Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
Int J Mol Sci ; 22(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638547

ABSTRACT

Cytochromes P450 (CYP) are one of the major xenobiotic metabolizing enzymes with increasing importance in pharmacogenetics. The CYP2C9 enzyme is responsible for the metabolism of a wide range of clinical drugs. More than sixty genetic variations have been identified in CYP2C9 with many demonstrating reduced activity compared to the wild-type (WT) enzyme. The CYP2C9*8 allele is predominantly found in persons of African ancestry and results in altered clearance of several drug substrates of CYP2C9. The X-ray crystal structure of CYP2C9*8, which represents an amino acid variation from arginine to histidine at position 150 (R150H), was solved in complex with losartan. The overall conformation of the CYP2C9*8-losartan complex was similar to the previously solved complex with wild type (WT) protein, but it differs in the occupancy of losartan. One molecule of losartan was bound in the active site and another on the surface in an identical orientation to that observed in the WT complex. However, unlike the WT structure, the losartan in the access channel was not observed in the *8 complex. Furthermore, isothermal titration calorimetry studies illustrated weaker binding of losartan to *8 compared to WT. Interestingly, the CYP2C9*8 interaction with losartan was not as weak as the CYP2C9*3 variant, which showed up to three-fold weaker average dissociation constant compared to the WT. Taken together, the structural and solution characterization yields insights into the similarities and differences of losartan binding to CYP2C9 variants and provides a useful framework for probing the role of amino acid substitution and substrate dependent activity.


Subject(s)
Catalytic Domain/genetics , Cytochrome P-450 CYP2C9/genetics , Inactivation, Metabolic/genetics , Losartan/metabolism , Alleles , Amino Acid Substitution/genetics , Binding Sites/genetics , Cytochrome P-450 CYP2C9/metabolism , Genetic Variation/genetics , Humans , Inactivation, Metabolic/physiology , Protein Conformation
3.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502039

ABSTRACT

The ATPase Family, AAA domain-containing protein 2 (ATAD2) bromodomain (BRD) has a canonical bromodomain structure consisting of four α-helices. ATAD2 functions as a co-activator of the androgen and estrogen receptors as well as the MYC and E2F transcription factors. ATAD2 also functions during DNA replication, recognizing newly synthesized histones. In addition, ATAD2 is shown to be up-regulated in multiple forms of cancer including breast, lung, gastric, endometrial, renal, and prostate. Furthermore, up-regulation of ATAD2 is strongly correlated with poor prognosis in many types of cancer, making the ATAD2 bromodomain an innovative target for cancer therapeutics. In this study, we describe the recognition of histone acetyllysine modifications by the ATAD2 bromodomain. Residue-specific information on the complex formed between the histone tail and the ATAD2 bromodomain, obtained through nuclear magnetic resonance spectroscopy (NMR) and X-ray crystallography, illustrates key residues lining the binding pocket, which are involved in coordination of di-acetylated histone tails. Analytical ultracentrifugation, NMR relaxation data, and isothermal titration calorimetry further confirm the monomeric state of the functionally active ATAD2 bromodomain in complex with di-acetylated histone ligands. Overall, we describe histone tail recognition by ATAD2 BRD and illustrate that one acetyllysine group is primarily engaged by the conserved asparagine (N1064), the "RVF" shelf residues, and the flexible ZA loop. Coordination of a second acetyllysine group also occurs within the same binding pocket but is essentially governed by unique hydrophobic and electrostatic interactions making the di-acetyllysine histone coordination more specific than previously presumed.


Subject(s)
ATPases Associated with Diverse Cellular Activities/chemistry , DNA-Binding Proteins/chemistry , Histones/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Acetylation , DNA-Binding Proteins/metabolism , Histone Code , Histones/chemistry , Humans , Protein Binding , Protein Domains
4.
Mol Pharmacol ; 98(5): 529-539, 2020 11.
Article in English | MEDLINE | ID: mdl-32938720

ABSTRACT

The human CYP2C9 plays a crucial role in the metabolic clearance of a wide range of clinical therapeutics. The *2 allele is a prevalent genetic variation in CYP2C9 that is found in various populations. A marked reduction of catalytic activity toward many important drug substrates has been demonstrated by CYP2C9*2, which represents an amino acid variation at position 144 from arginine to cysteine. The crystal structure of CYP2C9*2 in complex with an antihypertensive drug losartan was solved using X-ray crystallography at 3.1-Å resolution. The Arg144Cys variation in the *2 complex disrupts the hydrogen-bonding interactions that were observed between the side chain of arginine and neighboring residues in the losartan complex of CYP2C9 and the wild-type (WT) ligand-free structure. The conformation of several secondary structural elements is affected, thereby altering the binding and orientation of drug and important amino acid side chains in the distal active site cavity. The new structure revealed distinct interactions of losartan in the compact active site of CYP2C9*2 and differed in occupancy at the other binding sites previously identified in the WT-losartan complex. Furthermore, the binding studies in solution using losartan illustrated lower activity of the CYP2C9*2 compared with the WT. Together, the findings yield valuable insights into the decreased hydroxylation activity of losartan in patients carrying CYP2C9*2 allele and provide a useful framework to investigate the effect of a single-nucleotide polymorphism that leads to altered metabolism of diverse drug substrates. SIGNIFICANCE STATEMENT: The *2 allele of the human drug-metabolizing enzyme CYP2C9 is found in different populations and results in significantly reduced activity toward various drug substrates. How the CYP2C9*2 variant induces altered drug metabolism is poorly understood given that the Arg144Cys variation is located far away from the active site. This work yield insight into the effect of distal variation using multitude of techniques that include X-ray crystallography, isothermal titration calorimetry, enzymatic characterization, and computational studies.


Subject(s)
Cytochrome P-450 CYP2C9/genetics , Losartan/chemistry , Polymorphism, Single Nucleotide/genetics , Alleles , Antihypertensive Agents/chemistry , Catalytic Domain/genetics , Humans
5.
Proteins ; 87(2): 157-167, 2019 02.
Article in English | MEDLINE | ID: mdl-30520161

ABSTRACT

The ATPase family, AAA domain-containing protein 2 (ATAD2) has a C-terminal bromodomain, which functions as a chromatin reader domain recognizing acetylated lysine on the histone tails within the nucleosome. ATAD2 is overexpressed in many cancers and its expression is correlated with poor patient outcomes, making it an attractive therapeutic target and potential biomarker. We solved the crystal structure of the ATAD2 bromodomain and found that it contains a disulfide bridge near the base of the acetyllysine binding pocket (Cys1057-Cys1079). Site-directed mutagenesis revealed that removal of a free C-terminal cysteine (C1101) residue greatly improved the solubility of the ATAD2 bromodomain in vitro. Isothermal titration calorimetry experiments in combination with the Ellman's assay demonstrated that formation of an intramolecular disulfide bridge negatively impacts the ligand binding affinities and alters the thermodynamic parameters of the ATAD2 bromodomain interaction with a histone H4K5ac peptide as well as a small molecule bromodomain ligand. Molecular dynamics simulations indicate that the formation of the disulfide bridge in the ATAD2 bromodomain does not alter the structure of the folded state or flexibility of the acetyllysine binding pocket. However, consideration of this unique structural feature should be taken into account when examining ligand-binding affinity, or in the design of new bromodomain inhibitor compounds that interact with this acetyllysine reader module.


Subject(s)
ATPases Associated with Diverse Cellular Activities/chemistry , Adenosine Triphosphatases/chemistry , Cysteine/chemistry , DNA-Binding Proteins/chemistry , Disulfides/chemistry , Protein Domains , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Crystallography, X-Ray , Cysteine/genetics , Cysteine/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disulfides/metabolism , Histones/chemistry , Histones/metabolism , Humans , Ligands , Lysine/chemistry , Lysine/metabolism , Molecular Dynamics Simulation , Mutation , Protein Binding , Solubility , Thermodynamics
6.
J Cell Physiol ; 233(3): 1877-1886, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28500727

ABSTRACT

Bromodomain proteins function as epigenetic readers that recognize acetylated histone tails to facilitate the transcription of target genes. There are approximately 60 known human bromodomains, which are divided into eight sub-families based on structural conservation. The bromodomain-containing proteins in family IV include seven members (BRPF1, BRPF2, BRPF3, BRD7, BRD9, ATAD2, and ATAD2b). The bromodomains of each of these proteins recognize and bind acetyllysine residues on histone tails protruding from the nucleosome. However, the histone marks recognized by each bromodomain protein can be very different. The BRPF1 subunit of the MOZ histone acetyltransferase (HAT) recognizes acetylated histones H2AK5ac, H4K12ac, H3K14ac, H4K8ac, and H4K5ac. While the bromodomain of BRD7, a member of the SWI/SNF complex, was shown to preferentially recognize acetylated histones H3K9ac, H3K14ac, H4K8ac, H4K12ac, and H4K16ac. The bromodomains of BRPF2 and BRPF3 have similar sequences, and function as part of the HBO1 HAT complex, but there is limited data on which histone ligands they bind. Similarly, there is little known about the histone targets of the BRD9 and ATAD2b bromodomain proteins. Interestingly, the ATAD2 bromodomain was recently shown to preferentially bind to the di-acetylated H4K5acK12ac mark found in newly synthesized histones following DNA replication. However, despite the physiological importance of the family IV bromodomains, little is known about how they function at the molecular or atomic level. In this review, we summarize our understanding of how family IV bromodomains recognize and select for acetyllysine marks and discuss the importance of acetylated histone recognition for their biological functions.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetylation , Epigenomics , Histone Acetyltransferases , Histone Chaperones , Humans , Models, Molecular , Protein Binding
7.
J Biol Chem ; 291(35): 18326-41, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27281824

ABSTRACT

Aberrant access to genetic information disrupts cellular homeostasis and can lead to cancer development. One molecular mechanism that regulates access to genetic information includes recognition of histone modifications, which is carried out by protein modules that interact with chromatin and serve as landing pads for enzymatic activities that regulate gene expression. The ING3 tumor suppressor protein contains a plant homeodomain (PHD) that reads the epigenetic code via recognition of histone H3 tri-methylated at lysine 4 (H3K4me3), and this domain is lost or mutated in various human cancers. However, the molecular mechanisms targeting ING3 to histones and the role of this interaction in the cell remain elusive. Thus, we employed biochemical and structural biology approaches to investigate the interaction of the ING3 PHD finger (ING3PHD) with the active transcription mark H3K4me3. Our results demonstrate that association of the ING3PHD with H3K4me3 is in the sub-micromolar range (KD ranging between 0.63 and 0.93 µm) and is about 200-fold stronger than with the unmodified histone H3. NMR and computational studies revealed an aromatic cage composed of Tyr-362, Ser-369, and Trp-385 that accommodate the tri-methylated side chain of H3K4. Mutational analysis confirmed the critical importance of Tyr-362 and Trp-385 in mediating the ING3PHD-H3K4me3 interaction. Finally, the biological relevance of ING3PHD-H3K4me3 binding was demonstrated by the failure of ING3PHD mutant proteins to enhance ING3-mediated DNA damage-dependent cell death. Together, our results reveal the molecular mechanism of H3K4me3 selection by the ING3PHD and suggest that this interaction is important for mediating ING3 tumor suppressive activities.


Subject(s)
Histones/chemistry , Homeodomain Proteins/chemistry , Tumor Suppressor Proteins/chemistry , Amino Acid Substitution , Cell Death , DNA Damage , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Methylation , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , RING Finger Domains , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Pharm Res ; 32(3): 863-75, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25186441

ABSTRACT

PURPOSE: Curcumin is an ideal chemopreventive and antitumor agent characterized by poor bioavailability and low stability. The development of synthetic structural analogues like dimethoxycurcumin (DMC) could overcome these drawbacks. In this study we compared the cytotoxicity, metabolism and the epigenetic changes induced by both drugs in leukemia cells. METHODS: Apoptosis and cell cycle analysis were analyzed by flow cytometry. Real-time PCR was used for gene expression analysis. DNA methylation was analyzed by DNA pyrosequencing. The metabolic stability was determined using human pooled liver microsomes. Chromatin Immunoprecipitation was used to quantify histone methylation. RESULTS: Clinically relevant concentration of curcumin and DMC were not cytotoxic to leukemia cells and induced G2/M cell cycle arrest. DMC was more metabolically stable than curcumin. Curcumin and DMC were devoid of DNA hypomethylating activity. DMC induced the expression of promoter methylated genes without reversing DNA methylation and increased H3K36me3 mark near the promoter region of hypermethylated genes. CONCLUSION: DMC is a more stable analogue of curcumin that can induce epigenetic changes not induced by curcumin. DMC induced the expression of promoter methylated genes. The combination of DMC with DNA methyltransferase inhibitors could harness their combined induced epigenetic changes for optimal re-expression of epigenetically silenced genes.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/analogs & derivatives , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia/genetics , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Apoptosis/drug effects , Biotransformation , Cell Line, Tumor , Curcumin/chemistry , Curcumin/metabolism , Curcumin/pharmacology , Dose-Response Relationship, Drug , Drug Stability , G2 Phase Cell Cycle Checkpoints/drug effects , Histones/metabolism , Humans , Leukemia/metabolism , Leukemia/pathology , Methylation , Microsomes, Liver/metabolism , Promoter Regions, Genetic , Time Factors
9.
J Cell Physiol ; 229(11): 1571-4, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24633655

ABSTRACT

The monocytic leukemic zinc finger (MOZ) histone acetyltransferase (HAT) plays a role in acute myeloid leukemia (AML). It functions as a quaternary complex with the bromodomain PHD finger protein 1 (BRPF1), the human Esa1-associated factor 6 homolog (hEAF6), and the inhibitor of growth 5 (ING5). Each of these subunits contain chromatin reader domains that recognize specific post-translational modifications (PTMs) on histone tails, and this recognition directs the MOZ HAT complex to specific chromatin substrates. The structure and function of these epigenetic reader modules has now been elucidated, and a model describing how the cooperative action of these domains regulates HAT activity in response to the epigenetic landscape is proposed. The emerging role of epigenetic reader domains in disease, and their therapeutic potential for many types of cancer is also highlighted.


Subject(s)
Disease/genetics , Epigenesis, Genetic , Histone Acetyltransferases/metabolism , Signal Transduction/genetics , Animals , Disease Progression , Histone Acetyltransferases/chemistry , Humans , Models, Genetic
10.
J Inorg Biochem ; 258: 112622, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38852293

ABSTRACT

The human cytochrome P450 (CYP) 1, 2 and 3 families of enzymes are responsible for the biotransformation of a majority of the currently available pharmaceutical drugs. The highly polymorphic CYP2C9 predominantly metabolizes many drugs including anticoagulant S-warfarin, anti-hypertensive losartan, anti-diabetic tolbutamide, analgesic ibuprofen, etc. There are >80 single nucleotide changes identified in CYP2C9, many of which significantly alter the clearance of important drugs. Here we report the structural and biophysical analysis of two polymorphic variants, CYP2C9*14 (Arg125His) and CYP2C9*27 (Arg150Leu) complexed with losartan. The X-ray crystal structures of the CYP2C9*14 and *27 illustrate the binding of two losartan molecules, one in the active site near heme and another on the periphery. Both losartan molecules are bound in an identical conformation to that observed in the previously solved CYP2C9 wild-type complex, however, the number of losartan differs from the wild-type structure, which showed binding of three molecules. Additionally, isothermal titration calorimetry experiments reveal a lower binding affinity of losartan with *14 and *27 variants when compared to the wild-type. Overall, the results provide new insights into the effects of these genetic polymorphisms and suggests a possible mechanism contributing to reduced metabolic activity in patients carrying these alleles.


Subject(s)
Cytochrome P-450 CYP2C9 , Losartan , Losartan/chemistry , Losartan/metabolism , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/chemistry , Humans , Crystallography, X-Ray , Protein Binding
11.
J Med Chem ; 67(10): 8186-8200, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38733345

ABSTRACT

The ATPase family AAA+ domain containing 2 (ATAD2) protein and its paralog ATAD2B have a C-terminal bromodomain (BRD) that functions as a reader of acetylated lysine residues on histone proteins. Using a structure-function approach, we investigated the ability of the ATAD2/B BRDs to select acetylated lysine among multiple histone post-translational modifications. The ATAD2B BRD can bind acetylated histone ligands that also contain adjacent methylation or phosphorylation marks, while the presence of these modifications significantly weakened the acetyllysine binding activity of the ATAD2 BRD. Our structural studies provide mechanistic insights into how ATAD2/B BRD-binding pocket residues coordinate the acetyllysine group in the context of adjacent post-translational modifications. Furthermore, we investigated how sequence changes in amino acids of the histone ligands impact the recognition of an adjacent acetyllysine residue. Our study highlights how the interplay between multiple combinations of histone modifications influences the reader activity of the ATAD2/B BRDs, resulting in distinct binding modes.


Subject(s)
ATPases Associated with Diverse Cellular Activities , DNA-Binding Proteins , Histones , Lysine , Histones/metabolism , Histones/chemistry , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Lysine/metabolism , Lysine/chemistry , Acetylation , Protein Processing, Post-Translational , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Protein Binding , Protein Domains , Models, Molecular , Binding Sites
12.
Front Bioinform ; 2: 835892, 2022.
Article in English | MEDLINE | ID: mdl-36304339

ABSTRACT

Background: Bromodomains are a structurally conserved epigenetic reader domain that bind to acetylated lysine residues in both histone and non-histone proteins. Bromodomain-containing proteins (BRD proteins) often function as scaffolding proteins in the assembly of multi-protein complexes to regulate diverse biological processes. BRD proteins have been classified based on biological and functional similarity, however the functions of many BRD proteins remains unknown. PPI network analysis is useful for revealing organizational roles, identifying functional clusters, and predicting function for BRD proteins. Results: We used available data to construct protein-protein interaction networks (PPINs) to study the properties of the human bromodomain protein family. The network properties of the BRD PPIN establishes that the BRD proteins serve as hub proteins that are enriched near the global center to form an inter-connected PPIN. We identified dense subgraphs formed by BRD proteins and find that different BRD proteins share topological similarity and functional associations. We explored the functional relationships through clustering and Hallmark pathway gene set enrichment analysis and identify potential biological roles for different BRD proteins. Conclusion: In our network analysis we confirmed that BRD proteins are conserved central nodes in the human PPI network and function as scaffolds to form distinctive functional clusters. Overall, this study provides detailed insight into the predictive functions of BRD proteins in the context of functional complexes and biological pathways.

13.
Int J Biol Macromol ; 223(Pt A): 316-326, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36328269

ABSTRACT

Plasmodium falciparum requires a two-host system, moving between Anopheles mosquito and humans, to complete its life cycle. To overcome such dynamic growth conditions its histones undergo various post-translational modifications to regulate gene expression. The P. falciparum Bromodomain Protein 1 (PfBDP1) has been shown to interact with acetylated lysine modifications on histone H3 to regulate the expression of invasion-related genes. Here, we investigated the ability of the PfBDP1 bromodomain to interact with acetyllsyine modifications on additional core and variant histones. A crystal structure of the PfBDP1 bromodomain (PfBDP1-BRD) reveals it contains the conserved bromodomain fold, but our comparative analysis between the PfBDP1-BRD and human bromodomain families indicates it has a unique binding mechanism. Solution NMR spectroscopy and ITC binding assays carried out with acetylated histone ligands demonstrate that it preferentially recognizes tetra-acetylated histone H4, and we detected weaker interactions with multi-acetylated H2A.Z in addition to the previously reported interactions with acetylated histone H3. Our findings indicate PfBDP1 may play additional roles in the P. falciparum life cycle, and the distinctive features of its bromodomain binding pocket could be leveraged for the development of new therapeutic agents to help overcome the continuously evolving resistance of P. falciparum against currently available drugs.


Subject(s)
Histones , Plasmodium falciparum , Humans , Histones/metabolism , Ligands , Plasmodium falciparum/metabolism , Protein Binding , Protein Domains , Acetylation , Transcription Factor TFIIIB/metabolism
14.
Results Probl Cell Differ ; 70: 375-396, 2022.
Article in English | MEDLINE | ID: mdl-36348115

ABSTRACT

The cell cycle is governed by stringent epigenetic mechanisms that, in response to intrinsic and extrinsic regulatory cues, support fidelity of DNA replication and cell division. We will focus on (1) the complex and interdependent processes that are obligatory for control of proliferation and compromised in cancer, (2) epigenetic and topological domains that are associated with distinct phases of the cell cycle that may be altered in cancer initiation and progression, and (3) the requirement for mitotic bookmarking to maintain intranuclear localization of transcriptional regulatory machinery to reinforce cell identity throughout the cell cycle to prevent malignant transformation.


Subject(s)
Epigenesis, Genetic , Neoplasms , Humans , Cell Cycle/genetics , Cell Division , Neoplasms/genetics , Neoplasms/pathology , Chromatin , Gene Expression Regulation
15.
Results Probl Cell Differ ; 70: 339-373, 2022.
Article in English | MEDLINE | ID: mdl-36348114

ABSTRACT

Epigenetic gene regulatory mechanisms play a central role in the biological control of cell and tissue structure, function, and phenotype. Identification of epigenetic dysregulation in cancer provides mechanistic into tumor initiation and progression and may prove valuable for a variety of clinical applications. We present an overview of epigenetically driven mechanisms that are obligatory for physiological regulation and parameters of epigenetic control that are modified in tumor cells. The interrelationship between nuclear structure and function is not mutually exclusive but synergistic. We explore concepts influencing the maintenance of chromatin structures, including phase separation, recognition signals, factors that mediate enhancer-promoter looping, and insulation and how these are altered during the cell cycle and in cancer. Understanding how these processes are altered in cancer provides a potential for advancing capabilities for the diagnosis and identification of novel therapeutic targets.


Subject(s)
Epigenesis, Genetic , Neoplasms , Humans , Phenotype , Neoplasms/genetics , Neoplasms/pathology , Gene Expression Regulation , Chromatin
16.
Cancers (Basel) ; 13(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34298819

ABSTRACT

Histone acetylation is generally associated with an open chromatin configuration that facilitates many cellular processes including gene transcription, DNA repair, and DNA replication. Aberrant levels of histone lysine acetylation are associated with the development of cancer. Bromodomains represent a family of structurally well-characterized effector domains that recognize acetylated lysines in chromatin. As part of their fundamental reader activity, bromodomain-containing proteins play versatile roles in epigenetic regulation, and additional functional modules are often present in the same protein, or through the assembly of larger enzymatic complexes. Dysregulated gene expression, chromosomal translocations, and/or mutations in bromodomain-containing proteins have been correlated with poor patient outcomes in cancer. Thus, bromodomains have emerged as a highly tractable class of epigenetic targets due to their well-defined structural domains, and the increasing ease of designing or screening for molecules that modulate the reading process. Recent developments in pharmacological agents that target specific bromodomains has helped to understand the diverse mechanisms that bromodomains play with their interaction partners in a variety of chromatin processes, and provide the promise of applying bromodomain inhibitors into the clinical field of cancer treatment. In this review, we explore the expression and protein interactome profiles of bromodomain-containing proteins and discuss them in terms of functional groups. Furthermore, we highlight our current understanding of the roles of bromodomain-containing proteins in cancer, as well as emerging strategies to specifically target bromodomains, including combination therapies using bromodomain inhibitors alongside traditional therapeutic approaches designed to re-program tumorigenesis and metastasis.

17.
Curr Res Struct Biol ; 2: 104-115, 2020.
Article in English | MEDLINE | ID: mdl-33554132

ABSTRACT

Bromodomain-containing proteins are often part of chromatin-modifying complexes, and their activity can lead to altered expression of genes that drive cancer, inflammation and neurological disorders in humans. Bromodomain-PHD finger protein 1 (BRPF1) is part of the MOZ (monocytic leukemic zinc-finger protein) HAT (histone acetyltransferase) complex, which is associated with chromosomal translocations known to contribute to the development of acute myeloid leukemia (AML). BRPF1 contains a unique combination of chromatin reader domains including two plant homeodomain (PHD) fingers separated by a zinc knuckle (PZP domain), a bromodomain, and a proline-tryptophan-tryptophan-proline (PWWP) domain. BRPF1 is known to recruit the MOZ HAT complex to chromatin by recognizing acetylated lysine residues on the N-terminal histone tail region through its bromodomain. However, histone proteins can contain several acetylation modifications on their N-terminus, and it is unknown how additional marks influence bromodomain recruitment to chromatin. Here, we identify the BRPF1 bromodomain as a selective reader of di-acetyllysine modifications on histone H4. We used ITC assays to characterize the binding of di-acetylated histone ligands to the BRPF1 bromodomain and found that the domain binds preferentially to histone peptides H4K5acK8ac and H4K5acK12ac. Analytical ultracentrifugation (AUC) experiments revealed that the monomeric state of the BRPF1 bromodomain coordinates di-acetylated histone ligands. NMR chemical shift perturbation studies, along with binding and mutational analyses, revealed non-canonical regions of the bromodomain-binding pocket that are important for histone tail recognition. Together, our findings provide critical information on how the combinatorial action of post-translational modifications can modulate BRPF1 bromodomain binding and specificity.

18.
J Med Chem ; 63(21): 12799-12813, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33084328

ABSTRACT

Bromodomains exhibit preferences for specific patterns of post-translational modifications on core and variant histone proteins. We examined the ligand specificity of the ATAD2B bromodomain and compared it to its closely related paralogue in ATAD2. We show that the ATAD2B bromodomain recognizes mono- and diacetyllysine modifications on histones H4 and H2A. A structure-function approach was used to identify key residues in the acetyllysine-binding pocket that dictate the molecular recognition process, and we examined the binding of an ATAD2 bromodomain inhibitor by ATAD2B. Our analysis demonstrated that critical contacts required for bromodomain inhibitor coordination are conserved between the ATAD2/B bromodomains, with many residues playing a dual role in acetyllysine recognition. We further characterized an alternative splice variant of ATAD2B that results in a loss of function. Our results outline the structural and functional features of the ATAD2B bromodomain and identify a novel mechanism regulating the interaction of the ATAD2B protein with chromatin.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/genetics , Acetylation , Alternative Splicing , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Histones/chemistry , Humans , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Protein Domains , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
19.
Curr Pharm Teach Learn ; 10(12): 1565-1573, 2018 12.
Article in English | MEDLINE | ID: mdl-30527821

ABSTRACT

INTRODUCTION: In this study, we introduced a student self-directed interactive animation tool created to enhance student understanding of cytochrome P450 (CYP450) mediated drug metabolism. METHODS: The online learning tool was constructed in HTML5 computer code. It was implemented over four years in a second year pharmacy degree course where CYP450 metabolism was taught. Assessment was by comparing test scores of students using the learning tool with a previous class that did not and through survey data from the student users. RESULTS: Use of the Cyber-CYP learning tool enhanced test performance on CYP450 metabolism-related questions in all years tested. Survey responses indicated that the learning tool was easy to use and facilitated student learning of CYP450-mediated drug metabolism. CONCLUSIONS: This study has shown that complex and dynamic processes, such as CYP450 metabolism, can be more effectively communicated using student-centered, self-paced and interactive animations.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Learning , Metabolism/drug effects , Problem-Based Learning/methods , Education, Distance/methods , Education, Distance/standards , Education, Distance/trends , Education, Pharmacy/methods , Education, Pharmacy/standards , Educational Measurement/methods , Humans , Problem-Based Learning/standards , Problem-Based Learning/trends , Surveys and Questionnaires
20.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 10): 1389-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25286946

ABSTRACT

The bromodomain-PHD finger protein 1 (BRPF1) is an essential subunit of the monocytic leukemia zinc (MOZ) histone acetyltransferase (HAT) complex and is required for complex formation and enzymatic activation. BRPF1 contains a structurally conserved bromodomain, which recognizes specific acetyllysine residues on histone proteins. The MOZ HAT plays a direct role in hematopoiesis, and deregulation of its activity is linked to the development of acute myeloid leukemia. However, the molecular mechanism of histone-ligand recognition by the BRPF1 bromodomain is currently unknown. The 117-amino-acid BRPF1 bromodomain was overexpressed in Escherichia coli and purified to homogeneity. Crystallization experiments of the BRPF1 bromodomain in complex with its H4K12ac and H2AK5ac histone ligands yielded crystals that were suitable for high-resolution X-ray diffraction analysis. The BRPF1 bromodomain-H4K12ac crystals belonged to the tetragonal space group P43212, with unit-cell parameters a = 75.1, b = 75.1, c = 86.3 Å, and diffracted to a resolution of 1.94 Å. The BRPF1 bromodomain-H2AK5ac crystals grew in the monoclinic space group P21, with unit-cell parameters a = 60.9, b = 55.6, c = 82.1 Å, ß = 93.6°, and diffracted to a resolution of 1.80 Å. Complete data sets were collected from both crystal forms using synchrotron radiation on beamline X29 at Brookhaven National Laboratory (BNL).


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Histones/chemistry , Nuclear Proteins/chemistry , Crystallization , Crystallography, X-Ray , DNA-Binding Proteins , Humans , Ligands , Peptide Fragments/chemistry , Protein Binding , Protein Interaction Domains and Motifs
SELECTION OF CITATIONS
SEARCH DETAIL