Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
ChemistryOpen ; 7(9): 721-727, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30214852

ABSTRACT

Gefitinib is the molecular target drug for advanced non-small-cell lung cancer. The primary target of gefitinib is the positive mutation of epidermal growth factor receptor, but it also inhibits cyclin G-associated kinase (GAK). To reveal the molecular bases of GAK and gefitinib binding, structure analyses were conducted and determined two forms of the gefitinib-bound nanobody⋅GAK kinase domain complex structures. The first form, GAK_1, has one gefitinib at the ATP binding pocket, whereas the second form, GAK_2, binds one each in the ATP binding site and a novel binding site adjacent to the activation segment C-terminal helix, a unique element of the Numb-associated kinase family. In the novel binding site, gefitinib binds in the hydrophobic groove around the activation segment, disrupting the conserved hydrogen bonds for the catalytic activity. These structures suggest possibilities for the development of selective GAK inhibitors for viral infections, such as the hepatitis C virus.

2.
Epigenetics ; 13(4): 410-431, 2018.
Article in English | MEDLINE | ID: mdl-30080437

ABSTRACT

The bromodomain and extra-terminal domain (BET) proteins are promising drug targets for cancer and immune diseases. However, BET inhibition effects have been studied more in the context of bromodomain-containing protein 4 (BRD4) than BRD2, and the BET protein association to histone H4-hyperacetylated chromatin is not understood at the genome-wide level. Here, we report transcription start site (TSS)-resolution integrative analyses of ChIP-seq and transcriptome profiles in human non-small cell lung cancer (NSCLC) cell line H23. We show that di-acetylation at K5 and K8 of histone H4 (H4K5acK8ac) co-localizes with H3K27ac and BRD2 in the majority of active enhancers and promoters, where BRD2 has a stronger association with H4K5acK8ac than H3K27ac. Although BET inhibition by JQ1 led to complete reduction of BRD2 binding to chromatin, only local changes of H4K5acK8ac levels were observed, suggesting that recruitment of BRD2 does not influence global histone H4 hyperacetylation levels. This finding supports a model in which recruitment of BET proteins via histone H4 hyperacetylation is predominant over hyperacetylation of histone H4 by BET protein-associated acetyltransferases. In addition, we found that a remarkable number of BRD2-bound genes, including MYC and its downstream target genes, were transcriptionally upregulated upon JQ1 treatment. Using BRD2-enriched sites and transcriptional activity analysis, we identified candidate transcription factors potentially involved in the JQ1 response in BRD2-dependent and -independent manner.


Subject(s)
Azepines/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Histones/chemistry , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Triazoles/pharmacology , Acetylation/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Models, Molecular , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors , Transcription Initiation Site/drug effects
3.
Protein Expr Purif ; 150: 92-99, 2018 10.
Article in English | MEDLINE | ID: mdl-29793032

ABSTRACT

Since phosphorylation is involved in various physiological events, kinases and interacting factors can be potential targets for drug discovery. For the development and improvement of inhibitors from the point of view of mechanistic enzymology, a cell-free protein synthesis system would be advantageous, since it could prepare mutant proteins easily. However, especially in the case of protein kinase, product solubility remains one of the major challenges. To overcome this problem, we prepared a chaperone-supplemented extract from Escherichia coli BL21 cells harboring a plasmid encoding a set of chaperone genes, dnaK, dnaJ, and grpE. We explored cell-disruption procedures and constructed an efficient protein synthesis system. Employing this system, we produced the kinase domain of human hematopoietic cell kinase (HCK) to obtain further structural information about its molecular interaction with one of its inhibitors, previously developed by our group (RK-20449). Lower reaction temperature improved the solubility, and addition of a protein phosphatase (YpoH) facilitated the homogeneous production of the non-phosphorylated kinase domain. Crystals of the purified product were obtained and the kinase-inhibitor complex structure was solved at 1.7 Šresolution. In addition, results of kinase activity measurement, using a synthetic substrate, showed that the kinase activity was facilitated by autophosphorylation at Tyr416, as confirmed by the peptide mass mapping.


Subject(s)
Gene Expression , Proto-Oncogene Proteins c-hck , Cell-Free System/chemistry , Cell-Free System/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Humans , Phosphorylation , Protein Domains , Proto-Oncogene Proteins c-hck/biosynthesis , Proto-Oncogene Proteins c-hck/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
4.
PLoS One ; 13(2): e0193158, 2018.
Article in English | MEDLINE | ID: mdl-29462206

ABSTRACT

Growing numbers of therapeutic antibodies offer excellent treatment strategies for many diseases. Elucidation of the interaction between a potential therapeutic antibody and its target protein by structural analysis reveals the mechanism of action and offers useful information for developing rational antibody designs for improved affinity. Here, we developed a rapid, high-yield cell-free system using dialysis mode to synthesize antibody fragments for the structural analysis of antibody-antigen complexes. Optimal synthesis conditions of fragments (Fv and Fab) of the anti-EGFR antibody 059-152 were rapidly determined in a day by using a 30-µl-scale unit. The concentration of supplemented disulfide isomerase, DsbC, was critical to obtaining soluble antibody fragments. The optimal conditions were directly applicable to a 9-ml-scale reaction, with linear scalable yields of more than 1 mg/ml. Analyses of purified 059-152-Fv and Fab showed that the cell-free synthesized antibody fragments were disulfide-bridged, with antigen binding activity comparable to that of clinical antibodies. Examination of the crystal structure of cell-free synthesized 059-152-Fv in complex with the extracellular domain of human EGFR revealed that the epitope of 059-152-Fv broadly covers the EGF binding surface on domain III, including residues that formed critical hydrogen bonds with EGF (Asp355EGFR, Gln384EGFR, H409EGFR, and Lys465EGFR), so that the antibody inhibited EGFR activation. We further demonstrated the application of the cell-free system to site-specific integration of non-natural amino acids for antibody engineering, which would expand the availability of therapeutic antibodies based on structural information and rational design. This cell-free system could be an ideal antibody-fragment production platform for functional and structural analysis of potential therapeutic antibodies and for engineered antibody development.


Subject(s)
Antigen-Antibody Complex , Binding Sites, Antibody , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Variable Region/chemistry , Epitopes , Humans , Models, Molecular , Protein Conformation
5.
J Biochem ; 162(5): 357-369, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28992119

ABSTRACT

Cell-free protein synthesis is a useful method for preparing proteins for functional or structural analyses. However, batch-to-batch variability with regard to protein synthesis activity remains a problem for large-scale production of cell extract in the laboratory. To address this issue, we have developed a novel procedure for large-scale preparation of bacterial cell extract with high protein synthesis activity. The developed procedure comprises cell cultivation using a fermentor, harvesting and washing of cells by tangential flow filtration, cell disruption with high-pressure homogenizer and continuous diafiltration. By optimizing and combining these methods, ∼100 ml of the cell extract was prepared from 150 g of Escherichia coli cells. The protein synthesis activities, defined as the yield of protein per unit of absorbance at 260 nm of the cell extract, were shown to be reproducible, and the average activity of several batches was twice that obtained using a previously reported method. In addition, combinatorial use of the high-pressure homogenizer and diafiltration increased the scalability, indicating that the cell concentration at disruption varies from 0.04 to 1 g/ml. Furthermore, addition of Gam protein and examinations of the N-terminal sequence rendered the extract prepared here useful for rapid screening with linear DNA templates.


Subject(s)
Cell-Free System , DNA-Binding Proteins , Escherichia coli , Green Fluorescent Proteins , Protein Biosynthesis , Viral Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Protein Biosynthesis/physiology , Reproducibility of Results , Viral Proteins/genetics , Viral Proteins/metabolism
6.
Bioconjug Chem ; 27(1): 198-206, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26625213

ABSTRACT

Z-Lysine (ZLys) is a lysine derivative with a benzyloxycarbonyl group linked to the ε-nitrogen. It has been genetically encoded with the UAG stop codon, using the pair of an engineered variant of pyrrolysyl-tRNA synthetase (PylRS) and tRNA(Pyl). In the present study, we designed a novel Z-lysine derivative (AmAzZLys), which is doubly functionalized with amino and azido substituents at the meta positions of the benzyl moiety, and demonstrated its applicability for creating protein conjugates. AmAzZLys was incorporated into proteins in Escherichia coli, by using the ZLys-specific PylRS variant. AmAzZLys was then site-specifically incorporated into a camelid single-domain antibody specific to the epidermal growth factor receptor (EGFR). A one-pot reaction demonstrated that the phenyl amine and azide were efficiently linked to the 5 kDa polyethylene glycol and a fluorescent probe, respectively, through specific bio-orthogonal chemistry. The antibody was then tested for the ability to form a photo-cross-link between its phenylazide moiety and the antigen, while the amino group on the same ring was used for chemical labeling. When incorporated at a selected position in the antibody and exposed to 365 nm light, AmAzZLys formed a covalent bond with the EGFR ectodomain, with the phenylamine moiety labeled fluorescently prior to the reaction. The present results illuminated the versatility of the ZLys scaffold, which can accommodate multiple reactive groups useful for protein conjugation.


Subject(s)
Amino Acids/chemistry , Biochemistry/methods , Proteins/chemistry , Single-Domain Antibodies/chemistry , Antibodies/chemistry , Azides/chemistry , ErbB Receptors/chemistry , ErbB Receptors/immunology , Fluorescent Dyes/chemistry , Methanosarcina/enzymology , Polyethylene Glycols/chemistry , Surface Plasmon Resonance
7.
Biochem Biophys Res Commun ; 431(2): 296-301, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23291178

ABSTRACT

Many important therapeutic targets are secreted proteins with multiple disulfide bonds, such as antibodies, cytokines, hormones, and proteases. The preparation of these proteins for structural and functional analyses using cell-based expression systems still suffers from several issues, such as inefficiency, low yield, and difficulty in stable-isotope labeling. The cell-free (or in vitro) protein synthesis system has become a useful protein production method. The openness of the cell-free system allows direct control of the reaction environment to promote protein folding, making it well suited for the synthesis of disulfide-containing proteins. In this study, we developed the Escherichia coli (E. coli) cell lysate-based cell-free synthesis system for disulfide-containing proteins, which can produce sufficient amounts of functional proteins for NMR analyses. Disulfide bond formation was facilitated by the use of glutathione buffer. In addition, disulfide isomerase, DsbC, catalyzed the efficient shuffling of incorrectly formed disulfide bonds during the protein synthesis reaction. We successfully synthesized milligram quantities of functional (15)N-labeled higher eukaryotic proteins, bovine pancreatic trypsin inhibitor (BPTI) and human lysozyme C (LYZ). The NMR spectra and functional analyses indicated that the synthesized proteins are both catalytically functional and properly folded. Thus, the cell-free system is useful for the synthesis of disulfide-containing proteins for structural and functional analyses.


Subject(s)
Aprotinin/biosynthesis , Cell-Free System , Disulfides/chemistry , Muramidase/biosynthesis , Proteins/chemistry , Animals , Aprotinin/chemistry , Buffers , Cattle , Glutathione/chemistry , Humans , Muramidase/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Biosynthesis , Protein Folding
8.
Bioorg Med Chem ; 20(22): 6579-82, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23062822

ABSTRACT

Immuno-PET is a promising approach for improved cancer diagnosis, by taking advantage of the high specificity of antibodies. Here, we present a novel cell-free protein synthesis method for preparing a positron emitter labeled-antibody. Functional anti-human EGFRvIII single chain Fv, MR1-1, was successfully labeled with carbon-11 (half-life=20.4 min) in 5 min (36% yield) by the direct incorporation of the clinical PET tracer, l-[(11)C]methionine. The product [(11)C]MR1-1 was easily and rapidly isolated with high radiochemical purity (>95%) from the reaction solution, by affinity purification. This method would be widely applicable to the preparation of radiolabeled antibodies for PET imaging.


Subject(s)
Radiopharmaceuticals/chemistry , Single-Chain Antibodies/immunology , Animals , Carbon Radioisotopes/chemistry , Cell Line, Tumor , Cell-Free System , ErbB Receptors/immunology , ErbB Receptors/metabolism , Glioma/diagnostic imaging , Humans , Hydrazines/chemistry , Isotope Labeling , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Rats , Single-Chain Antibodies/chemistry
9.
Nucleic Acids Res ; 40(20): 10576-84, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22909996

ABSTRACT

At earlier stages in the evolution of the universal genetic code, fewer than 20 amino acids were considered to be used. Although this notion is supported by a wide range of data, the actual existence and function of the genetic codes with a limited set of canonical amino acids have not been addressed experimentally, in contrast to the successful development of the expanded codes. Here, we constructed artificial genetic codes involving a reduced alphabet. In one of the codes, a tRNAAla variant with the Trp anticodon reassigns alanine to an unassigned UGG codon in the Escherichia coli S30 cell-free translation system lacking tryptophan. We confirmed that the efficiency and accuracy of protein synthesis by this Trp-lacking code were comparable to those by the universal genetic code, by an amino acid composition analysis, green fluorescent protein fluorescence measurements and the crystal structure determination. We also showed that another code, in which UGU/UGC codons are assigned to Ser, synthesizes an active enzyme. This method will provide not only new insights into primordial genetic codes, but also an essential protein engineering tool for the assessment of the early stages of protein evolution and for the improvement of pharmaceuticals.


Subject(s)
Genetic Code , Protein Engineering , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Codon , Genetic Variation , Molecular Sequence Data , Protein Biosynthesis , RNA, Transfer, Ala/chemistry , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics
10.
Anal Biochem ; 411(2): 223-9, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21256106

ABSTRACT

During recent years, the targets of protein structure analysis using nuclear magnetic resonance spectroscopy have become larger and more complicated. As a result, a complete and precise stable isotope labeling technique has been desired. A cell-free protein synthesis system is appropriate for this purpose. In the current study, we achieved precise and complete (15)N and (2)H labeling using an Escherichia coli cell extract-based cell-free protein synthesis system by controlling the metabolic reactions in the system with their chemical inhibitors. The addition of aminooxyacetate, d-malate, l-methionine sulfoximine, S-methyl-l-cysteine sulfoximine, 6-diazo-5-oxo-l-norleucine, and 5-diazo-4-oxo-l-norvaline was quite effective for precise amino acid-selective (15)N labeling even for aspartic acid, asparagine, glutamic acid, and glutamine, which generally suffer from severe isotope scrambling and dilution when using the conventional cell-free system. For (2)H labeling, the back-protonation of the H(α) and H(ß) positions, which commonly occurred in the conventional system, was dramatically suppressed by simply adding aminooxyacetate and d-malate to the cell-free system except for the H(α) positions in methionine and cysteine.


Subject(s)
Escherichia coli Proteins/biosynthesis , Isotope Labeling/methods , Amino Acids/chemistry , Amino Acids/metabolism , Cell-Free System , Deuterium/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Nitrogen Isotopes/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
J Biomol NMR ; 48(4): 193-201, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21052777

ABSTRACT

Improvement of the cell-free protein synthesis system (CF) over the past decade have made it one of the most powerful protein production methods. The CF approach is especially useful for stable-isotope (SI) labeling of proteins for NMR analysis. However, it is less popular than expected, partly because the SI-labeled amino acids used for SI labeling by the CF are too expensive. In the present study, we developed a simple and inexpensive method for producing an SI-labeled protein using Escherichia coli cell extract-based CF. This method takes advantage of endogenous metabolic conversions to generate SI-labeled asparagine, glutamine, cysteine, and tryptophan, which are much more expensive than the other 16 kinds of SI-labeled amino acids, from inexpensive sources, such as SI-labeled algal amino acid mixture, SI-labeled indole, and sodium sulfide, during the CF reaction. As compared with the conventional method employing 20 kinds of SI-labeled amino acids, highly enriched uniform SI-labeling with similar labeling efficiency was achieved at a greatly reduced cost with the newly developed method. Therefore, our method solves the cost problem of the SI labeling of proteins using the CF.


Subject(s)
Escherichia coli , Isotope Labeling/methods , Nitrogen Isotopes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry , Amino Acids/analysis , Amino Acids/chemistry , Amino Acids/metabolism , Asparagine/chemistry , Cysteine/chemistry , Escherichia coli/chemistry , Escherichia coli/cytology , Escherichia coli/metabolism , Glutamine/chemistry , Protein Biosynthesis , Proteins/metabolism , Tryptophan/chemistry
12.
Structure ; 18(9): 1127-39, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826339

ABSTRACT

The zinc finger CW (zf-CW) domain is a motif of about 60 residues that is frequently found in proteins involved in epigenetic regulation. Here, we determined the NMR solution structure of the zf-CW domain of the human zf-CW and PWWP domain containing protein 1 (ZCWPW1). The zf-CW domain adopts a new fold in which a zinc ion is coordinated tetrahedrally by four conserved Cys ligand residues. The tertiary structure of the zf-CW domain partially resembles that adopted by the plant homeo domain (PHD) finger bound to the histone tail, suggesting that the zf-CW domain and the PHD finger have similar functions. The solution structure of the complex of the zf-CW domain with the histone H3 tail peptide (1-10) with trimethylated K4 clarified its binding mode. Our structural and biochemical studies have identified the zf-CW domain as a member of the histone modification reader modules for epigenetic regulation.


Subject(s)
Histones/chemistry , Zinc Fingers , Amino Acid Sequence , Binding Sites , Epigenesis, Genetic , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
13.
Protein Expr Purif ; 68(2): 128-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19664715

ABSTRACT

High-throughput protein production systems have become an important issue, because protein production is one of the bottleneck steps in large-scale structural and functional analyses of proteins. We have developed a dialysis reactor and a fully automated system for protein production using the dialysis cell-free synthesis method, which we previously established to produce protein samples on a milligram scale in a high-throughput manner. The dialysis reactor was designed to be suitable for an automated system and has six dialysis cups attached to a flat dialysis membrane. The automated system is based on a Tecan Freedom EVO 200 workstation in a three-arm configuration, and is equipped with shaking incubators, a vacuum module, a robotic centrifuge, a plate heat sealer, and a custom-made tilting carrier for collection of reaction solutions from the flat-bottom cups with dialysis membranes. The consecutive process, from the dialysis cell-free protein synthesis to the partial purification by immobilized metal affinity chromatography on a 96-well filtration plate, was performed within ca. 14h, including 8h of cell-free protein synthesis. The proteins were eluted stepwise in a high concentration using EDTA by centrifugation, while the resin in the filtration plate was washed on the vacuum manifold. The system was validated to be able to simultaneously and automatically produce up to 96 proteins in yields of several milligrams with high well-to-well reliability, sufficient for structural and functional analyses of proteins. The protein samples produced by the automated system have been utilized for NMR screening to judge the protein foldedness and for structure determinations using heteronuclear multi-dimensional NMR spectroscopy. The automated high-throughput protein production system represents an important breakthrough in the structural and functional studies of proteins and has already contributed a massive amount of results in the structural genomics project at the RIKEN Structural Genomics/Proteomics Initiative (RSGI).


Subject(s)
Automation, Laboratory/instrumentation , Cell-Free System , Dialysis/instrumentation , Protein Biosynthesis , Proteins/isolation & purification , Chromatography, Affinity/instrumentation , Chromatography, Affinity/methods , Dialysis/methods , Electrophoresis, Polyacrylamide Gel , Equipment Design , Histidine , Oligopeptides , Proteins/genetics
14.
Rapid Commun Mass Spectrom ; 23(11): 1647-53, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19418491

ABSTRACT

Sodium dodecyl sulfate (SDS), an anionic surfactant, is widely used in peptide and protein sample preparation. When the sample is analyzed by matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), this surfactant can often cause signal suppression. We have previously reported an on-probe sample preparation method using a suspension of anion-exchange silica gel and sinapinic acid (i.e., gel-SA suspension) as a matrix, thereby greatly improving the MALDI signal detection of the protein solutions containing SDS. In this study, we found that a certain amount of SDS enhanced the MALDI signal intensity for protein samples. This effect was also observed when using sodium decyl sulfate and sodium tetradecyl sulfate instead of SDS. Furthermore, this on-probe sample preparation method using both SDS and the gel-SA suspension improved the detection limit of protein samples in the MALDI-MS analysis by about ten-fold as compared to that of protein samples without SDS and the gel-SA suspension. This method can be applied not only to the MALDI-MS analysis of samples containing SDS, but also to the examination of proteins at femtomole levels or insoluble proteins such as membrane proteins.


Subject(s)
Proteins/chemistry , Sodium Dodecyl Sulfate/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Horses , Silica Gel , Silicon Dioxide/chemistry
15.
Protein Sci ; 18(1): 80-91, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19177353

ABSTRACT

The muscleblind-like (MBNL) proteins 1, 2, and 3, which contain four CCCH zinc finger motifs (ZF1-4), are involved in the differentiation of muscle inclusion by controlling the splicing patterns of several pre-mRNAs. Especially, MBNL1 plays a crucial role in myotonic dystrophy. The CCCH zinc finger is a sequence motif found in many RNA binding proteins and is suggested to play an important role in the recognition of RNA molecules. Here, we solved the solution structures of both tandem zinc finger (TZF) motifs, TZF12 (comprising ZF1 and ZF2) and TZF34 (ZF3 and ZF4), in MBNL2 from Homo sapiens. In TZF12 of MBNL2, ZF1 and ZF2 adopt a similar fold, as reported previously for the CCCH-type zinc fingers in the TIS11d protein. The linker between ZF1 and ZF2 in MBNL2 forms an antiparallel beta-sheet with the N-terminal extension of ZF1. Furthermore, ZF1 and ZF2 in MBNL2 interact with each other through hydrophobic interactions. Consequently, TZF12 forms a single, compact global fold, where ZF1 and ZF2 are approximately symmetrical about the C2 axis. The structure of the second tandem zinc finger (TZF34) in MBNL2 is similar to that of TZF12. This novel three-dimensional structure of the TZF domains in MBNL2 provides a basis for functional studies of the CCCH-type zinc finger motifs in the MBNL protein family.


Subject(s)
RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Zinc Fingers , Amino Acid Sequence , Binding Sites , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , Sequence Analysis, Protein
16.
Protein Sci ; 16(8): 1577-87, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17656577

ABSTRACT

The zinc finger HIT domain is a sequence motif found in many proteins, including thyroid hormone receptor interacting protein 3 (TRIP-3), which is possibly involved in maturity-onset diabetes of the young (MODY). Novel zinc finger motifs are suggested to play important roles in gene regulation and chromatin remodeling. Here, we determined the high-resolution solution structure of the zinc finger HIT domain in ZNHIT2 (protein FON) from Homo sapiens, by an NMR method based on 567 upper distance limits derived from NOE intensities measured in three-dimensional NOESY spectra. The structure yielded a backbone RMSD to the mean coordinates of 0.19 A for the structured residues 12-48. The fold consists of two consecutive antiparallel beta-sheets and two short C-terminal helices packed against the second beta-sheet, and binds two zinc ions. Both zinc ions are coordinated tetrahedrally via a CCCC-CCHC motif to the ligand residues of the zf-HIT domain in an interleaved manner. The tertiary structure of the zinc finger HIT domain closely resembles the folds of the B-box, RING finger, and PHD domains with a cross-brace zinc coordination mode, but is distinct from them. The unique three-dimensional structure of the zinc finger HIT domain revealed a novel zinc-binding fold, as a new member of the treble clef domain family. On the basis of the structural data, we discuss the possible functional roles of the zinc finger HIT domain.


Subject(s)
Phosphoproteins/chemistry , Zinc Fingers , Amino Acid Sequence , Consensus Sequence , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphoproteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Solutions/chemistry , Surface Properties , Zinc/chemistry , Zinc/metabolism
17.
Protein Sci ; 16(8): 1788-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17600150

ABSTRACT

The general transcription factor TFII-I, with the corresponding gene name GTF2I, is an unusual transcriptional regulator that associates with both basal and signal-induced transcription factors. TFII-I consists of six GTF2I repeat domains, called I-repeats R1-R6. The structure and function of the GTF2I domain are not clearly understood, even though it contains a helix-loop-helix motif, which is considered to be the protein-protein interaction area, based on biochemical analyses. Here, we report the solution structure of the fifth repeat of the six GTF2I repeat domains from murine TFII-I, which was determined by heteronuclear multidimensional NMR spectroscopy (PDB code 1Q60). The three-dimensional structure of the GTF2I domain is classified as a new fold, consisting of four helices (residues 8-24, 34-39, 63-71, and 83-91), two antiparallel beta strands (residues 44-47 and 77-80), and a well-defined loop containing two beta-turns between sheet 1 and helix 3. All of the repeats probably have similar folds to that of repeat 5, because the conserved residues in the GTF2I repeat domains are assembled on the hydrophobic core, turns, and secondary structure elements, as revealed by a comparison of the sequences of the first through the sixth GTF2I repeats in TFII-I.


Subject(s)
Transcription Factors, TFII/chemistry , Amino Acid Sequence , Animals , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , Sequence Alignment , Solutions , Transcription Factors, TFII/genetics
18.
J Mol Biol ; 369(1): 222-38, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17428495

ABSTRACT

SWIRM is a conserved domain found in several chromatin-associated proteins. Based on their sequences, the SWIRM family members can be classified into three subfamilies, which are represented by Swi3, LSD1, and Ada2. Here we report the SWIRM structure of human MYb-like, Swirm and Mpn domain-containing protein-1 (MYSM1). The MYSM1 SWIRM structure forms a compact HTH-related fold comprising five alpha-helices, which best resembles the Swi3 SWIRM structure, among the known SWIRM structures. The MYSM1 and Swi3 SWIRM structures are more similar to the LSD1 structure than the Ada2alpha structure. The SWIRM domains of MYSM1 and LSD1 lacked DNA binding activity, while those of Ada2alpha and the human Swi3 counterpart, SMARCC2, bound DNA. The dissimilarity in the DNA-binding ability of the MYSM1 and SMARCC2 SWIRM domains might be due to a couple of amino acid differences in the last helix. These results indicate that the SWIRM family has indeed diverged into three structural subfamilies (Swi3/MYSM1, LSD1, and Ada2 types), and that the Swi3/MYSM1-type subfamily has further diverged into two functionally distinct groups. We also solved the structure of the SANT domain of MYSM1, and demonstrated that it bound DNA with a similar mode to that of the c-Myb DNA-binding domain.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , DNA/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Solutions , Structure-Activity Relationship , Trans-Activators , Ubiquitin-Specific Proteases
19.
J Biomol NMR ; 37(3): 225-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17237976

ABSTRACT

Cell-free protein synthesis is suitable for stable-isotope labeling of proteins for NMR analysis. The Escherichia coli cell-free system containing potassium acetate for efficient translation (KOAc system) is usually used for stable-isotope labeling, although it is less productive than other systems. A system containing a high concentration of potassium L-glutamate (L-Glu system), instead of potassium acetate, is highly productive, but cannot be used for stable-isotope labeling of Glu residues. In this study, we have developed a new cell-free system that uses potassium D-glutamate (D-Glu system). The productivity of the D-Glu system is approximately twice that of the KOAc system. The cross peak intensities in the 1H-15N HSQC spectrum of the uniformly stable-isotope labeled Ras protein, prepared with the D-Glu system, were similar to those obtained with the KOAc system, except that the Asp intensities were much higher for the protein produced with the D-Glu system. These results indicate that the D-Glu system is a highly productive cell-free system that is especially useful for stable-isotope labeling of proteins.


Subject(s)
Carbon Isotopes/chemistry , Isotope Labeling/methods , Magnetic Resonance Spectroscopy/methods , Nitrogen Isotopes/chemistry , Protein Biosynthesis , Proteins/chemistry , Aspartic Acid/chemistry , Cell-Free System , Escherichia coli/metabolism , Glutamates/chemistry , Isotopes , Proline/chemistry , ras Proteins/metabolism
20.
J Struct Funct Genomics ; 7(2): 93-100, 2006 Jun.
Article in English | MEDLINE | ID: mdl-17146616

ABSTRACT

Cell-free protein synthesis has become one of the standard methods for protein expression. The cell-free method is suitable for the synthesis of a protein that requires a ligand for its enzymatic activity and/or structure formation and stabilization, since it is an open system, which allows us to add the proper ligand to the reaction mixture. A large number of proteins that require zinc for their function are involved in diverse cellular processes, including transcription, DNA replication, metabolism, and cell signaling. In this study, we analyzed the effects of zinc on the cell-free synthesis of plant-specific zinc-binding transcription factors. The solubility and/or stability of the proteins were significantly increased in the presence of the proper concentration of zinc during the cell-free reaction. NMR analyses confirmed that correctly folded proteins were synthesized by the cell-free method. These results indicate that the cell-free method can be used to synthesize correctly folded and functional zinc-binding proteins.


Subject(s)
Arabidopsis Proteins/biosynthesis , Genomics/methods , Plant Proteins/biosynthesis , Zinc/pharmacology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Cell-Free System , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/chemistry , Chloramphenicol O-Acetyltransferase/genetics , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Nitrogen Isotopes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/biosynthesis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Folding , Protein Structure, Tertiary , Repressor Proteins/biosynthesis , Repressor Proteins/chemistry , Repressor Proteins/genetics , Solubility , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...