Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Chembiochem ; 21(1-2): 256-264, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31612581

ABSTRACT

The SMYD2 protein lysine methyltransferase methylates various histone and non-histone proteins and is overexpressed in several cancers. Using peptide arrays, we investigated the substrate specificity of the enzyme, revealing a recognition of leucine (or weaker phenylalanine) at the -1 peptide site and disfavor of acidic residues at the +1 to +3 sites. Using this motif, novel SMYD2 peptide substrates were identified, leading to the discovery of 32 novel peptide substrates with a validated target site. Among them, 19 were previously reported to be methylated at the target lysine in human cells, strongly suggesting that SMYD2 is the protein lysine methyltransferase responsible for this activity. Methylation of some of the novel peptide substrates was tested at the protein level, leading to the identification of 14 novel protein substrates of SMYD2, six of which were more strongly methylated than p53, the best SMYD2 substrate described so far. The novel SMYD2 substrate proteins are involved in diverse biological processes such as chromatin regulation, transcription, and intracellular signaling. The results of our study provide a fundament for future investigations into the role of this important enzyme in normal development and cancer.


Subject(s)
Histone-Lysine N-Methyltransferase/analysis , Circular Dichroism , HEK293 Cells , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Substrate Specificity
2.
Biochemistry ; 58(35): 3634-3645, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31389685

ABSTRACT

SET and MYND domain-containing protein 3 (SMYD3) is a lysine methyltransferase that plays a central role in a variety of cancer diseases, exerting its pro-oncogenic activity by methylation of key proteins, of both nuclear and cytoplasmic nature. However, the role of SMYD3 in the initiation and progression of cancer is not yet fully understood and further biochemical characterization is required to support the discovery of therapeutics targeting this enzyme. We have therefore developed robust protocols for production, handling, and crystallization of SMYD3 and biophysical and biochemical assays for clarification of SMYD3 biochemistry and identification of useful lead compounds. Specifically, a time-resolved biosensor assay was developed for kinetic characterization of SMYD3 interactions. Functional differences in SMYD3 interactions with its natural small molecule ligands SAM and SAH were revealed, with SAM forming a very stable complex. A variety of peptides mimicking putative substrates of SMYD3 were explored in order to expose structural features important for recognition. The interaction between SMYD3 and some peptides was influenced by SAM. A nonradioactive SMYD3 activity assay using liquid chromatography-mass spectrometry (LC-MS) analysis explored substrate features of importance also for methylation. Methylation was notable only toward MAP kinase kinase kinase 2 (MAP3K2_K260)-mimicking peptides, although binary and tertiary complexes were detected also with other peptides. The analysis supported a random bi-bi mechanistic model for SMYD3 methyltransferase catalysis. Our work unveiled complexities in SMYD3 biochemistry and resulted in procedures suitable for further studies and identification of novel starting points for design of effective and specific leads for this potential oncology target.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Circular Dichroism , Crystallography, X-Ray , Enzyme Stability , Epigenesis, Genetic/genetics , Escherichia coli , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Kinetics , Ligands , Protein Conformation , Protein Unfolding , Structure-Activity Relationship , Temperature , Thermodynamics
3.
Nat Commun ; 10(1): 3541, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388018

ABSTRACT

SETD3 is an actin histidine-N3 methyltransferase, whereas other characterized SET-domain enzymes are protein lysine methyltransferases. We report that in a pre-reactive complex SETD3 binds the N3-protonated form (N3-H) of actin His73, and in a post-reactive product complex, SETD3 generates the methylated histidine in an N1-protonated (N1-H) and N3-methylated form. During the reaction, the imidazole ring of His73 rotates ~105°, which shifts the proton from N3 to N1, thus ensuring that the target atom N3 is deprotonated prior to the methyl transfer. Under the conditions optimized for lysine deprotonation, SETD3 has weak lysine methylation activity on an actin peptide in which the target His73 is substituted by a lysine. The structure of SETD3 with Lys73-containing peptide reveals a bent conformation of Lys73, with its side chain aliphatic carbons tracing along the edge of imidazole ring and the terminal ε-amino group occupying a position nearly identical to the N3 atom of unmethylated histidine.


Subject(s)
Actins/chemistry , Catalytic Domain , Histone-Lysine N-Methyltransferase/chemistry , Actins/metabolism , Crystallography, X-Ray , Histidine/chemistry , Histidine/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Lysine/chemistry , Lysine/metabolism , Methylation , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity
4.
J Med Chem ; 62(17): 7669-7683, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31415173

ABSTRACT

The first chemical probe to primarily occupy the co-factor binding site of a Su(var)3-9, enhancer of a zeste, trithorax (SET) domain containing protein lysine methyltransferase (PKMT) is reported. Protein methyltransferases require S-adenosylmethionine (SAM) as a co-factor (methyl donor) for enzymatic activity. However, SAM itself represents a poor medicinal chemistry starting point for a selective, cell-active inhibitor given its extreme physicochemical properties and its role in multiple cellular processes. A previously untested medicinal chemistry strategy of deliberate file enrichment around molecules bearing the hallmarks of SAM, but with improved lead-like properties from the outset, yielded viable hits against SET and MYND domain-containing protein 2 (SMYD2) that were shown to bind in the co-factor site. These leads were optimized to identify a highly biochemically potent, PKMT-selective, and cell-active chemical probe. While substrate-based inhibitors of PKMTs are known, this represents a novel, co-factor-derived strategy for the inhibition of SMYD2 which may also prove applicable to lysine methyltransferase family members previously thought of as intractable.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , S-Adenosylmethionine/pharmacology , Small Molecule Libraries/pharmacology , Binding Sites/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Humans , Models, Molecular , Molecular Structure , S-Adenosylmethionine/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
5.
Bioorg Med Chem ; 27(17): 3866-3878, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31327677

ABSTRACT

SET domain bifurcated protein 1 (SETDB1) is a human histone-lysine methyltransferase which is amplified in human cancers and was shown to be crucial in the growth of non-small and small cell lung carcinoma. In addition to its catalytic domain, SETDB1 harbors a unique tandem tudor domain which recognizes histone sequences containing both methylated and acetylated lysines, and likely contributes to its localization on chromatin. Using X-ray crystallography and NMR spectroscopy fragment screening approaches, we have identified the first small molecule fragment hits that bind to histone peptide binding groove of the Tandem Tudor Domain (TTD) of SETDB1. Herein, we describe the binding modes of these fragments and analogues and the biophysical characterization of key compounds. These confirmed small molecule fragments will inform the development of potent antagonists of SETDB1 interaction with histones.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Histones/antagonists & inhibitors , Histones/metabolism , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tudor Domain/drug effects
6.
Nat Commun ; 9(1): 2583, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968706

ABSTRACT

Embryonic stem cells (ESCs) maintain pluripotency through unique epigenetic states. When ESCs commit to a specific lineage, epigenetic changes in histones and DNA accompany the transition to specialized cell types. Investigating how epigenetic regulation controls lineage specification is critical in order to generate the required cell types for clinical applications. Uhrf1 is a widely known hemi-methylated DNA-binding protein, playing a role in DNA methylation through the recruitment of Dnmt1 and in heterochromatin formation alongside G9a, Trim28, and HDACs. Although Uhrf1 is not essential in ESC self-renewal, it remains elusive how Uhrf1 regulates cell specification. Here we report that Uhrf1 forms a complex with the active trithorax group, the Setd1a/COMPASS complex, to maintain bivalent histone marks, particularly those associated with neuroectoderm and mesoderm specification. Overall, our data demonstrate that Uhrf1 safeguards proper differentiation via bivalent histone modifications.


Subject(s)
Cellular Reprogramming/genetics , Histone Code/genetics , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , Cellular Reprogramming Techniques , Chimera , DNA Methylation/physiology , Epigenesis, Genetic , Female , Fibroblasts , Gene Knockout Techniques , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/isolation & purification , Histones/metabolism , Humans , Male , Mesoderm/cytology , Mesoderm/physiology , Mice , Mouse Embryonic Stem Cells , Neural Plate/cytology , Neural Plate/physiology , Nuclear Proteins/genetics , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases
7.
Bioorg Med Chem ; 25(16): 4414-4423, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28662962

ABSTRACT

Given the high homology between the protein lysine methyltransferases G9a-like protein (GLP) and G9a, it has been challenging to develop potent and selective inhibitors for either enzyme. Recently, we reported two quinazoline compounds, MS0124 and MS012, as GLP selective inhibitors. To further investigate the structure-activity relationships (SAR) of the quinazoline scaffold, we designed and synthesized a range of analogs bearing different 2-amino substitutions and evaluated their inhibition potencies against both GLP and G9a. These studies led to the identification of two new GLP selective inhibitors, 13 (MS3748) and 17 (MS3745), with 59- and 65-fold higher potency for GLP over G9a, which were confirmed by isothermal titration calorimetry (ITC). Crystal structures of GLP and G9a in complex with 13 and 17 provide insight into the interactions of the inhibitors with both proteins. In addition, we generated GLP selective inhibitors bearing a quinoline core instead of the quinazoline core.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Quinazolines/pharmacology , Quinolines/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Humans , Models, Molecular , Molecular Structure , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(2): 246-51, 2017 Feb.
Article in Chinese | MEDLINE | ID: mdl-29763214

ABSTRACT

Objective: To purify the recombinant Setd8 protein and prepare rabbit anti-mouse Setd8 polyclonal antibody. Methods: The recombinant plasmid p ET-30a-Setd8 was constructed by double enzyme digestion and linkage,and then transformed into E. coli BL21. The expression of the target protein was induced by IPTG and the expression product was purified by Ni-NTA affinity chromatograph. The purified protein was used to immunize New Zealand white rabbits to produce polyclonal antibody. The titer and specificity of the antibody were identified by ELISA,Western blotting and immunohistochemistry. Results: The prokaryotic expression vector p ET-30a-Setd8 was constructed successfully. After induced by IPTG,the recombinant Setd8 protein was expressed effectively in E. coli BL21. Polyclonal antibody against Setd8 was generated by immunizing rabbits with the routine method. ELISA showed that the titer of rabbit anti-Setd8 antiserum was 1 ∶ 1 000 000.Western blotting demonstrated that the polyclonal antibody could recognize the native mouse Setd8 protein. Immunohistochemistry revealed that Setd8 protein recognized by the polyclonal antibody was mainly distributed in the nucleus of spermatogonia in adult mouse testis. Conclusion: Using the prokaryotic expression vector p ET-30a-Setd8,we have prepared successfully the polyclonal antibody with high affinity and specificity.


Subject(s)
Antibodies/immunology , Antibody Affinity , Antibody Specificity , Histone-Lysine N-Methyltransferase/immunology , Animals , Antibodies/isolation & purification , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Immunohistochemistry , Mice , Plasmids , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
Methods Enzymol ; 573: 209-40, 2016.
Article in English | MEDLINE | ID: mdl-27372755

ABSTRACT

In eukaryotes, several lysine residues on histone proteins are methylated. This posttranslational modification is linked to a myriad of nuclear-based transactions such as epigenetic inheritance of heterochromatin, regulation of gene expression, DNA damage repair, and DNA replication. The majority of the enzymes responsible for writing these marks onto chromatin belong to the SET domain family of histone lysine methyltransferases. Although they often share important structural features, including a conserved catalytic domain, SET domain enzymes use different mechanisms to achieve substrate recognition, mono-, di-, or trimethylate lysine residues and some require other proteins to achieve maximal methyltransferase activity. In this chapter, we summarize our efforts to purify, crystallize, and enzymatically characterize SET domain enzymes with a specific focus on the histone H3K27 monomethyltransferase ATXR5.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray/methods , Enzyme Assays/methods , Escherichia coli/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Models, Molecular , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ricinus/chemistry , Ricinus/genetics , Ricinus/metabolism , Sequence Alignment , Up-Regulation
10.
Methods Mol Biol ; 1366: 207-217, 2016.
Article in English | MEDLINE | ID: mdl-26585137

ABSTRACT

Methylation of estrogen receptor α by the histone lysine methyltransferase SMYD2 regulates ERα chromatin recruitment and its target gene expression. This protocol describes SMYD2 purification and crystallization of SMYD2 in complex with an ERα peptide. Recombinant SMYD2 is overexpressed in Escherichia coli cells. After release from the cells by French Press, SMYD2 is purified to apparent homogeneity with multiple chromatography methods. Nickel affinity column purifies SMYD2 based on specific interaction of its 6×His tag with the bead-immobilized nickel ions. Desalting column is used for protein buffer exchange. Gel filtration column purifies SMYD2 based on molecular size. The entire purification process is monitored and analyzed by SDS-polyacrylamide gel electrophoresis. Crystallization of SMYD2 is performed with the hanging drop vapor diffusion method. Crystals of the SMYD2-ERα peptide complex are obtained by microseeding using seeding bead. This method can give rise to large size of crystals which are suitable for X-ray diffraction data collection. X-ray crystallographic study of the SMYD2-ERα complex can provide structural insight into posttranslational regulation of ERα signaling.


Subject(s)
Chromatography, Affinity , Chromatography, Gel , Crystallography, X-Ray , Estrogen Receptor alpha/isolation & purification , Histone-Lysine N-Methyltransferase/isolation & purification , Crystallization , Electrophoresis, Polyacrylamide Gel , Escherichia coli/enzymology , Escherichia coli/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Weight , Protein Binding , Recombinant Proteins/isolation & purification , Workflow
11.
Protein Expr Purif ; 97: 44-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24583182

ABSTRACT

Chromatin-associated proteins are heterogeneously and dynamically composed. To gain a complete understanding of DNA packaging and basic nuclear functions, it is important to generate a comprehensive inventory of these proteins. However, biochemical purification of chromatin-associated proteins is difficult and is accompanied by concerns over complex stability, protein solubility and yield. Here, we describe a new method for optimized purification of the endogenously expressed fission yeast Set2 complex, histone H3K36 methyltransferase. Using the standard centrifugation procedure for purification, approximately half of the Set2 protein separated into the insoluble chromatin pellet fraction, making it impossible to recover the large amounts of soluble Set2. To overcome this poor recovery, we developed a novel protein purification technique termed the filtration/immunoaffinity purification/mass spectrometry (FIM) method, which eliminates the need for centrifugation. Using the FIM method, in which whole cell lysates were filtered consecutively through eight different pore sizes (53-0.8µm), a high yield of soluble FLAG-tagged Set2 was obtained from fission yeast. The technique was suitable for affinity purification and produced a low background. A mass spectrometry analysis of anti-FLAG immunoprecipitated proteins revealed that Rpb1, Rpb2 and Rpb3, which have all been reported previously as components of the budding yeast Set2 complex, were isolated from fission yeast using the FIM method. In addition, other subunits of RNA polymerase II and its phosphatase were also identified. In conclusion, the FIM method is valid for the efficient purification of protein complexes that separate into the insoluble chromatin pellet fraction during centrifugation.


Subject(s)
Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Schizosaccharomyces pombe Proteins/isolation & purification , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Chromatography, Liquid/methods , Filtration/methods , Immunoblotting/methods , Immunoprecipitation/methods , Mass Spectrometry/methods , Schizosaccharomyces/chemistry
12.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 595-602, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23519668

ABSTRACT

SET7/9 is a protein lysine methyltransferase that methylates histone H3 and nonhistone proteins such as p53, TAF10 and oestrogen receptor α. In previous work, novel inhibitors of SET7/9 that are amine analogues of the coenzyme S-(5'-adenosyl)-L-methionine (AdoMet) have been developed. Here, crystal structures of SET7/9 are reported in complexes with two AdoMet analogues, designated DAAM-3 and AAM-1, in which an n-hexylaminoethyl group or an n-hexyl group is attached to the N atom that replaces the S atom of AdoMet, respectively. In both structures, the inhibitors bind to the coenzyme-binding site and their additional alkyl chain binds in the lysine-access channel. The N atom in the azaalkyl chain of DAAM-3 is located at almost the same position as the N-methyl C atom of the methylated lysine side chain in the substrate-peptide complex structures and stabilizes complex formation by hydrogen bonding to the substrate-binding site residues of SET7/9. On the other hand, the alkyl chain of AAM-1, which is a weaker inhibitor than DAAM-3, binds in the lysine-access channel only through hydrophobic and van der Waals interactions. Unexpectedly, the substrate-binding site of SET7/9 complexed with AAM-1 specifically interacts with the artificial N-terminal sequence of an adjacent symmetry-related molecule, presumably stabilizing the alkyl chain of AAM-1.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Macromolecular Substances/chemistry , S-Adenosylmethionine/chemistry , Crystallography, X-Ray , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Macromolecular Substances/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , S-Adenosylmethionine/metabolism
13.
Mol Cell Biol ; 33(10): 2067-77, 2013 May.
Article in English | MEDLINE | ID: mdl-23508102

ABSTRACT

Methylation of lysine 4 on histone H3 (H3K4) at promoters is tightly linked to transcriptional regulation in human cells. At least six different COMPASS-like multisubunit (SET1/MLL) complexes that contain methyltransferase activity for H3K4 have been described, but a comprehensive and quantitative analysis of these SET1/MLL complexes is lacking. We applied label-free quantitative mass spectrometry to determine the subunit composition and stoichiometry of the human SET1/MLL complexes. We identified both known and novel, unique and shared interactors and determined their distribution and stoichiometry over the different SET1/MLL complexes. In addition to being a core COMPASS subunit, the Dpy30 protein is a genuine subunit of the NURF chromatin remodeling complex. Furthermore, we identified the Bod1 protein as a discriminator between the SET1B and SET1A complexes, and we show that the H3K36me-interactor Psip1 preferentially binds to the MLL2 complex. Finally, absolute protein quantification in crude lysates mirrors many of the observed SET1/MLL complex stoichiometries. Our findings provide a molecular framework for understanding the diversity and abundance of the different SET1/MLL complexes, which together establish the H3K4 methylation landscape in human cells.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplasm Proteins/metabolism , Protein Subunits/metabolism , Adaptor Proteins, Signal Transducing/isolation & purification , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Chromatography, Affinity , DNA-Binding Proteins/isolation & purification , HeLa Cells , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Intracellular Signaling Peptides and Proteins , Myeloid-Lymphoid Leukemia Protein/isolation & purification , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/isolation & purification , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Protein Interaction Mapping , Protein Subunits/isolation & purification , Proto-Oncogene Proteins/isolation & purification , Proto-Oncogene Proteins/metabolism , Transcription Factors/isolation & purification , Transcription Factors/metabolism
14.
Clin Chim Acta ; 415: 181-90, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23123287

ABSTRACT

BACKGROUND: No substantial work has been conducted to date in context to cationic proteins with antilithiatic activity. We explored the antilithiatic cationic proteins present in human calcium oxalate (CaOx) stones and also examined their molecular interactions with calcium oxalate crystals in silico. METHODS: Proteins were isolated from the matrix of human CaOx containing kidney stones. Proteins having MW>3 kDa were subjected to cation exchange chromatography followed by molecular-sieve chromatography. The effect of these purified cationic proteins was tested against CaOx nucleation and growth and on oxalate injured MDCK cells for their activity. Proteins were identified by MALDI-TOF MS. Molecular interaction studies with COM crystals in silico were also investigated. RESULTS: Three antilithiatic cationic proteins were identified as histone-lysine N-methyltransferase, inward rectifier K channel and protein Wnt-2 (MW~53, ~44, and ~42 kDa respectively) by MALDI-TOF MS based on database search with MASCOT server. Further molecular modeling calculations revealed the mode of interaction of these proteins with CaOx at the molecular level. CONCLUSION: We identified histone-lysine N-methyltransferase, inward rectifier K channel and protein Wnt-2 as novel antilithiatic proteins which play a vital role in the kidney function and have been associated with various kidney diseases.


Subject(s)
Calcium Oxalate/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Kidney Calculi/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Wnt2 Protein/chemistry , Adult , Animals , Binding Sites , Cations , Cell Survival/drug effects , Chromatography, Gel , Chromatography, Ion Exchange , Crystallization , Cytoprotection , Dogs , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/pharmacology , Humans , Madin Darby Canine Kidney Cells , Molecular Docking Simulation , Potassium Channels, Inwardly Rectifying/isolation & purification , Potassium Channels, Inwardly Rectifying/pharmacology , Protein Binding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Wnt2 Protein/isolation & purification , Wnt2 Protein/pharmacology
15.
J Am Chem Soc ; 135(3): 1048-56, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23244065

ABSTRACT

Protein methyltransferases (PMTs) have emerged as important epigenetic regulators in myriad biological processes in both normal physiology and disease conditions. However, elucidating PMT-regulated epigenetic processes has been hampered by ambiguous knowledge about in vivo activities of individual PMTs particularly because of their overlapping but nonredundant functions. To address limitations of conventional approaches in mapping chromatin modification of specific PMTs, we have engineered the chromatin-modifying apparatus and formulated a novel technology, termed clickable chromatin enrichment with parallel DNA sequencing (CliEn-seq), to probe genome-wide chromatin modification within living cells. The three-step approach of CliEn-seq involves in vivo synthesis of S-adenosyl-L-methionine (SAM) analogues from cell-permeable methionine analogues by engineered SAM synthetase (methionine adenosyltransferase or MAT), in situ chromatin modification by engineered PMTs, subsequent enrichment and sequencing of the uniquely modified chromatins. Given critical roles of the chromatin-modifying enzymes in epigenetics and structural similarity among many PMTs, we envision that the CliEn-seq technology is generally applicable in deciphering chromatin methylation events of individual PMTs in diverse biological settings.


Subject(s)
Chromatin/metabolism , DNA Methylation , Genetic Engineering , Glucagon-Like Peptide 1/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Protein Processing, Post-Translational , Chromatin/genetics , Epigenomics , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/isolation & purification , HEK293 Cells , Histocompatibility Antigens/genetics , Histocompatibility Antigens/isolation & purification , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Models, Molecular , S-Adenosylmethionine/biosynthesis , S-Adenosylmethionine/chemistry
16.
J Bacteriol ; 194(23): 6410-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23002218

ABSTRACT

Rickettsia prowazekii, the etiologic agent of epidemic typhus, is a potential biological threat agent. Its outer membrane protein B (OmpB) is an immunodominant antigen and plays roles as protective envelope and as adhesins. The observation of the correlation between methylation of lysine residues in rickettsial OmpB and bacterial virulence has suggested the importance of an enzymatic system for the methylation of OmpB. However, no rickettsial lysine methyltransferase has been characterized. Bioinformatic analysis of genomic DNA sequences of Rickettsia identified putative lysine methyltransferases. The genes of the potential methyltransferases were synthesized, cloned, and expressed in Escherichia coli, and expressed proteins were purified by nickel-nitrilotriacetic acid (Ni-NTA) affinity chromatography. The methyltransferase activities of the purified proteins were analyzed by methyl incorporation of radioactively labeled S-adenosylmethionine into recombinant fragments of OmpB. Two putative recombinant methyltransferases (rRP789 and rRP027-028) methylated recombinant OmpB fragments. The specific activity of rRP789 is 10- to 30-fold higher than that of rRP027-028. Western blot analysis using specific antibodies against trimethyl lysine showed that both rRP789 and rRP027-028 catalyzed trimethylation of recombinant OmpB fragments. Liquid chromatography-tandem mass spectrometry (LC/MS-MS) analysis showed that rRP789 catalyzed mono-, di-, and trimethylation of lysine, while rRP027-028 catalyzed exclusively trimethylation. To our knowledge, rRP789 and rRP027-028 are the first biochemically characterized lysine methyltransferases of outer membrane proteins from Gram-negative bacteria. The production and characterization of rickettsial lysine methyltransferases provide new tools to investigate the mechanism of methylation of OmpB, effects of methylation on the structure and function of OmpB, and development of methylated OmpB-based diagnostic assays and vaccine candidates.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Rickettsia prowazekii/enzymology , Rickettsia prowazekii/metabolism , Blotting, Western , Chromatography, Affinity , Chromatography, Liquid , Cloning, Molecular , Computational Biology , Escherichia coli/genetics , Gene Expression , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/isolation & purification , Isotope Labeling , Kinetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Rickettsia prowazekii/genetics , S-Adenosylmethionine/metabolism , Tandem Mass Spectrometry
17.
Annu Rev Biochem ; 81: 65-95, 2012.
Article in English | MEDLINE | ID: mdl-22663077

ABSTRACT

The Saccharomyces cerevisiae Set1/COMPASS was the first histone H3 lysine 4 (H3K4) methylase identified over 10 years ago. Since then, it has been demonstrated that Set1/COMPASS and its enzymatic product, H3K4 methylation, is highly conserved across the evolutionary tree. Although there is only one COMPASS in yeast, Drosophila possesses three and humans bear six COMPASS family members, each capable of methylating H3K4 with nonredundant functions. In yeast, the histone H2B monoubiquitinase Rad6/Bre1 is required for proper H3K4 and H3K79 trimethylations. The machineries involved in this process are also highly conserved from yeast to human. In this review, the process of histone H2B monoubiquitination-dependent and -independent histone H3K4 methylation as a mark of active transcription, enhancer signatures, and developmentally poised genes is discussed. The misregulation of histone H2B monoubiquitination and H3K4 methylation result in the pathogenesis of human diseases, including cancer. Recent findings in this regard are also examined.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , DNA Repair , Gene Expression Regulation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/isolation & purification , Humans , Neoplasms/metabolism , Transcription, Genetic
18.
Methods Mol Biol ; 809: 465-72, 2012.
Article in English | MEDLINE | ID: mdl-22113294

ABSTRACT

Methylation on histone H3 lysine 4 (H3K4) correlates with actively transcribed genes. In mammalian cells, there exist multiple Set1-like histone H3K4 methyltransferase complexes, which have overlapping but distinct subunit compositions. Developing methods to isolate each of these histone H3K4 methyltransferase complexes would help understand the molecular mechanisms by which histone H3K4 methylation regulates mammalian gene expression. In this chapter, we provide a one-step affinity purification protocol on isolation of the MLL3/MLL4 histone H3K4 methyltransferase complex using FLAG-tagged PA1, a unique subunit of the MLL3/MLL4 complex.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Histone Methyltransferases , Humans , Immunoprecipitation
19.
Nucleic Acids Res ; 38(19): 6389-403, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20530532

ABSTRACT

The dynamic exchange of histone lysine methylation status by histone methyltransferases and demethylases has been previously implicated as an important factor in chromatin structure and transcriptional regulation. Using immunoaffinity TAP analysis, we purified the WHISTLE-interacting protein complexes, which include the heat shock protein HSP90α and the jumonji C-domain harboring the histone demethylase JMJD1C. In this study, we demonstrate that JMJD1C specifically demethylates histone H3K9 mono- and di-methylation, and mediates transcriptional activation. We also provide evidence suggesting that both WHISTLE and JMJD1C performs functions in the development of mouse testes by regulating the expression of the steroidogenesis marker, p450c17, via SF-1-mediated transcription. Furthermore, we demonstrate that WHISTLE is recruited to the p450c17 promoter via SF-1 and represses the transcription of prepubertal stages of steroidogenesis, after which JMJD1C replaces WHISTLE and activates the expression of target genes via SF-1-mediated interactions. Our results demonstrate that the histone methylation balance mediated by HMTase WHISTLE and demethylase JMJD1C perform a transcriptional regulatory function in mouse testis development.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Steroids/biosynthesis , Animals , Cell Line , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/isolation & purification , Jumonji Domain-Containing Histone Demethylases/physiology , Male , Methylation , Mice , Steroid 17-alpha-Hydroxylase/genetics , Testis/growth & development , Testis/metabolism
20.
Nucleic Acids Res ; 38(17): 5681-91, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20457750

ABSTRACT

Metnase is a human protein with methylase (SET) and nuclease domains that is widely expressed, especially in proliferating tissues. Metnase promotes non-homologous end-joining (NHEJ), and knockdown causes mild hypersensitivity to ionizing radiation. Metnase also promotes plasmid and viral DNA integration, and topoisomerase IIα (TopoIIα)-dependent chromosome decatenation. NHEJ factors have been implicated in the replication stress response, and TopoIIα has been proposed to relax positive supercoils in front of replication forks. Here we show that Metnase promotes cell proliferation, but it does not alter cell cycle distributions, or replication fork progression. However, Metnase knockdown sensitizes cells to replication stress and confers a marked defect in restart of stalled replication forks. Metnase promotes resolution of phosphorylated histone H2AX, a marker of DNA double-strand breaks at collapsed forks, and it co-immunoprecipitates with PCNA and RAD9, a member of the PCNA-like RAD9-HUS1-RAD1 intra-S checkpoint complex. Metnase also promotes TopoIIα-mediated relaxation of positively supercoiled DNA. Metnase is not required for RAD51 focus formation after replication stress, but Metnase knockdown cells show increased RAD51 foci in the presence or absence of replication stress. These results establish Metnase as a key factor that promotes restart of stalled replication forks, and implicate Metnase in the repair of collapsed forks.


Subject(s)
DNA Repair , DNA Replication , Histone-Lysine N-Methyltransferase/physiology , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/isolation & purification , Cell Proliferation , Cell Survival , DNA Topoisomerases, Type II/metabolism , DNA, Superhelical/metabolism , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/isolation & purification , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Immunoprecipitation , Proliferating Cell Nuclear Antigen/isolation & purification , Rad51 Recombinase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL