Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Commun ; 9(1): 4574, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385749

ABSTRACT

The YEATS domain has been identified as a reader of histone acylation and more recently emerged as a promising anti-cancer therapeutic target. Here, we detail the structural mechanisms for π-π-π stacking involving the YEATS domains of yeast Taf14 and human AF9 and acylated histone H3 peptides and explore DNA-binding activities of these domains. Taf14-YEATS selects for crotonyllysine, forming π stacking with both the crotonyl amide and the alkene moiety, whereas AF9-YEATS exhibits comparable affinities to saturated and unsaturated acyllysines, engaging them through π stacking with the acyl amide. Importantly, AF9-YEATS is capable of binding to DNA, whereas Taf14-YEATS is not. Using a structure-guided approach, we engineered a mutant of Taf14-YEATS that engages crotonyllysine through the aromatic-aliphatic-aromatic π stacking and shows high selectivity for the crotonyl H3K9 modification. Our findings shed light on the molecular principles underlying recognition of acyllysine marks and reveal a previously unidentified DNA-binding activity of AF9-YEATS.


Subject(s)
DNA/metabolism , Histone Code , Nuclear Proteins/metabolism , Protein Domains , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIID/metabolism , Acetylation , Acylation , Crystallography, X-Ray , DNA/ultrastructure , Humans , Lysine/metabolism , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/ultrastructure , Protein Binding , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factor TFIID/ultrastructure
2.
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
3.
Nat Commun ; 8(1): 1088, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057918

ABSTRACT

Recognition of modified histones by "reader" proteins constitutes a key mechanism regulating diverse chromatin-associated processes important for normal and neoplastic development. We recently identified the YEATS domain as a novel acetyllysine-binding module; however, the functional importance of YEATS domain-containing proteins in human cancer remains largely unknown. Here, we show that the YEATS2 gene is highly amplified in human non-small cell lung cancer (NSCLC) and is required for cancer cell growth and survival. YEATS2 binds to acetylated histone H3 via its YEATS domain. The YEATS2-containing ATAC complex co-localizes with H3K27 acetylation (H3K27ac) on the promoters of actively transcribed genes. Depletion of YEATS2 or disruption of the interaction between its YEATS domain and acetylated histones reduces the ATAC complex-dependent promoter H3K9ac levels and deactivates the expression of essential genes. Taken together, our study identifies YEATS2 as a histone H3K27ac reader that regulates a transcriptional program essential for NSCLC tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/physiopathology , Histones/metabolism , Lung Neoplasms/physiopathology , Acetylation , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Histones/genetics , Humans , Lung Neoplasms/genetics , Protein Processing, Post-Translational/genetics
4.
Bioorg Med Chem ; 25(16): 4368-4374, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28693916

ABSTRACT

Bacteroides fragilis, a human pathogen, helps in the formation of intra-abdominal abscesses and is involved in 90% of anaerobic peritoneal infections. Phosphonopyruvate decarboxylase (PnPDC), a thiamin diphosphate (ThDP)-dependent enzyme, plays a key role in the formation of 2-aminoethylphosphonate, a component of the cell wall of B. fragilis. As such PnPDC is a possible target for therapeutic intervention in this, and other phosphonate producing organisms. However, the enzyme is of more general interest as it appears to be an evolutionary forerunner to the decarboxylase family of ThDP-dependent enzymes. To date, PnPDC has proved difficult to crystallize and no X-ray structures are available. In the past we have shown that ThDP-dependent enzymes will often crystallize if the cofactor has been irreversibly inactivated. To explore this possibility, and the utility of inhibitors of phosphonate biosynthesis as potential antibiotics, we synthesized phosphonodifluoropyruvate (PnDFP) as a prospective mechanism-based inhibitor of PnPDC. Here we provide evidence that PnDFP indeed inactivates the enzyme, that the inactivation is irreversible, and is accompanied by release of fluoride ion, i.e., PnDFP bears all the hallmarks of a mechanism-based inhibitor. Unfortunately, the enzyme remains refractive to crystallization.


Subject(s)
Bacteroides fragilis/enzymology , Carboxy-Lyases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyruvates/pharmacology , Carboxy-Lyases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Pyruvates/chemical synthesis , Pyruvates/chemistry , Structure-Activity Relationship
5.
Structure ; 25(4): 650-654.e2, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28286003

ABSTRACT

The monocytic leukemia zinc-finger protein-related factor (MORF) is a transcriptional coactivator and a catalytic subunit of the lysine acetyltransferase complex implicated in cancer and developmental diseases. We have previously shown that the double plant homeodomain finger (DPF) of MORF is capable of binding to acetylated histone H3. Here we demonstrate that the DPF of MORF recognizes many newly identified acylation marks. The mass spectrometry study provides comprehensive analysis of H3K14 acylation states in vitro and in vivo. The crystal structure of the MORF DPF-H3K14butyryl complex offers insight into the selectivity of this reader toward lipophilic acyllysine substrates. Together, our findings support the mechanism by which the acetyltransferase MORF promotes spreading of histone acylation.


Subject(s)
Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histones/chemistry , Histones/metabolism , Acetylation , Binding Sites , Crystallography, X-Ray , HeLa Cells , Humans , Lysine/chemistry , Mass Spectrometry , Protein Binding , Protein Domains , Protein Processing, Post-Translational
6.
Proc Natl Acad Sci U S A ; 114(7): E1072-E1080, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137841

ABSTRACT

MYC is a major cancer driver but is documented to be a difficult therapeutic target itself. Here, we report on the biological activity, the structural basis, and therapeutic effects of the family of multitargeted compounds that simultaneously disrupt functions of two critical MYC-mediating factors through inhibiting the acetyllysine binding of BRD4 and the kinase activity of PI3K. We show that the dual-action inhibitor impairs PI3K/BRD4 signaling in vitro and in vivo and affords maximal MYC down-regulation. The concomitant inhibition of PI3K and BRD4 blocks MYC expression and activation, promotes MYC degradation, and markedly inhibits cancer cell growth and metastasis. Collectively, our findings suggest that the dual-activity inhibitor represents a highly promising lead compound for the development of novel anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Morpholines/pharmacology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Pyrans/pharmacology , Thiophenes/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/secondary , Cell Cycle Proteins , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Models, Molecular , Morpholines/therapeutic use , Neoplasm Metastasis/drug therapy , Neoplasm Proteins/physiology , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/pathology , Neuroblastoma/secondary , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Conformation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-myc/physiology , Pyrans/therapeutic use , Signal Transduction/drug effects , Thiophenes/therapeutic use , Transcription Factors/chemistry , Transcription Factors/physiology , Xenograft Model Antitumor Assays
7.
J Mol Biol ; 429(13): 2066-2074, 2017 06 30.
Article in English | MEDLINE | ID: mdl-27697561

ABSTRACT

The plant homeodomain (PHD) finger of Set3 binds methylated lysine 4 of histone H3 in vitro and in vivo; however, precise selectivity of this domain has not been fully characterized. Here, we explore the determinants of methyllysine recognition by the PHD fingers of Set3 and its orthologs. We use X-ray crystallographic and spectroscopic approaches to show that the Set3 PHD finger binds di- and trimethylated states of H3K4 with comparable affinities and employs similar molecular mechanisms to form complexes with either mark. Composition of the methyllysine-binding pocket plays an essential role in determining the selectivity of the PHD fingers. The finding that the histone-binding activity is not conserved in the PHD finger of Set4 suggests different functions for the Set3 and Set4 paralogs.


Subject(s)
Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Histones/chemistry , Histones/metabolism , Crystallography, X-Ray , Lysine/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Conformation
8.
Cell Rep ; 16(12): 3195-3207, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653685

ABSTRACT

MORC3 is linked to inflammatory myopathies and cancer; however, the precise role of MORC3 in normal cell physiology and disease remains poorly understood. Here, we present detailed genetic, biochemical, and structural analyses of MORC3. We demonstrate that MORC3 is significantly upregulated in Down syndrome and that genetic abnormalities in MORC3 are associated with cancer. The CW domain of MORC3 binds to the methylated histone H3K4 tail, and this interaction is essential for recruitment of MORC3 to chromatin and accumulation in nuclear bodies. We show that MORC3 possesses intrinsic ATPase activity that requires DNA, but it is negatively regulated by the CW domain, which interacts with the ATPase domain. Natively linked CW impedes binding of the ATPase domain to DNA, resulting in a decrease in the DNA-stimulated enzymatic activity. Collectively, our studies provide a molecular framework detailing MORC3 functions and suggest that its modulation may contribute to human disease.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Histidine Kinase/metabolism , Adenosine Triphosphatases/chemistry , Cells, Cultured , Chromatin/metabolism , DNA-Binding Proteins/chemistry , Down Syndrome/genetics , Down Syndrome/metabolism , Histidine Kinase/chemistry , Humans , Neoplasms/genetics , Neoplasms/metabolism , Protein Conformation , Protein Domains
9.
Nat Chem Biol ; 12(9): 662-8, 2016 Aug 18.
Article in English | MEDLINE | ID: mdl-27538025

ABSTRACT

The field of chromatin biology has been advancing at an accelerated pace. Recent discoveries of previously uncharacterized sites and types of post-translational modifications (PTMs) and the identification of new sets of proteins responsible for the deposition, removal, and reading of these marks continue raising the complexity of an already exceedingly complicated biological phenomenon. In this Perspective article we examine the biological importance of new types and sites of histone PTMs and summarize the molecular mechanisms of chromatin engagement by newly discovered epigenetic readers. We also highlight the imperative role of structural insights in understanding PTM-reader interactions and discuss future directions to enhance the knowledge of PTM readout.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Epigenesis, Genetic , Histones/chemistry , Histones/metabolism , Protein Processing, Post-Translational , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/genetics , Humans , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Models, Molecular , Molecular Structure , Phosphoproteins/chemistry , Phosphoproteins/metabolism
10.
Nat Chem Biol ; 12(6): 396-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27089029

ABSTRACT

The discovery of new histone modifications is unfolding at startling rates; however, the identification of effectors capable of interpreting these modifications has lagged behind. Here we report the YEATS domain as an effective reader of histone lysine crotonylation, an epigenetic signature associated with active transcription. We show that the Taf14 YEATS domain engages crotonyllysine via a unique π-π-π-stacking mechanism and that other YEATS domains have crotonyllysine-binding activity.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Histones/chemistry , Lysine/chemistry , Models, Molecular , Molecular Structure , Protein Domains
11.
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
12.
Transcription ; 7(1): 14-20, 2016.
Article in English | MEDLINE | ID: mdl-26934307

ABSTRACT

The YEATS domains of AF9 and Taf14 have recently been found to recognize the histone H3K9ac modification. In this commentary, we discuss the mechanistic and biological implications of this interaction. We compare structures of the YEATS-H3K9ac complexes the highlighting a novel mechanism for the acetyllysine recognition through the aromatic cage. We also summarize the latest findings underscoring a critical role of the acetyllysine binding function of AF9 and Taf14 in transcriptional regulation and DNA repair.


Subject(s)
Lysine/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Acetylation , Binding Sites , Gene Expression Regulation , Histone-Lysine N-Methyltransferase , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Nuclear Proteins/genetics , Protein Conformation , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic
13.
ACS Chem Biol ; 11(3): 547-53, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26726824

ABSTRACT

Methyllysine post-translational modifications (PTMs) of histones create binding sites for evolutionarily conserved reader domains that link nuclear host proteins and chromatin-modifying complexes to specific genomic regions. In the context of these events, adjacent histone PTMs are capable of altering the binding activity of readers toward their target marks. This provides a mechanism of "combinatorial readout" of PTMs that can enhance, decrease, or eliminate the association of readers with chromatin. In this Perspective, we focus on recent studies describing the impact of dynamic phospho-serine/threonine/tyrosine marks on the interaction of methyllysine readers with histones, summarize mechanistic aspects of the phospho/methyl readout, and highlight the significance of crosstalk between these PTMs. We also demonstrate that in addition to inhibiting binding and serving as a true switch, promoting dissociation of the methyllysine readers from chromatin, the phospho/methyl combination can act together in a cooperative manner--thus adding a new layer of regulatory information that can be encoded in these dual histone PTMs.


Subject(s)
Epigenesis, Genetic , Histones/metabolism , Lysine/metabolism , Binding Sites , Chromatin , Histones/genetics , Humans , Methylation , Models, Molecular , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Protein Processing, Post-Translational
14.
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
15.
Genes Dev ; 29(17): 1795-800, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26341557

ABSTRACT

The YEATS domain, found in a number of chromatin-associated proteins, has recently been shown to have the capacity to bind histone lysine acetylation. Here, we show that the YEATS domain of Taf14, a member of key transcriptional and chromatin-modifying complexes in yeast, is a selective reader of histone H3 Lys9 acetylation (H3K9ac). Structural analysis reveals that acetylated Lys9 is sandwiched in an aromatic cage formed by F62 and W81. Disruption of this binding in cells impairs gene transcription and the DNA damage response. Our findings establish a highly conserved acetyllysine reader function for the YEATS domain protein family and highlight the significance of this interaction for Taf14.


Subject(s)
DNA Repair/genetics , Gene Expression Regulation, Fungal/genetics , Histones/metabolism , Models, Molecular , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factor TFIID/metabolism , Acetylation , DNA Damage , Histones/chemistry , Histones/genetics , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism
16.
Biochim Biophys Acta ; 1854(8): 1001-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25936776

ABSTRACT

Benzoylformate decarboxylase (BFDC) is a thiamin diphosphate (ThDP)-dependent enzyme that catalyzes the nonoxidative decarboxylation of benzoylformate. It is the penultimate enzyme in both the mandelate pathway and the d-phenylglycine degradation pathway. The ThDP-dependent Enzyme Engineering Database (TEED) now lists more than 800 sequences annotated as BFDCs, including one from Mycobacterium smegmatis (MsBFDC). However, there is no evidence that either pathway for benzoylformate formation exists in the M. smegmatis genome. Further, sequence alignments of MsBFDC with the well characterized enzyme isolated from Pseudomonas putida (PpBFDC) indicate that there will be active site substitutions in MsBFDC likely to reduce activity with benzoylformate. Taken together these data would suggest that the annotation is unlikely to be correct. To test this hypothesis the putative MsBFDC was cloned, expressed, purified, and the X-ray structure was solved to a resolution of 2.2Å. While showing no evidence for ThDP in the active site, the structure was very similar to that of PpBFDC. A number of 2-oxo acids were tested as substrates. For MsBFDC the K(m) value for benzoylformate was ~23 mM, nearly 100-fold greater than that of PpBFDC while the k(cat) value was reduced 60-fold. These values would suggest that benzoylformate is not the physiological substrate for this enzyme, and that annotation as a 2-oxo acid decarboxylase may be more appropriate.


Subject(s)
Bacterial Proteins/chemistry , Carboxy-Lyases/chemistry , Glyoxylates/chemistry , Mandelic Acids/chemistry , Mycobacterium smegmatis/enzymology , Thiamine Pyrophosphate/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Catalytic Domain , Crystallography, X-Ray , Glyoxylates/metabolism , Kinetics , Mandelic Acids/metabolism , Mycobacterium smegmatis/genetics , Thiamine Pyrophosphate/metabolism
17.
Biochemistry ; 53(27): 4358-67, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24956165

ABSTRACT

The X-ray structure of benzoylformate decarboxylase (BFDC) from Pseudomonas putida ATCC 12633 shows it to be a tetramer. This was believed to be typical of all thiamin diphosphate-dependent decarboxylases until recently when the structure of KdcA, a branched-chain 2-keto acid decarboxylase from Lactococcus lactis, showed it to be a homodimer. This lent credence to earlier unfolding experiments on pyruvate decarboxylase from Saccharomyces cerevisiae that indicated that it might be active as a dimer. To investigate this possibility in BFDC, we sought to shift the equilibrium toward dimer formation. Point mutations were made in the noncatalytic monomer-monomer interfaces, but these had a minimal effect on both tetramer formation and catalytic activity. Subsequently, the R141E/Y288A/A306F variant was shown by analytical ultracentrifugation to be partially dimeric. It was also found to be catalytically inactive. Further experiments revealed that just two mutations, R141E and A306F, were sufficient to markedly alter the dimer-tetramer equilibrium and to provide an ~450-fold decrease in kcat. Equilibrium denaturation studies suggested that the residual activity was possibly due to the presence of residual tetramer. The structures of the R141E and A306F variants, determined to <1.5 Å resolution, hinted that disruption of the monomer interfaces will be accompanied by movement of a loop containing Leu109 and Leu110. As these residues contribute to the hydrophobicity of the active site and the correct positioning of the substrate, it seems that tetramer formation may well be critical to the catalytic activity of BFDC.


Subject(s)
Bacterial Proteins/chemistry , Carboxy-Lyases/chemistry , Bacterial Proteins/genetics , Carboxy-Lyases/genetics , Crystallography, X-Ray , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Point Mutation , Protein Denaturation , Protein Multimerization , Protein Structure, Quaternary , Pseudomonas putida/enzymology
18.
Biochemistry ; 53(13): 2145-52, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24628377

ABSTRACT

Identification of enzyme-bound intermediates via their spectroscopic signatures, which then allows direct monitoring of the kinetic fate of these intermediates, poses a continuing challenge. As an electrophilic covalent catalyst, the thiamin diphosphate (ThDP) coenzyme forms a number of noncovalent and covalent intermediates along its reaction pathways, and multiple UV-vis and circular dichroism (CD) bands have been identified at Rutgers pertinent to several among them. These electronic transitions fall into two classes: those for which the conjugated system provides a reasonable guide to the observed λmax and others in which there is no corresponding conjugated system and the observed CD bands are best ascribed to charge transfer (CT) transitions. Herein is reported the reaction of four ThDP enzymes with alternate substrates: (a) acetyl pyruvate, its methyl ester, and fluoropyruvate, these providing the shortest side chains attached at the thiazolium C2 atom and leading to CT bands with λmax values of >390 nm, not pertinent to any on-pathway conjugated systems (estimated λmax values of <330 nm), and (b) (E)-4-(4-chlorophenyl)-2-oxo-3-butenoic acid displaying both a conjugated enamine (430 nm) and a CT transition (480 nm). We suggest that the CT transitions result from an interaction of the π bond on the ThDP C2 side chain as a donor, and the positively charged thiazolium ring as an acceptor, and correspond to covalent ThDP-bound intermediates. Time resolution of these bands allows the rate constants for individual steps to be determined. These CD methods can be applied to the entire ThDP superfamily of enzymes and should find applications with other enzymes.


Subject(s)
Pyruvate Decarboxylase/metabolism , Thiamine Pyrophosphate/metabolism , Thiamine/metabolism , Circular Dichroism , Electron Transport , Molecular Structure , Pyruvate Decarboxylase/chemistry , Thiamine/chemistry , Thiamine Pyrophosphate/chemistry
19.
FEBS J ; 280(24): 6395-411, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23895593

ABSTRACT

For almost 20 years, site-saturation mutagenesis (SSM) has been used to evolve stereoselective enzymes as catalysts for synthetic organic chemistry. Much of this work has focused on enzymes such as lipases and esterases, although the range is rapidly expanding. By contrast, using SSM to study enzyme mechanisms is much less common. Instead, site-directed mutagenesis is more generally employed, with a particular emphasis on alanine variants. In the present review, we provide examples of the growing use of SSM to study not only substrate and reaction selectivity, but also the reaction mechanism of thiamin diphosphate (ThDP)-dependent enzymes. We report that the use of SSM to examine the roles of the catalytic residues of benzoylformate decarboxylase gave rise to results that were at odds with earlier kinetic and structural studies using alanine substitutions and also questioned their conclusions. SSM was also employed to examine the long held tenet that a bulky hydrophobic residue provides a fulcrum by which the V-conformation of the ThDP cofactor is maintained. X-ray structures showed that ThDP stayed in the V-conformation even when the replacement residues were charged or did not contact the cofactor. We also summarize the results obtained when SSM was used to evolve new substrate specificity and/or enantioselectivity in ThDP-dependent enzymes such as benzoylformate decarboxylase, transketolase, 2-succinyl-5-enolpyruvyl-6-hydroxy-3-cyclohexene-1-carboxylate synthase and the E1 component of the 2-oxoglutarate dehydrogenase complex.


Subject(s)
Carboxy-Lyases/metabolism , Mutation/genetics , Thiamine Pyrophosphate/chemistry , Thiamine Pyrophosphate/metabolism , Animals , Carboxy-Lyases/genetics , Catalysis , Humans , Mutagenesis, Site-Directed , Stereoisomerism
20.
Biochemistry ; 52(18): 3028-30, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23607689

ABSTRACT

It is widely accepted that, in thiamin diphosphate (ThDP)-dependent enzymes, much of the rate acceleration is provided by the cofactor. Inter alia, the reactive conformation of ThDP, known as the V-conformation, has been attributed to the presence of a bulky hydrophobic residue located directly below the cofactor. Here we report the use of site-saturation mutagenesis to generate variants of this residue (Leu403) in benzoylformate decarboxylase. The observed 3 orders of magnitude range in k(cat)/K(m) values suggested that conformational changes in the cofactor could be influencing catalysis. However, X-ray structures of several variants were determined, and there was remarkably little change in ThDP conformation. Rather, it seemed that, once the V-conformation was attained, residue size and hydrophobicity were more important for enzyme activity.


Subject(s)
Carboxy-Lyases/metabolism , Thiamine Pyrophosphate/chemistry , Carboxy-Lyases/chemistry , Carboxy-Lyases/genetics , Molecular Conformation , Mutagenesis, Site-Directed , Thiamine Pyrophosphate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...