta (TGF-ß) inhibition efficiently promote reprogramming. However, the mechanisms by which TGF-ß blockade promotes cardiac reprogramming remain unknown. Here, we identify interactions between the histone H3 lysine 27 trimethylation (H3K27me3) demethylase JMJD3, the SWI/SNF remodeling complex subunit BRG1, and cardiac transcription factors. Furthermore, canonical TGF-ß signaling regulates the interaction between GATA4 and JMJD3. TGF-ß activation impairs the ability of GATA4 to bind target genes and prevents demethylation of H3K27 at cardiac gene promoters during cardiac reprogramming. Finally, a mutation in GATA4 (V267M) that is associated with congenital heart disease exhibits reduced binding to JMJD3 and impairs cardiomyogenesis. Thus, we have identified an epigenetic mechanism wherein canonical TGF-ß pathway activation impairs cardiac gene programming, in part by interfering with GATA4-JMJD3 interactions.


Subject(s)
GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Myocytes, Cardiac/cytology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , DNA Methylation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , GATA4 Transcription Factor/genetics , Histones/chemistry , Humans , Induced Pluripotent Stem Cells/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism
3.
Genes Dev ; 27(18): 2009-24, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-24065767

ABSTRACT

Histone acetyltransferases (HATs) assemble into multisubunit complexes in order to target distinct lysine residues on nucleosomal histones. Here, we characterize native HAT complexes assembled by the BRPF family of scaffold proteins. Their plant homeodomain (PHD)-Zn knuckle-PHD domain is essential for binding chromatin and is restricted to unmethylated H3K4, a specificity that is reversed by the associated ING subunit. Native BRPF1 complexes can contain either MOZ/MORF or HBO1 as catalytic acetyltransferase subunit. Interestingly, while the previously reported HBO1 complexes containing JADE scaffold proteins target histone H4, the HBO1-BRPF1 complex acetylates only H3 in chromatin. We mapped a small region to the N terminus of scaffold proteins responsible for histone tail selection on chromatin. Thus, alternate choice of subunits associated with HBO1 can switch its specificity between H4 and H3 tails. These results uncover a crucial new role for associated proteins within HAT complexes, previously thought to be intrinsic to the catalytic subunit.


Subject(s)
Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Chromatin/metabolism , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Homeodomain Proteins/metabolism , Humans , Methylation , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
4.
Biochemistry ; 59(4): 389-399, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31746185

ABSTRACT

Transcription factor 19 (TCF19) plays critical roles in type 1 diabetes and the maintenance of pancreatic ß cells. Recent studies have also implicated TCF19 in cell proliferation of hepatic carcinoma and non-small cell lung carcinoma; however, the mechanism underlying this regulation remains elusive. At the molecular level, TCF19 contains two modules, the plant homeodomain (PHD) finger and the forkhead-associated (FHA) domain, of unclear function. Here, we show that TCF19 mediates hepatocellular carcinoma HepG2 cell proliferation through its PHD finger that recognizes trimethylated lysine 4 of histone 3 (H3K4me3). W316 of the PHD finger of TCF19 is one of the critical residues eliciting this function. Whole genome microarray analysis and orthogonal cell-based assays identified a large subset of genes involved in cell survival and proliferation that depend on TCF19. Our data suggest that TCF19 acts as a pro-oncogene in hepatocellular carcinoma cells and that its functional PHD finger is critical in cell proliferation.


Subject(s)
Histones/metabolism , Transcription Factors/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Hep G2 Cells , Histone Code , Histones/genetics , Humans , Liver Neoplasms/metabolism , Lysine/metabolism , Methylation , Models, Molecular , PHD Zinc Fingers/physiology , Protein Binding , Transcription Factors/physiology
5.
Nucleic Acids Res ; 46(1): 421-430, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29145630

ABSTRACT

Yaf9 is an integral part of the NuA4 acetyltransferase and the SWR1 chromatin remodeling complexes. Here, we show that Yaf9 associates with acetylated histone H3 with high preference for H3K27ac. The crystal structure of the Yaf9 YEATS domain bound to the H3K27ac peptide reveals that the sequence C-terminal to K27ac stabilizes the complex. The side chain of K27ac inserts between two aromatic residues, mutation of which abrogates the interaction in vitro and leads in vivo to phenotypes similar to YAF9 deletion, including loss of SWR1-dependent incorporation of variant histone H2A.Z. Our findings reveal the molecular basis for the recognition of H3K27ac by a YEATS reader and underscore the importance of this interaction in mediating Yaf9 function within the NuA4 and SWR1 complexes.


Subject(s)
Adenosine Triphosphatases/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetylation , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Binding Sites/genetics , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Histones/chemistry , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid
6.
Nucleic Acids Res ; 44(13): 6102-12, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27016734

ABSTRACT

Histone post-translational modifications, and specific combinations they create, mediate a wide range of nuclear events. However, the mechanistic bases for recognition of these combinations have not been elucidated. Here, we characterize crosstalk between H3T3 and H3T6 phosphorylation, occurring in mitosis, and H3K4me3, a mark associated with active transcription. We detail the molecular mechanisms by which H3T3ph/K4me3/T6ph switches mediate activities of H3K4me3-binding proteins, including those containing plant homeodomain (PHD) and double Tudor reader domains. Our results derived from nuclear magnetic resonance chemical shift perturbation analysis, orthogonal binding assays and cell fluorescence microscopy studies reveal a strong anti-correlation between histone H3T3/T6 phosphorylation and retention of PHD finger proteins in chromatin during mitosis. Together, our findings uncover the mechanistic rules of chromatin engagement for H3K4me3-specific readers during cell division.


Subject(s)
Chromatin/genetics , Heterochromatin/genetics , Mitosis/genetics , Protein Processing, Post-Translational/genetics , Histone Code/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Methylation , Phosphorylation , Protein Binding/genetics , Tudor Domain/genetics
7.
Nucleic Acids Res ; 44(1): 472-84, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26626149

ABSTRACT

BRPF1 (bromodomain PHD finger 1) is a core subunit of the MOZ histone acetyltransferase (HAT) complex, critical for normal developmental programs and implicated in acute leukemias. BRPF1 contains a unique assembly of zinc fingers, termed a PZP domain, the physiological role of which remains unclear. Here, we elucidate the structure-function relationship of this novel epigenetic reader and detail the biological and mechanistic consequences of its interaction with nucleosomes. PZP has a globular architecture and forms a 2:1 stoichiometry complex with the nucleosome, bivalently interacting with histone H3 and DNA. This binding impacts the nucleosome dynamics, shifting the DNA unwrapping/rewrapping equilibrium toward the unwrapped state and increasing DNA accessibility. We demonstrate that the DNA-binding function of the BRPF1 PZP domain is required for the MOZ-BRPF1-ING5-hEaf6 HAT complex to be recruited to chromatin and to acetylate nucleosomal histones. Our findings reveal a novel link between chromatin dynamics and MOZ-mediated acetylation.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Chromatin/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Protein Interaction Domains and Motifs , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Chromatin/genetics , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Nuclear Proteins/genetics , Nucleosomes/genetics , Protein Binding , Protein Conformation , Protein Stability , Sequence Alignment
8.
Nature ; 451(7180): 851-4, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18235446

ABSTRACT

The transcription apparatus in Archaea can be described as a simplified version of its eukaryotic RNA polymerase (RNAP) II counterpart, comprising an RNAPII-like enzyme as well as two general transcription factors, the TATA-binding protein (TBP) and the eukaryotic TFIIB orthologue TFB. It has been widely understood that precise comparisons of cellular RNAP crystal structures could reveal structural elements common to all enzymes and that these insights would be useful in analysing components of each enzyme that enable it to perform domain-specific gene expression. However, the structure of archaeal RNAP has been limited to individual subunits. Here we report the first crystal structure of the archaeal RNAP from Sulfolobus solfataricus at 3.4 A resolution, completing the suite of multi-subunit RNAP structures from all three domains of life. We also report the high-resolution (at 1.76 A) crystal structure of the D/L subcomplex of archaeal RNAP and provide the first experimental evidence of any RNAP possessing an iron-sulphur (Fe-S) cluster, which may play a structural role in a key subunit of RNAP assembly. The striking structural similarity between archaeal RNAP and eukaryotic RNAPII highlights the simpler archaeal RNAP as an ideal model system for dissecting the molecular basis of eukaryotic transcription.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Sulfolobus solfataricus/enzymology , Crystallography, X-Ray , DNA-Directed RNA Polymerases/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Models, Molecular , Protein Folding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/enzymology , Taq Polymerase/chemistry
9.
Proc Natl Acad Sci U S A ; 108(2): 546-50, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187417

ABSTRACT

Spt4/5 in archaea and eukaryote and its bacterial homolog NusG is the only elongation factor conserved in all three domains of life and plays many key roles in cotranscriptional regulation and in recruiting other factors to the elongating RNA polymerase. Here, we present the crystal structure of Spt4/5 as well as the structure of RNA polymerase-Spt4/5 complex using cryoelectron microscopy reconstruction and single particle analysis. The Spt4/5 binds in the middle of RNA polymerase claw and encloses the DNA, reminiscent of the DNA polymerase clamp and ring helicases. The transcription elongation complex model reveals that the Spt4/5 is an upstream DNA holder and contacts the nontemplate DNA in the transcription bubble. These structures reveal that the cellular RNA polymerases also use a strategy of encircling DNA to enhance its processivity as commonly observed for many nucleic acid processing enzymes including DNA polymerases and helicases.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , DNA-Directed RNA Polymerases/metabolism , DNA/genetics , Nuclear Proteins/physiology , Pyrococcus furiosus/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcriptional Elongation Factors/physiology , Amino Acid Sequence , Animals , Cryoelectron Microscopy/methods , Crystallography, X-Ray/methods , DNA-Directed DNA Polymerase/genetics , Humans , Molecular Sequence Data , Nucleic Acids/chemistry , Sequence Homology, Amino Acid , Transcription, Genetic
10.
Nat Struct Mol Biol ; 31(7): 1039-1049, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38448574

ABSTRACT

JADE is a core subunit of the HBO1 acetyltransferase complex that regulates developmental and epigenetic programs and promotes gene transcription. Here we describe the mechanism by which JADE facilitates recruitment of the HBO1 complex to chromatin and mediates its enzymatic activity. Structural, genomic and complex assembly in vivo studies show that the PZP (PHD1-zinc-knuckle-PHD2) domain of JADE engages the nucleosome through binding to histone H3 and DNA and is necessary for the association with chromatin targets. Recognition of unmethylated H3K4 by PZP directs enzymatic activity of the complex toward histone H4 acetylation, whereas H3K4 hypermethylation alters histone substrate selectivity. We demonstrate that PZP contributes to leukemogenesis, augmenting transforming activity of the NUP98-JADE2 fusion. Our findings highlight biological consequences and the impact of the intact JADE subunit on genomic recruitment, enzymatic function and pathological activity of the HBO1 complex.


Subject(s)
Histone Acetyltransferases , Histones , Humans , Histones/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Animals , Chromatin/metabolism , Acetylation , Mice , Nucleosomes/metabolism , Protein Binding , Methylation , Models, Molecular , Protein Domains , Homeodomain Proteins , Tumor Suppressor Proteins
11.
Nat Commun ; 15(1): 5335, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914563

ABSTRACT

The NuA3 complex is a major regulator of gene transcription and the cell cycle in yeast. Five core subunits are required for complex assembly and function, but it remains unclear how these subunits interact to form the complex. Here, we report that the Taf14 subunit of the NuA3 complex binds to two other subunits of the complex, Yng1 and Sas3, and describe the molecular mechanism by which the extra-terminal domain of Taf14 recognizes the conserved motif present in Yng1 and Sas3. Structural, biochemical, and mutational analyses show that two motifs are sandwiched between the two extra-terminal domains of Taf14. The head-to-toe dimeric complex enhances the DNA binding activity of Taf14, and the formation of the hetero-dimer involving the motifs of Yng1 and Sas3 is driven by sequence complementarity. In vivo assays in yeast demonstrate that the interactions of Taf14 with both Sas3 and Yng1 are required for proper function of the NuA3 complex in gene transcription and DNA repair. Our findings suggest a potential basis for the assembly of three core subunits of the NuA3 complex, Taf14, Yng1 and Sas3.


Subject(s)
Protein Binding , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/chemistry , Protein Subunits/metabolism , Protein Subunits/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/chemistry , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Protein Multimerization , Models, Molecular , Transcription, Genetic , Amino Acid Sequence
12.
Biochim Biophys Acta Gene Regul Mech ; 1866(3): 194961, 2023 09.
Article in English | MEDLINE | ID: mdl-37482120

ABSTRACT

Taf14 is a subunit of multiple fundamental complexes implicated in transcriptional regulation and DNA damage repair in yeast cells. Here, we investigate the association of Taf14 with the consensus sequence present in other subunits of these complexes and describe the mechanistic features that affect this association. We demonstrate that the precise molecular mechanisms and biological outcomes underlying the Taf14 interactions depend on the accessibility of binding interfaces, the ability to recognize other ligands, and a degree of sensitivity to temperature and chemical and osmotic stresses. Our findings aid in a better understanding of how the distribution of Taf14 among the complexes is mediated.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIID/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Gene Expression Regulation
13.
Nat Struct Mol Biol ; 30(9): 1265-1274, 2023 09.
Article in English | MEDLINE | ID: mdl-37524969

ABSTRACT

The inhibitor of apoptosis protein BIRC2 regulates fundamental cell death and survival signaling pathways. Here we show that BIRC2 accumulates in the nucleus via binding of its second and third BIR domains, BIRC2BIR2 and BIRC2BIR3, to the histone H3 tail and report the structure of the BIRC2BIR3-H3 complex. RNA-seq analysis reveals that the genes involved in interferon and defense response signaling and cell-cycle regulation are most affected by depletion of BIRC2. Overexpression of BIRC2 delays DNA damage repair and recovery of the cell-cycle progression. We describe the structural mechanism for targeting of BIRC2BIR3 by a potent but biochemically uncharacterized small molecule inhibitor LCL161 and demonstrate that LCL161 disrupts the association of endogenous BIRC2 with H3 and stimulates cell death in cancer cells. We further show that LCL161 mediates degradation of BIRC2 in human immunodeficiency virus type 1-infected human CD4+ T cells. Our findings provide mechanistic insights into the nuclear accumulation of and blocking BIRC2.


Subject(s)
Inhibitor of Apoptosis Proteins , Thiazoles , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Apoptosis/genetics , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Nat Commun ; 14(1): 697, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754959

ABSTRACT

Human acetyltransferases MOZ and MORF are implicated in chromosomal translocations associated with aggressive leukemias. Oncogenic translocations involve the far amino terminus of MOZ/MORF, the function of which remains unclear. Here, we identified and characterized two structured winged helix (WH) domains, WH1 and WH2, in MORF and MOZ. WHs bind DNA in a cooperative manner, with WH1 specifically recognizing unmethylated CpG sequences. Structural and genomic analyses show that the DNA binding function of WHs targets MORF/MOZ to gene promoters, stimulating transcription and H3K23 acetylation, and WH1 recruits oncogenic fusions to HOXA genes that trigger leukemogenesis. Cryo-EM, NMR, mass spectrometry and mutagenesis studies provide mechanistic insight into the DNA-binding mechanism, which includes the association of WH1 with the CpG-containing linker DNA and binding of WH2 to the dyad of the nucleosome. The discovery of WHs in MORF and MOZ and their DNA binding functions could open an avenue in developing therapeutics to treat diseases associated with aberrant MOZ/MORF acetyltransferase activities.


Subject(s)
Acetyltransferases , Histone Acetyltransferases , Leukemia , Humans , Acetylation , Acetyltransferases/metabolism , CpG Islands/genetics , Histone Acetyltransferases/metabolism , Leukemia/genetics , Translocation, Genetic
15.
Nat Commun ; 13(1): 3177, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35676274

ABSTRACT

The assembly and function of the yeast general transcription factor TFIID complex requires specific contacts between its Taf14 and Taf2 subunits, however, the mechanism underlying these contacts remains unclear. Here, we determined the molecular and structural basis by which the YEATS and ET domains of Taf14 bind to the C-terminal tail of Taf2 and identified a unique DNA-binding activity of the linker region connecting the two domains. We show that in the absence of ligands the linker region of Taf14 is occluded by the surrounding domains, and therefore the DNA binding function of Taf14 is autoinhibited. Binding of Taf2 promotes a conformational rearrangement in Taf14, resulting in a release of the linker for the engagement with DNA and the nucleosome. Genetic in vivo data indicate that the association of Taf14 with both Taf2 and DNA is essential for transcriptional regulation. Our findings provide a basis for deciphering the role of individual TFIID subunits in mediating gene transcription.


Subject(s)
Saccharomyces cerevisiae Proteins , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , DNA/metabolism , Gene Expression Regulation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism
16.
Curr Opin Struct Biol ; 71: 1-6, 2021 12.
Article in English | MEDLINE | ID: mdl-33993059

ABSTRACT

The past two decades have witnessed rapid advances in the identification and characterization of epigenetic readers, capable of recognizing or reading post-translational modifications in histones. More recently, a new set of readers with the ability to interact with the nucleosome through concomitant binding to histones and DNA has emerged. In this review, we discuss mechanistic insights underlying bivalent histone and DNA recognition by newly characterized readers and highlight the importance of binding to DNA for their association with chromatin.


Subject(s)
Histones , Nucleosomes , Chromatin , DNA/genetics , Epigenesis, Genetic , Histones/metabolism , Protein Processing, Post-Translational
17.
STAR Protoc ; 2(2): 100479, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33982013

ABSTRACT

The core subunit of the MORF acetyltransferase complex BRPF1 contains a unique combination of zinc fingers, including a plant homeodomain (PHD) finger followed by a zinc knuckle and another PHD finger, which together form a PZP domain (BRPF1PZP). BRPF1PZP has been shown to bind to the nucleosome and make contacts with both histone H3 tail and DNA. Here, we describe biophysical and structural methods for characterization of the interactions between BRPF1PZP, H3 tail, DNA, and the intact nucleosome. For complete details on the use and execution of this protocol, please refer to Klein et al. (2020).


Subject(s)
Adaptor Proteins, Signal Transducing , DNA-Binding Proteins , Nucleosomes , Protein Domains/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Biophysical Phenomena , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Fluorescence Polarization , Histones/chemistry , Histones/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Nucleosomes/chemistry , Nucleosomes/metabolism
18.
J Med Chem ; 64(15): 10997-11013, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34279931

ABSTRACT

Eleven-nineteen leukemia (ENL) protein is a histone acetylation reader essential for disease maintenance in acute leukemias, in particular, the mixed-lineage leukemia (MLL)-rearranged leukemia. In this study, we carried out high-throughput screening of a small-molecule library to identify inhibitors for the ENL YEATS domain. Structure-activity relationship studies of the hits and structure-based inhibitor design led to two compounds, 11 and 24, with IC50 values below 100 nM in inhibiting the ENL-acetyl-H3 interaction. Both compounds, and their precursor compound 7, displayed strong selectivity toward the ENL YEATS domain over all other human YEATS domains. Moreover, 7 exhibited on-target inhibition of ENL in cultured cells and a synergistic effect with the bromodomain and extraterminal domain inhibitor JQ1 in killing leukemia cells. Together, we have developed selective chemical probes for the ENL YEATS domain, providing the basis for further medicinal chemistry-based optimization to advance both basic and translational research of ENL.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Leukemia, Myeloid, Acute/drug therapy , Small Molecule Libraries/pharmacology , Transcriptional Elongation Factors/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Structure , Protein Domains/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Transcriptional Elongation Factors/metabolism
19.
Nat Commun ; 12(1): 4130, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34226546

ABSTRACT

Chromosomal translocations of the AF10 (or MLLT10) gene are frequently found in acute leukemias. Here, we show that the PZP domain of AF10 (AF10PZP), which is consistently impaired or deleted in leukemogenic AF10 translocations, plays a critical role in blocking malignant transformation. Incorporation of functional AF10PZP into the leukemogenic CALM-AF10 fusion prevents the transforming activity of the fusion in bone marrow-derived hematopoietic stem and progenitor cells in vitro and in vivo and abrogates CALM-AF10-mediated leukemogenesis in vivo. Crystallographic, biochemical and mutagenesis studies reveal that AF10PZP binds to the nucleosome core particle through multivalent contacts with the histone H3 tail and DNA and associates with chromatin in cells, colocalizing with active methylation marks and discriminating against the repressive H3K27me3 mark. AF10PZP promotes nuclear localization of CALM-AF10 and is required for association with chromatin. Our data indicate that the disruption of AF10PZP function in the CALM-AF10 fusion directly leads to transformation, whereas the inclusion of AF10PZP downregulates Hoxa genes and reverses cellular transformation. Our findings highlight the molecular mechanism by which AF10 targets chromatin and suggest a model for the AF10PZP-dependent CALM-AF10-mediated leukemogenesis.


Subject(s)
Acute Disease , Leukemia, Myeloid, Acute/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Translocation, Genetic/genetics , Animals , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Chromatin , HEK293 Cells , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Methylation , Mice , Models, Molecular , Monomeric Clathrin Assembly Proteins/genetics , Monomeric Clathrin Assembly Proteins/metabolism , Nucleosomes , Protein Conformation
20.
Structure ; 28(1): 105-110.e3, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31711755

ABSTRACT

The assembly of human histone acetyltransferase MOZ/MORF complexes relies on the scaffolding bromodomain plant homeodomain (PHD) finger 1 (BRPF1) subunit. The PHD-zinc-knuckle-PHD module of BRPF1 (BRPF1PZP) has been shown to associate with the histone H3 tail and DNA; however, the molecular mechanism underlying recognition of H3 and the relationship between the histone and DNA-binding activities remain unclear. In this study, we report the crystal structure of BRPF1PZP bound to the H3 tail and characterize the role of the bipartite interaction in the engagement of BRPF1PZP with the nucleosome core particle (NCP). We find that although both interactions of BRPF1PZP with the H3 tail and DNA are required for tight binding to NCP and for acetyltransferase function of the BRPF1-MORF-ING5-MEAF6 complex, binding to extranucleosomal DNA dominates. Our findings suggest that functionally active BRPF1PZP might be important in stabilization of the MOZ/MORF complexes at chromatin with accessible DNA.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , DNA/metabolism , Histones/metabolism , Humans , Protein Binding , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL