Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38712034

ABSTRACT

Disease-causing missense mutations that occur within structurally and functionally unannotated protein regions can guide researchers to new mechanisms of protein regulation and dysfunction. Here, we report that the thrombocytopenia-, myelodysplastic syndromes-, and leukemia-associated P214L mutation in the transcriptional regulator ETV6 creates an XPO1-dependent nuclear export signal to cause protein mislocalization. Strategies to disrupt XPO1 activity fully restore ETV6 P214L protein nuclear localization and transcription regulation activity. Mechanistic insight inspired the design of a 'humanized' ETV6 mice, which we employ to demonstrate that the germline P214L mutation is sufficient to elicit severe defects in thrombopoiesis and hematopoietic stem cell maintenance. Beyond ETV6, we employed computational methods to uncover rare disease-associated missense mutations in unrelated proteins that create a nuclear export signal to disrupt protein function. Thus, missense mutations that operate through this mechanism should be predictable and may suggest rational therapeutic strategies for associated diseases.

2.
Nat Commun ; 15(1): 1000, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38307862

ABSTRACT

The chromatin remodeler ALC1 is activated by DNA damage-induced poly(ADP-ribose) deposited by PARP1/PARP2 and their co-factor HPF1. ALC1 has emerged as a cancer drug target, but how it is recruited to ADP-ribosylated nucleosomes to affect their positioning near DNA breaks is unknown. Here we find that PARP1/HPF1 preferentially initiates ADP-ribosylation on the histone H2B tail closest to the DNA break. To dissect the consequences of such asymmetry, we generate nucleosomes with a defined ADP-ribosylated H2B tail on one side only. The cryo-electron microscopy structure of ALC1 bound to such an asymmetric nucleosome indicates preferential engagement on one side. Using single-molecule FRET, we demonstrate that this asymmetric recruitment gives rise to directed sliding away from the DNA linker closest to the ADP-ribosylation site. Our data suggest a mechanism by which ALC1 slides nucleosomes away from a DNA break to render it more accessible to repair factors.


Subject(s)
Nucleosomes , Poly ADP Ribosylation , Nucleosomes/genetics , Cryoelectron Microscopy , Poly (ADP-Ribose) Polymerase-1/metabolism , Chromatin , DNA Repair , DNA Breaks
3.
J Am Chem Soc ; 145(25): 14000-14009, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37315125

ABSTRACT

We report here chemoenzymatic and fully synthetic methodologies to modify aspartate and glutamate side chains with ADP-ribose at specific sites on peptides. Structural analysis of aspartate and glutamate ADP-ribosylated peptides reveals near-quantitative migration of the side chain linkage from the anomeric carbon to the 2″- or 3″-ADP-ribose hydroxyl moieties. We find that this linkage migration pattern is unique to aspartate and glutamate ADP-ribosylation and propose that the observed isomer distribution profile is present in biochemical and cellular environments. After defining distinct stability properties of aspartate and glutamate ADP-ribosylation, we devise methods to install homogenous ADP-ribose chains at specific glutamate sites and assemble glutamate-modified peptides into full-length proteins. By implementing these technologies, we show that histone H2B E2 tri-ADP-ribosylation is able to stimulate the chromatin remodeler ALC1 with similar efficiency to histone serine ADP-ribosylation. Our work reveals fundamental principles of aspartate and glutamate ADP-ribosylation and enables new strategies to interrogate the biochemical consequences of this widespread protein modification.


Subject(s)
Aspartic Acid , Glutamic Acid , Aspartic Acid/metabolism , Glutamic Acid/metabolism , ADP-Ribosylation , Histones/metabolism , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Peptides/chemistry
4.
Science ; 377(6601): eabn5582, 2022 07.
Article in English | MEDLINE | ID: mdl-35771920

ABSTRACT

Protein domains of low sequence complexity do not fold into stable, three-dimensional structures. Nevertheless, proteins with these sequences assist in many aspects of cell organization, including assembly of nuclear and cytoplasmic structures not surrounded by membranes. The dynamic nature of these cellular assemblies is caused by the ability of low-complexity domains (LCDs) to transiently self-associate through labile, cross-ß structures. Mechanistic studies useful for the study of LCD self-association have evolved over the past decade in the form of simple assays of phase separation. Here, we have used such assays to demonstrate that the interactions responsible for LCD self-association can be dictated by labile protein structures poised close to equilibrium between the folded and unfolded states. Furthermore, missense mutations causing Charcot-Marie-Tooth disease, frontotemporal dementia, and Alzheimer's disease manifest their pathophysiology in vitro and in cultured cell systems by enhancing the stability of otherwise labile molecular structures formed upon LCD self-association.


Subject(s)
Alzheimer Disease , Charcot-Marie-Tooth Disease , DNA-Binding Proteins , Frontotemporal Dementia , Alzheimer Disease/genetics , Cells, Cultured , Charcot-Marie-Tooth Disease/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Frontotemporal Dementia/genetics , Humans , Mutation, Missense , Protein Domains , Protein Folding , Protein Stability
5.
ACS Chem Biol ; 17(4): 810-815, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35312285

ABSTRACT

Recently developed chemical and enzyme-based technologies to install serine ADP-ribosylation onto synthetic peptides have enabled new approaches to study poly(ADP-ribose) polymerase (PARP) biology. Here, we establish a generalizable strategy to prepare ADP-ribosylated peptides that are compatible with N-terminal, C-terminal, and sequential protein ligation reactions. Two unique protein-assembly routes are employed to generate full-length linker histone constructs that are homogeneously ADP-ribosylated at known DNA damage-dependent modification sites. We found that serine mono-ADP-ribosylation is sufficient to alleviate linker histone-dependent chromatin compaction and that this effect is amplified by ADP-ribose chain elongation. Our work will greatly expand the scope of ADP-ribose-modified proteins that can be constructed via semisynthesis, which is rapidly emerging as a robust approach to elucidate the direct effects that site-specific serine mono- and poly-ADP-ribosylation have on protein function.


Subject(s)
Histones , Serine , ADP-Ribosylation , Adenosine Diphosphate Ribose/metabolism , Histones/metabolism , Peptides/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Serine/metabolism
6.
Elife ; 102021 12 07.
Article in English | MEDLINE | ID: mdl-34874266

ABSTRACT

Serine ADP-ribosylation (ADPr) is a DNA damage-induced post-translational modification catalyzed by the PARP1/2:HPF1 complex. As the list of PARP1/2:HPF1 substrates continues to expand, there is a need for technologies to prepare mono- and poly-ADP-ribosylated proteins for biochemical interrogation. Here, we investigate the unique peptide ADPr activities catalyzed by PARP1 in the absence and presence of HPF1. We then exploit these activities to develop a method that facilitates installation of ADP-ribose polymers onto peptides with precise control over chain length and modification site. Importantly, the enzymatically mono- and poly-ADP-ribosylated peptides are fully compatible with protein ligation technologies. This chemoenzymatic protein synthesis strategy was employed to assemble a series of full-length, ADP-ribosylated histones and show that ADPr at histone H2B serine 6 or histone H3 serine 10 converts nucleosomes into robust substrates for the chromatin remodeler ALC1. We found ALC1 preferentially remodels 'activated' substrates within heterogeneous mononucleosome populations and asymmetrically ADP-ribosylated dinucleosome substrates, and that nucleosome serine ADPr is sufficient to stimulate ALC1 activity in nuclear extracts. Our study identifies a biochemical function for nucleosome serine ADPr and describes a new, highly modular approach to explore the impact that site-specific serine mono- and poly-ADPr have on protein function.


Subject(s)
ADP-Ribosylation , Chromatin Assembly and Disassembly , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Nucleosomes/metabolism , Serine/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans
7.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593918

ABSTRACT

Low complexity (LC) head domains 92 and 108 residues in length are, respectively, required for assembly of neurofilament light (NFL) and desmin intermediate filaments (IFs). As studied in isolation, these IF head domains interconvert between states of conformational disorder and labile, ß-strand-enriched polymers. Solid-state NMR (ss-NMR) spectroscopic studies of NFL and desmin head domain polymers reveal spectral patterns consistent with structural order. A combination of intein chemistry and segmental isotope labeling allowed preparation of fully assembled NFL and desmin IFs that could also be studied by ss-NMR. Assembled IFs revealed spectra overlapping with those observed for ß-strand-enriched polymers formed from the isolated NFL and desmin head domains. Phosphorylation and disease-causing mutations reciprocally alter NFL and desmin head domain self-association yet commonly impede IF assembly. These observations show how facultative structural assembly of LC domains via labile, ß-strand-enriched self-interactions may broadly influence cell morphology.


Subject(s)
Desmin/chemistry , Desmin/metabolism , Intermediate Filaments/chemistry , Intermediate Filaments/metabolism , Humans , Phosphorylation , Protein Conformation , Protein Domains
8.
Nat Chem Biol ; 16(2): 134-142, 2020 02.
Article in English | MEDLINE | ID: mdl-31819269

ABSTRACT

Recent studies have implicated the nucleosome acidic patch in the activity of ATP-dependent chromatin remodeling machines. We used a photocrosslinking-based nucleosome profiling technology (photoscanning) to identify a conserved basic motif within the catalytic subunit of ISWI remodelers, SNF2h, which engages this nucleosomal epitope. This region of SNF2h is essential for chromatin remodeling activity in a reconstituted biochemical system and in cells. Our studies suggest that the basic motif in SNF2h plays a critical role in anchoring the remodeler to the nucleosomal surface. We also examine the functional consequences of several cancer-associated histone mutations that map to the nucleosome acidic patch. Kinetic studies using physiologically relevant heterotypic nucleosomal substrates ('Janus' nucleosomes) indicate that these cancer-associated mutations can disrupt regularly spaced chromatin structure by inducing ISWI-mediated unidirectional nucleosome sliding. These results indicate a potential mechanistic link between oncogenic histones and alterations to the chromatin landscape.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Nucleosomes/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Motifs , Binding Sites , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Cysteine/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histones/metabolism , Humans , Nucleosomes/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Angew Chem Int Ed Engl ; 58(13): 4144-4162, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30153374

ABSTRACT

The emergence of high-throughput DNA sequencing technologies sparked a revolution in the field of genomics that has rippled into many branches of the life and physical sciences. The remarkable sensitivity, specificity, throughput, and multiplexing capacity that are inherent to parallel DNA sequencing have since motivated its use as a broad-spectrum molecular counter. A key aspect of extrapolating DNA sequencing to non-traditional applications is the need to append nucleic-acid barcodes to entities of interest. In this review, we describe the chemical and biochemical approaches that have enabled nucleic-acid barcoding of proteinaceous and non-proteinaceous materials and provide examples of downstream technologies that have been made possible by DNA-encoded molecules. As commercially available high-throughput sequencers were first released less than 15 years ago, we believe related applications will continue to mature and close by proposing new frontiers to support this assertion.


Subject(s)
DNA Barcoding, Taxonomic/methods , High-Throughput Nucleotide Sequencing/methods , Nucleic Acids/analysis , Proteins/metabolism , Animals , Computational Biology , Humans , Proteins/genetics
10.
Nat Chem Biol ; 14(9): 837-840, 2018 09.
Article in English | MEDLINE | ID: mdl-30013063

ABSTRACT

Recent studies report serine ADP-ribosylation on nucleosomes during the DNA damage response. We unveil histone H3 serine 10 as the primary acceptor residue for chromatin ADP-ribosylation and find that specific histone acetylation marks block this activity. Our results provide a molecular explanation for the well-documented phenomenon of rapid deacetylation at DNA damage sites and support the combinatorial application of PARP and HDAC inhibitors for the treatment of PARP-dependent cancers.


Subject(s)
ADP-Ribosylation , Chromatin/metabolism , DNA Damage , Histones/metabolism , Acetylation , Cloning, Molecular , DNA/genetics , DNA/metabolism , Histones/genetics , Humans , Tumor Cells, Cultured
11.
Nature ; 548(7669): 607-611, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28767641

ABSTRACT

ATP-dependent chromatin remodellers regulate access to genetic information by controlling nucleosome positions in vivo. However, the mechanism by which remodellers discriminate between different nucleosome substrates is poorly understood. Many chromatin remodelling proteins possess conserved protein domains that interact with nucleosomal features. Here we used a quantitative high-throughput approach, based on the use of a DNA-barcoded mononucleosome library, to profile the biochemical activity of human ISWI family remodellers in response to a diverse set of nucleosome modifications. We show that accessory (non-ATPase) subunits of ISWI remodellers can distinguish between differentially modified nucleosomes, directing remodelling activity towards specific nucleosome substrates according to their modification state. Unexpectedly, we show that the nucleosome acidic patch is necessary for maximum activity of all ISWI remodellers evaluated. This dependence also extends to CHD and SWI/SNF family remodellers, suggesting that the acidic patch may be generally required for chromatin remodelling. Critically, remodelling activity can be regulated by modifications neighbouring the acidic patch, signifying that it may act as a tunable interaction hotspot for ATP-dependent chromatin remodellers and, by extension, many other chromatin effectors that engage this region of the nucleosome surface.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Nucleosomes/chemistry , Nucleosomes/metabolism , Substrate Specificity , Transcription Factors/metabolism , DNA Barcoding, Taxonomic , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/genetics , Protein Subunits/metabolism
12.
Proc Natl Acad Sci U S A ; 114(4): 681-686, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069948

ABSTRACT

Recent advances in the field of programmable DNA-binding proteins have led to the development of facile methods for genomic localization of genetically encodable entities. Despite the extensive utility of these tools, locus-specific delivery of synthetic molecules remains limited by a lack of adequate technologies. Here we combine the flexibility of chemical synthesis with the specificity of a programmable DNA-binding protein by using protein trans-splicing to ligate synthetic elements to a nuclease-deficient Cas9 (dCas9) in vitro and subsequently deliver the dCas9 cargo to live cells. The versatility of this technology is demonstrated by delivering dCas9 fusions that include either the small-molecule bromodomain and extra-terminal family bromodomain inhibitor JQ1 or a peptide-based PRC1 chromodomain ligand, which are capable of recruiting endogenous copies of their cognate binding partners to targeted genomic binding sites. We expect that this technology will allow for the genomic localization of a wide array of small molecules and modified proteinaceous materials.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/physiology , Amino Acid Sequence , Binding Sites , DNA-Binding Proteins/metabolism , Epigenomics/methods , Escherichia coli/metabolism , Genomics/methods , Recombinant Proteins/metabolism
13.
J Biol Chem ; 291(51): 26468-26477, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27803161

ABSTRACT

Protein acetylation is a prevalent posttranslational modification that is regulated by diverse acetyltransferase enzymes. Although histone acetyltransferases (HATs) have been well characterized both structurally and mechanistically, far less is known about non-histone acetyltransferase enzymes. The human ESCO1 and ESCO2 paralogs acetylate the cohesin complex subunit SMC3 to regulate the separation of sister chromatids during mitosis and meiosis. Missense mutations within the acetyltransferase domain of these proteins correlate with diseases, including endometrial cancers and Roberts syndrome. Despite their biological importance, the mechanisms underlying acetylation by the ESCO proteins are not understood. Here, we report the X-ray crystal structure of the highly conserved zinc finger-acetyltransferase moiety of ESCO1 with accompanying structure-based mutagenesis and biochemical characterization. We find that the ESCO1 acetyltransferase core is structurally homologous to the Gcn5 HAT, but contains unique additional features including a zinc finger and an ∼40-residue loop region that appear to play roles in protein stability and SMC3 substrate binding. We identify key residues that play roles in substrate binding and catalysis, and rationalize the functional consequences of disease-associated mutations. Together, these studies reveal the molecular basis for SMC3 acetylation by ESCO1 and have broader implications for understanding the structure/function of non-histone acetyltransferases.


Subject(s)
Acetyltransferases/chemistry , Cell Cycle Proteins/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Acetylation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amino Acid Substitution , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , Humans , Mutation, Missense , Protein Domains , Structural Homology, Protein , Structure-Activity Relationship , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism , Cohesins
14.
Nat Chem Biol ; 12(3): 188-93, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26807716

ABSTRACT

Specialized chromatin domains contribute to nuclear organization and regulation of gene expression. Gene-poor regions are di- and trimethylated at lysine 9 of histone H3 (H3K9me2 and H3K9me3) by the histone methyltransferase Suv39h1. This enzyme harnesses a positive feedback loop to spread H3K9me2 and H3K9me3 over extended heterochromatic regions. However, little is known about how feedback loops operate on complex biopolymers such as chromatin, in part because of the difficulty in obtaining suitable substrates. Here we describe the synthesis of multidomain 'designer chromatin' templates and their application to dissecting the regulation of human Suv39h1. We uncovered a two-step activation switch where H3K9me3 recognition and subsequent anchoring of the enzyme to chromatin allosterically promotes methylation activity and confirmed that this mechanism contributes to chromatin recognition in cells. We propose that this mechanism serves as a paradigm in chromatin biochemistry, as it enables highly dynamic sampling of chromatin state combined with targeted modification of desired genomic regions.


Subject(s)
Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Enzyme Activation/physiology , Feedback, Physiological , Genomic Structural Variation , Histones/metabolism , Humans , Insecta , Methylation , Methyltransferases/genetics , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Repressor Proteins/genetics
15.
Structure ; 23(2): 332-41, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25619998

ABSTRACT

N-terminal acetylation is among the most common protein modifications in eukaryotes and is mediated by evolutionarily conserved N-terminal acetyltransferases (NATs). NatD is among the most selective NATs; its only known substrates are histones H4 and H2A, containing the N-terminal sequence SGRGK in humans. Here we characterize the molecular basis for substrate-specific acetylation by NatD by reporting its crystal structure bound to cognate substrates and performing related biochemical studies. A novel N-terminal segment wraps around the catalytic core domain to make stabilizing interactions, and the α1-α2 and ß6-ß7 loops adopt novel conformations to properly orient the histone N termini in the binding site. Ser1 and Arg3 of the histone make extensive contacts to highly conserved NatD residues in the substrate binding pocket, and flanking glycine residues also appear to contribute to substrate-specific binding by NatD, together defining a Ser-Gly-Arg-Gly recognition sequence. These studies have implications for understanding substrate-specific acetylation by NAT enzymes.


Subject(s)
Histones/chemistry , N-Terminal Acetyltransferase D/chemistry , Schizosaccharomyces/enzymology , Acetylation , Amino Acid Sequence , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , N-Terminal Acetyltransferase D/genetics , N-Terminal Acetyltransferase D/metabolism , Sequence Alignment , Species Specificity , Static Electricity , Substrate Specificity
16.
Hum Mol Genet ; 24(7): 1956-76, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25489052

ABSTRACT

The X-linked lethal Ogden syndrome was the first reported human genetic disorder associated with a mutation in an N-terminal acetyltransferase (NAT) gene. The affected males harbor an Ser37Pro (S37P) mutation in the gene encoding Naa10, the catalytic subunit of NatA, the major human NAT involved in the co-translational acetylation of proteins. Structural models and molecular dynamics simulations of the human NatA and its S37P mutant highlight differences in regions involved in catalysis and at the interface between Naa10 and the auxiliary subunit hNaa15. Biochemical data further demonstrate a reduced catalytic capacity and an impaired interaction between hNaa10 S37P and Naa15 as well as Naa50 (NatE), another interactor of the NatA complex. N-Terminal acetylome analyses revealed a decreased acetylation of a subset of NatA and NatE substrates in Ogden syndrome cells, supporting the genetic findings and our hypothesis regarding reduced Nt-acetylation of a subset of NatA/NatE-type substrates as one etiology for Ogden syndrome. Furthermore, Ogden syndrome fibroblasts display abnormal cell migration and proliferation capacity, possibly linked to a perturbed retinoblastoma pathway. N-Terminal acetylation clearly plays a role in Ogden syndrome, thus revealing the in vivo importance of N-terminal acetylation in human physiology and disease.


Subject(s)
Genetic Diseases, X-Linked/metabolism , Proteins/metabolism , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amino Acid Motifs , Catalytic Domain , Female , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , Humans , Male , Mutation , Pedigree , Proteins/chemistry , Proteins/genetics
17.
Nat Struct Mol Biol ; 20(9): 1098-105, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23912279

ABSTRACT

N-terminal acetylation is ubiquitous among eukaryotic proteins and controls a myriad of biological processes. Of the N-terminal acetyltransferases (NATs) that facilitate this cotranslational modification, the heterodimeric NatA complex has the most diversity for substrate selection and modifies the majority of all N-terminally acetylated proteins. Here, we report the X-ray crystal structure of the 100-kDa holo-NatA complex from Schizosaccharomyces pombe, in the absence and presence of a bisubstrate peptide-CoA-conjugate inhibitor, as well as the structure of the uncomplexed Naa10p catalytic subunit. The NatA-Naa15p auxiliary subunit contains 13 tetratricopeptide motifs and adopts a ring-like topology that wraps around the NatA-Naa10p subunit, an interaction that alters the Naa10p active site for substrate-specific acetylation. These studies have implications for understanding the mechanistic details of other NAT complexes and how regulatory subunits modulate the activity of the broader family of protein acetyltransferases.


Subject(s)
Acetyltransferases/chemistry , N-Terminal Acetyltransferase A/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Acetyltransferases/genetics , Acetyltransferases/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase A/metabolism , Protein Structure, Quaternary , Protein Subunits , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
18.
Proc Natl Acad Sci U S A ; 110(36): 14652-7, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959863

ABSTRACT

Amino-terminal acetylation is a ubiquitous modification in eukaryotes that is involved in a growing number of biological processes. There are six known eukaryotic amino-terminal acetyltransferases (NATs), which are differentiated from one another on the basis of substrate specificity. To date, two eukaryotic NATs, NatA and NatE, have been structurally characterized, of which NatA will acetylate the α-amino group of a number of nonmethionine amino-terminal residue substrates such as serine; NatE requires a substrate amino-terminal methionine residue for activity. Interestingly, these two NATs use different catalytic strategies to accomplish substrate-specific acetylation. In archaea, where this modification is less prevalent, only one NAT enzyme has been identified. Surprisingly, this enzyme is able to acetylate NatA and NatE substrates and is believed to represent an ancestral NAT variant from which the eukaryotic NAT machinery evolved. To gain insight into the evolution of NAT enzymes, we determined the X-ray crystal structure of an archaeal NAT from Sulfolobus solfataricus (ssNAT). Through the use of mutagenesis and kinetic analysis, we show that the active site of ssNAT represents a hybrid of the NatA and NatE active sites, and we highlight features of this protein that allow it to facilitate catalysis of distinct substrates through different catalytic strategies, which is a unique characteristic of this enzyme. Taken together, the structural and biochemical data presented here have implications for the evolution of eukaryotic NAT enzymes and the substrate specificities therein.


Subject(s)
Archaeal Proteins/chemistry , N-Terminal Acetyltransferases/chemistry , Protein Structure, Tertiary , Sulfolobus solfataricus/enzymology , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Acetylation , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Catalytic Domain/genetics , Crystallography, X-Ray , Eukaryotic Cells/enzymology , Evolution, Molecular , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , N-Terminal Acetyltransferase A/chemistry , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/chemistry , N-Terminal Acetyltransferase E/genetics , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , Protein Binding , Protein Multimerization , Sequence Homology, Amino Acid
19.
PLoS One ; 7(12): e52642, 2012.
Article in English | MEDLINE | ID: mdl-23285125

ABSTRACT

Nt-acetylation is among the most common protein modifications in eukaryotes. Although thought for a long time to protect proteins from degradation, the role of Nt-acetylation is still debated. It is catalyzed by enzymes called N-terminal acetyltransferases (NATs). In eukaryotes, several NATs, composed of at least one catalytic domain, target different substrates based on their N-terminal sequences. In order to better understand the substrate specificity of human NATs, we investigated in silico the enzyme-substrate interactions in four catalytic subunits of human NATs (Naa10p, Naa20p, Naa30p and Naa50p). To date hNaa50p is the only human subunit for which X-ray structures are available. We used the structure of the ternary hNaa50p/AcCoA/MLG complex and a structural model of hNaa10p as a starting point for multiple molecular dynamics simulations of hNaa50p/AcCoA/substrate (substrate=MLG, EEE, MKG), hNaa10p/AcCoA/substrate (substrate=MLG, EEE). Nine alanine point-mutants of the hNaa50p/AcCoA/MLG complex were also simulated. Homology models of hNaa20p and hNaa30p were built and compared to hNaa50p and hNaa10p. The simulations of hNaa50p/AcCoA/MLG reproduce the interactions revealed by the X-ray data. We observed strong hydrogen bonds between MLG and tyrosines 31, 138 and 139. Yet the tyrosines interacting with the substrate's backbone suggest that their role in specificity is limited. This is confirmed by the simulations of hNaa50p/AcCoA/EEE and hNaa10p/AcCoA/MLG, where these hydrogen bonds are still observed. Moreover these tyrosines are all conserved in hNaa20p and hNaa30p. Other amino acids tune the specificity of the S1' sites that is different for hNaa10p (acidic), hNaa20p (hydrophobic/basic), hNaa30p (basic) and hNaa50p (hydrophobic). We also observe dynamic correlation between the ligand binding site and helix [Formula: see text] that tightens under substrate binding. Finally, by comparing the four structures we propose maps of the peptide-enzyme interactions that should help rationalizing substrate-specificity and lay the ground for inhibitor design.


Subject(s)
Catalytic Domain , N-Terminal Acetyltransferases/chemistry , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Amino Acid Sequence , Binding Sites , Humans , Hydrogen Bonding , Kinetics , Ligands , Molecular Dynamics Simulation , Molecular Sequence Data , N-Terminal Acetyltransferase E/chemistry , N-Terminal Acetyltransferase E/genetics , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferases/genetics , N-Terminal Acetyltransferases/metabolism , Nitrogen/chemistry , Peptides/chemistry , Point Mutation , Protein Binding , Sequence Alignment , Substrate Specificity
20.
J Biol Chem ; 286(42): 37002-10, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21900231

ABSTRACT

The co-translational modification of N-terminal acetylation is ubiquitous among eukaryotes and has been reported to have a wide range of biological effects. The human N-terminal acetyltransferase (NAT) Naa50p (NAT5/SAN) acetylates the α-amino group of proteins containing an N-terminal methionine residue and is essential for proper sister chromatid cohesion and chromosome condensation. The elevated activity of NATs has also been correlated with cancer, making these enzymes attractive therapeutic targets. We report the x-ray crystal structure of Naa50p bound to a native substrate peptide fragment and CoA. We found that the peptide backbone of the substrate is anchored to the protein through a series of backbone hydrogen bonds with the first methionine residue specified through multiple van der Waals contacts, together creating an α-amino methionine-specific pocket. We also employed structure-based mutagenesis; the results support the importance of the α-amino methionine-specific pocket of Naa50p and are consistent with the proposal that conserved histidine and tyrosine residues play important catalytic roles. Superposition of the ternary Naa50p complex with the peptide-bound Gcn5 histone acetyltransferase revealed that the two enzymes share a Gcn5-related N-acetyltransferase fold but differ in their respective substrate-binding grooves such that Naa50p can accommodate only an α-amino substrate and not a side chain lysine substrate that is acetylated by lysine acetyltransferase enzymes such as Gcn5. The structure of the ternary Naa50p complex also provides the first molecular scaffold for the design of NAT-specific small molecule inhibitors with possible therapeutic applications.


Subject(s)
Acetyltransferases/chemistry , Peptides/chemistry , Protein Folding , p300-CBP Transcription Factors/chemistry , Acetylation , Acetyltransferases/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Humans , Hydrogen Bonding , N-Terminal Acetyltransferase E , Peptides/metabolism , Protein Binding , Protein Structure, Quaternary , Substrate Specificity , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...