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










Publication year range
1.
J Mol Biol ; 431(12): 2298-2319, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31026448

ABSTRACT

The INhibitor of Growth (ING) family of tumor suppressors regulates the transcriptional state of chromatin by recruiting remodeling complexes to sites with histone H3 trimethylated at lysine 4 (H3K4me3). This modification is recognized by the plant homeodomain (PHD) present at the C-terminus of the five ING proteins. ING5 facilitates histone H3 acetylation by the HBO1 complex, and also H4 acetylation by the MOZ/MORF complex. We show that ING5 forms homodimers through its N-terminal domain, which folds independently into an elongated coiled-coil structure. The central region of ING5, which contains the nuclear localization sequence, is flexible and disordered, but it binds dsDNA with micromolar affinity. NMR analysis of the full-length protein reveals that the two PHD fingers of the dimer are chemically equivalent and independent of the rest of the molecule, and they bind H3K4me3 in the same way as the isolated PHD. We have observed that ING5 can form heterodimers with the highly homologous ING4, and that two of three primary tumor-associated mutants in the N-terminal domain strongly destabilize the coiled-coil structure. They also affect cell proliferation and cell cycle phase distribution, suggesting a driver role in cancer progression.


Subject(s)
Histones/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Histones/chemistry , Humans , Models, Molecular , Protein Domains , Protein Multimerization , Sequence Alignment , Transcription Factors/chemistry , Tumor Suppressor Proteins/chemistry
2.
Nat Commun ; 8: 15163, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28516903

ABSTRACT

Heterotrimeric G proteins are quintessential signalling switches activated by nucleotide exchange on Gα. Although activation is predominantly carried out by G-protein-coupled receptors (GPCRs), non-receptor guanine-nucleotide exchange factors (GEFs) have emerged as critical signalling molecules and therapeutic targets. Here we characterize the molecular mechanism of G-protein activation by a family of non-receptor GEFs containing a Gα-binding and -activating (GBA) motif. We combine NMR spectroscopy, computational modelling and biochemistry to map changes in Gα caused by binding of GBA proteins with residue-level resolution. We find that the GBA motif binds to the SwitchII/α3 cleft of Gα and induces changes in the G-1/P-loop and G-2 boxes (involved in phosphate binding), but not in the G-4/G-5 boxes (guanine binding). Our findings reveal that G-protein-binding and activation mechanisms are fundamentally different between GBA proteins and GPCRs, and that GEF-mediated perturbation of nucleotide phosphate binding is sufficient for Gα activation.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanosine Diphosphate/metabolism , Microfilament Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Motifs/physiology , Cell Line , Enzyme Activation/physiology , HEK293 Cells , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/physiology , Signal Transduction/physiology
3.
PLoS One ; 12(2): e0172507, 2017.
Article in English | MEDLINE | ID: mdl-28207895

ABSTRACT

The metastasis suppressor KISS1 is reported to be involved in the progression of several solid neoplasias, making it a promising molecular target for controlling their metastasis. The KISS1 sequence contains an N-terminal secretion signal and several dibasic sequences that are proposed to be the proteolytic cleavage sites. We present the first structural characterization of KISS1 by circular dichroism, multi-angle light scattering, small angle X-Ray scattering and NMR spectroscopy. An analysis of the KISS1 backbone NMR chemical shifts does not reveal any preferential conformation and deviation from a random coil ensemble. The backbone 15N transverse relaxation times indicate a mildly reduced mobility for two regions that are rich in bulky residues. The small angle X-ray scattering curve of KISS1 is likewise consistent with a predominantly random coil ensemble, although an ensemble optimization analysis indicates some preference for more extended conformations possibly due to positive charge repulsion between the abundant basic residues. Our results support the hypothesis that KISS1 mostly samples a random coil conformational space, which is consistent with its high susceptibility to proteolysis and the generation of Kisspeptin fragments.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Kisspeptins/chemistry , Protein Conformation , Circular Dichroism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Folding , Scattering, Small Angle
4.
FEBS Lett ; 591(2): 425-432, 2017 01.
Article in English | MEDLINE | ID: mdl-27926782

ABSTRACT

The tumor suppressor inhibitor of growth 4 (ING4) regulates chromatin structure by recruiting the histone acetyl transferase complex HBO1 to sites with histone H3 trimethylated at K4. ING4 dimerizes through its N-terminal domain and recognizes H3K4me3 by the C-terminal plant homeodomain (PHD). The central region of ING4 is disordered and contains the nuclear localization signal. Here, utilizing electrophoresis and nuclear magnetic resonance, we show that ING4 binds double-stranded DNA through its central region with micromolar affinity. Our findings suggest that the cooperativity arising from the presence of two DNA-binding regions in the ING4 dimer, as well as two H3K4me3-binding PHD fingers, may strengthen nucleosome binding and HBO1 complex recruitment.


Subject(s)
Cell Cycle Proteins/metabolism , DNA/chemistry , DNA/metabolism , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Biophysical Phenomena , Cell Cycle Proteins/chemistry , Electrophoretic Mobility Shift Assay , Fluorescence , Homeodomain Proteins/chemistry , Humans , Magnetic Resonance Spectroscopy , Mutant Proteins/metabolism , Protein Binding , Protein Domains , Protein Multimerization , Titrimetry , Tumor Suppressor Proteins/chemistry
5.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 6): 473-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27303901

ABSTRACT

Homing endonucleases are highly specific DNA-cleaving enzymes that recognize and cleave long stretches of DNA. The engineering of these enzymes provides instruments for genome modification in a wide range of fields, including gene targeting. The homing endonuclease I-SceI from the yeast Saccharomyces cerevisiae has been purified after overexpression in Escherichia coli and its crystal structure has been determined in complex with its target DNA. In order to evaluate the number of ions that are involved in the cleavage process, thus determining the catalytic mechanism, crystallization experiments were performed in the presence of Mn(2+), yielding crystals that were suitable for X-ray diffraction analysis. The crystals belonged to the orthorhombic space group P212121, with unit-cell parameters a = 80.11, b = 80.57, c = 130.87 Å, α = ß = γ = 90°. The self-rotation function and the Matthews coefficient suggested the presence of two protein-DNA complexes in the asymmetric unit. The crystals diffracted to a resolution limit of 2.9 Šusing synchrotron radiation. From the anomalous data, it was determined that three cations are involved in catalysis and it was confirmed that I-SceI follows a two-metal-ion DNA-strand cleavage mechanism.


Subject(s)
DNA/chemistry , Deoxyribonucleases, Type II Site-Specific/chemistry , Metals/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Catalysis
6.
Sci Rep ; 6: 28643, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27345490

ABSTRACT

Here, we describe a new strategy that allows the rapid and efficient engineering of mono and multispecific trivalent antibodies. By fusing single-domain antibodies from camelid heavy-chain-only immunoglobulins (VHHs) to the N-terminus of a human collagen XVIII trimerization domain (TIE(XVIII)) we produced monospecific trimerbodies that were efficiently secreted as soluble functional proteins by mammalian cells. The purified VHH-TIE(XVIII) trimerbodies were trimeric in solution and exhibited excellent antigen binding capacity. Furthermore, by connecting with two additional glycine-serine-based linkers three VHH-TIE(XVIII) modules on a single polypeptide chain, we present an approach for the rational design of multispecific tandem trimerbodies with defined stoichiometry and controlled orientation. Using this technology we report here the construction and characterization of a tandem VHH-based trimerbody capable of simultaneously binding to three different antigens: carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR) and green fluorescence protein (GFP). Multispecific tandem VHH-based trimerbodies were well expressed in mammalian cells, had good biophysical properties and were capable of simultaneously binding their targeted antigens. Importantly, these antibodies were very effective in inhibiting the proliferation of human epidermoid carcinoma A431 cells. Multispecific VHH-based trimerbodies are therefore ideal candidates for future applications in various therapeutic areas.


Subject(s)
Antibodies, Bispecific , Protein Engineering , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Camelids, New World , Humans , Mice , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
7.
J Biol Chem ; 290(48): 28727-36, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26363068

ABSTRACT

Homing endonucleases recognize and generate a DNA double-strand break, which has been used to promote gene targeting. These enzymes recognize long DNA stretches; they are highly sequence-specific enzymes and display a very low frequency of cleavage even in complete genomes. Although a large number of homing endonucleases have been identified, the landscape of possible target sequences is still very limited to cover the complexity of the whole eukaryotic genome. Therefore, the finding and molecular analysis of homing endonucleases identified but not yet characterized may widen the landscape of possible target sequences. The previous characterization of protein-DNA interaction before the engineering of new homing endonucleases is essential for further enzyme modification. Here we report the crystal structure of I-CvuI in complex with its target DNA and with the target DNA of I-CreI, a homologue enzyme widely used in genome engineering. To characterize the enzyme cleavage mechanism, we have solved the I-CvuI DNA structures in the presence of non-catalytic (Ca(2+)) and catalytic ions (Mg(2+)). We have also analyzed the metal dependence of DNA cleavage using Mg(2+) ions at different concentrations ranging from non-cleavable to cleavable concentrations obtained from in vitro cleavage experiments. The structure of I-CvuI homing endonuclease expands the current repertoire for engineering custom specificities, both by itself as a new scaffold alone and in hybrid constructs with other related homing endonucleases or other DNA-binding protein templates.


Subject(s)
Chlorella vulgaris/enzymology , Deoxyribonuclease I/chemistry , Plant Proteins/chemistry , Chlorella vulgaris/genetics , Crystallography, X-Ray , Deoxyribonuclease I/genetics , Plant Proteins/genetics , Protein Structure, Tertiary , Structure-Activity Relationship
8.
Nat Commun ; 6: 6439, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25762514

ABSTRACT

The intrinsically disordered protein p15(PAF) regulates DNA replication and repair by binding to the proliferating cell nuclear antigen (PCNA) sliding clamp. We present the structure of the human p15(PAF)-PCNA complex. Crystallography and NMR show the central PCNA-interacting protein motif (PIP-box) of p15(PAF) tightly bound to the front-face of PCNA. In contrast to other PCNA-interacting proteins, p15(PAF) also contacts the inside of, and passes through, the PCNA ring. The disordered p15(PAF) termini emerge at opposite faces of the ring, but remain protected from 20S proteasomal degradation. Both free and PCNA-bound p15(PAF) binds DNA mainly through its histone-like N-terminal tail, while PCNA does not, and a model of the ternary complex with DNA inside the PCNA ring is consistent with electron micrographs. We propose that p15(PAF) acts as a flexible drag that regulates PCNA sliding along the DNA and facilitates the switch from replicative to translesion synthesis polymerase binding.


Subject(s)
Carrier Proteins/chemistry , DNA Repair , DNA Replication , DNA/chemistry , Intrinsically Disordered Proteins/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Amino Acid Motifs , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , DNA/metabolism , DNA-Binding Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
9.
Microb Cell Fact ; 13: 116, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25112455

ABSTRACT

BACKGROUND: Recombinant antibodies are highly successful in many different pathological conditions and currently enjoy overwhelming recognition of their potential. There are a wide variety of protein expression systems available, but almost all therapeutic antibodies are produced in mammalian cell lines, which mimic human glycosylation. The production of clinical-grade antibodies in mammalian cells is, however, extremely expensive. Compared to mammalian systems, protein production in yeast strains such as Pichia pastoris, is simpler, faster and usually results in higher yields. RESULTS: In this work, a trivalent single-chain fragment variable (scFv)-based N-terminal trimerbody, specific for the human carcinoembryonic antigen (CEA), was expressed in human embryonic kidney 293 cells and in Pichia pastoris. Mammalian- and yeast-produced anti-CEA trimerbody molecules display similar functional and structural properties, yet, the yield of trimerbody expressed in P. pastoris is about 20-fold higher than in human cells. CONCLUSIONS: P. pastoris is an efficient expression system for multivalent trimerbody molecules, suitable for their commercial production.


Subject(s)
Biotechnology/methods , Pichia/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Carcinoembryonic Antigen/immunology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , HEK293 Cells , Humans , Immobilized Proteins/metabolism , Protein Stability , Protein Structure, Tertiary , Serum/metabolism , Single-Chain Antibodies/isolation & purification
10.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 2): 256-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24637769

ABSTRACT

Homing endonucleases are highly specific DNA-cleaving enzymes that recognize long stretches of DNA. The engineering of these enzymes provides novel instruments for genome modification in a wide range of fields, including gene targeting, by inducing specific double-strand breaks. I-CvuI is a homing endonuclease from the green alga Chlorella vulgaris. This enzyme was purified after overexpression in Escherichia coli. Crystallization experiments of I-CvuI in complex with its DNA target in the presence of Mg(2+) yielded crystals suitable for X-ray diffraction analysis. The crystals belonged to the orthorhombic space group P212121, with unit-cell parameters a = 62.83, b = 83.56, c = 94.40 Å. The self-rotation function and the Matthews coefficient suggested the presence of one protein-DNA complex per asymmetric unit. The crystals diffracted to a resolution limit of 1.9 Å using synchrotron radiation.


Subject(s)
Chlorella vulgaris/enzymology , Crystallography, X-Ray/methods , DNA/metabolism , Endonucleases/chemistry , Chromatography, Liquid , Crystallization , Electrophoresis, Polyacrylamide Gel , Endonucleases/metabolism , Protein Conformation
11.
Biophys J ; 106(4): 865-74, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24559989

ABSTRACT

We present to our knowledge the first structural characterization of the proliferating-cell-nuclear-antigen-associated factor p15(PAF), showing that it is monomeric and intrinsically disordered in solution but has nonrandom conformational preferences at sites of protein-protein interactions. p15(PAF) is a 12 kDa nuclear protein that acts as a regulator of DNA repair during DNA replication. The p15(PAF) gene is overexpressed in several types of human cancer. The nearly complete NMR backbone assignment of p15(PAF) allowed us to measure 86 N-H(N) residual dipolar couplings. Our residual dipolar coupling analysis reveals nonrandom conformational preferences in distinct regions, including the proliferating-cell-nuclear-antigen-interacting protein motif (PIP-box) and the KEN-box (recognized by the ubiquitin ligase that targets p15(PAF) for degradation). In accordance with these findings, analysis of the (15)N R2 relaxation rates shows a relatively reduced mobility for the residues in these regions. The agreement between the experimental small angle x-ray scattering curve of p15(PAF) and that computed from a statistical coil ensemble corrected for the presence of local secondary structural elements further validates our structural model for p15(PAF). The coincidence of these transiently structured regions with protein-protein interaction and posttranslational modification sites suggests a possible role for these structures as molecular recognition elements for p15(PAF).


Subject(s)
Carrier Proteins/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/metabolism , DNA-Binding Proteins , Humans , Molecular Sequence Data , Protein Binding
12.
PLoS One ; 7(11): e48390, 2012.
Article in English | MEDLINE | ID: mdl-23139781

ABSTRACT

PCNA is an essential factor for DNA replication and repair. It forms a ring shaped structure of 86 kDa by the symmetric association of three identical protomers. The ring encircles the DNA and acts as a docking platform for other proteins, most of them containing the PCNA Interaction Protein sequence (PIP-box). We have used NMR to characterize the interactions of PCNA with several other proteins and fragments in solution. The binding of the PIP-box peptide of the cell cycle inhibitor p21 to PCNA is consistent with the crystal structure of the complex. A shorter p21 peptide binds with reduced affinity but retains most of the molecular recognition determinants. However the binding of the corresponding peptide of the tumor suppressor ING1 is extremely weak, indicating that slight deviations from the consensus PIP-box sequence dramatically reduce the affinity for PCNA, in contrast with a proposed less stringent PIP-box sequence requirement. We could not detect any binding between PCNA and the MCL-1 or the CDK2 protein, reported to interact with PCNA in biochemical assays. This suggests that they do not bind directly to PCNA, or they do but very weakly, with additional unidentified factors stabilizing the interactions in the cell. Backbone dynamics measurements show three PCNA regions with high relative flexibility, including the interdomain connector loop (IDCL) and the C-terminus, both of them involved in the interaction with the PIP-box. Our work provides the basis for high resolution studies of direct ligand binding to PCNA in solution.


Subject(s)
Magnetic Resonance Spectroscopy , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Sequence , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Nuclear Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Proliferating Cell Nuclear Antigen/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Solutions , Thermodynamics , Tumor Suppressor Proteins/metabolism
13.
Protein Expr Purif ; 85(2): 246-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22917812

ABSTRACT

Meganucleases are highly specific endonucleases that recognize and cleave long DNA sequences, making them powerful tools for gene targeting. We describe the production of active recombinant meganucleases suitable for functional and structural studies using a batch-based cell-free protein synthesis method. Isotopic labeling of the I-CreI meganuclease is demonstrated opening the way for structural and ligand binding studies in solution by nuclear magnetic resonance (NMR)(2) which was previously hampered by the problems associated with the toxicity of the enzyme for Escherichia coli limiting its growth. The method can be adapted for the synthesis of soluble engineered variants that are produced as inclusion bodies in bacterial cells, thus facilitating their purification as soluble proteins instead of using denaturing-refolding protocols.


Subject(s)
Cell-Free System , DNA Restriction Enzymes/biosynthesis , Recombinant Proteins/biosynthesis , Biotechnology , DNA/metabolism , DNA Restriction Enzymes/analysis , DNA Restriction Enzymes/chemistry , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Solubility
14.
PLoS One ; 7(6): e39097, 2012.
Article in English | MEDLINE | ID: mdl-22723936

ABSTRACT

Laminins are large heterotrimeric cross-shaped extracellular matrix glycoproteins with terminal globular domains and a coiled-coil region through which the three chains are assembled and covalently linked. Laminins are key components of basement membranes, and they serve as attachment sites for cell adhesion, migration and proliferation. In this work, we produced a recombinant fragment comprising the entire laminin coiled-coil of the α1-, ß1-, and γ1-chains that assemble into a stable heterotrimeric coiled-coil structure independently of the rest of the molecule. This domain was biologically active and not only failed to serve as a substrate for cell attachment, spreading and focal adhesion formation but also inhibited cell adhesion to laminin when added to cells in a soluble form at the time of seeding. Furthermore, gene array expression profiling in cells cultured in the presence of the laminin coiled-coil domain revealed up-regulation of genes involved in cell motility and invasion. These findings were confirmed by real-time quantitative PCR and zymography assays. In conclusion, this study shows for the first time that the laminin coiled-coil domain displays anti-adhesive functions and has potential implications for cell migration during matrix remodeling.


Subject(s)
Cell Movement/genetics , Laminin/genetics , Laminin/metabolism , Protein Interaction Domains and Motifs/physiology , Protein Multimerization , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Enzyme Activation , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Laminin/chemistry , Matrix Metalloproteinase 2/metabolism , Mice , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reproducibility of Results
15.
MAbs ; 4(2): 226-32, 2012.
Article in English | MEDLINE | ID: mdl-22453098

ABSTRACT

We recently described the in vitro and in vivo properties of an engineered homotrimeric antibody made by fusing the N-terminal trimerization region of collagen XVIII NC1 domain to the C-terminus of a scFv fragment [trimerbody (scFv-NC1) 3; 110 kDa]. Here, we demonstrated the utility of the N-terminal trimerization region of collagen XV NC1 domain in the engineering of trivalent antibodies. We constructed several scFv-based trimerbodies containing the human type XV trimerization domain and demonstrated that all the purified trimerbodies were trimeric in solution and exhibited excellent antigen binding capacity. Importantly, type XV trimerbodies demonstrated substantially greater thermal and serum stability and resistance to protease digestion than type XVIII trimerbodies. In summary, the small size, high expression level, solubility and stability of the trimerization domain of type XV collagen make it the ideal choice for engineering homotrimeric antibodies for cancer detection and therapy.


Subject(s)
Antibodies, Neoplasm , Collagen , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Collagen/biosynthesis , Collagen/genetics , Collagen/immunology , HEK293 Cells , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology , Protein Stability , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
16.
J Biol Chem ; 287(14): 10876-84, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334692

ABSTRACT

The protein ING4 binds to histone H3 trimethylated at Lys-4 (H3K4me3) through its C-terminal plant homeodomain, thus recruiting the HBO1 histone acetyltransferase complex to target promoters. The structure of the plant homeodomain finger bound to an H3K4me3 peptide has been described, as well as the disorder and flexibility in the ING4 central region. We report the crystal structure of the ING4 N-terminal domain, which shows an antiparallel coiled-coil homodimer with each protomer folded into a helix-loop-helix structure. This arrangement suggests that ING4 can bind simultaneously two histone tails on the same or different nucleosomes. Dimerization has a direct impact on ING4 tumor suppressor activity because monomeric mutants lose the ability to induce apoptosis after genotoxic stress. Homology modeling based on the ING4 structure suggests that other ING dimers may also exist.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Protein Multimerization , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation , Crystallography, X-Ray , Histone Acetyltransferases/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Tertiary , Tumor Suppressor Proteins/genetics
17.
PLoS One ; 6(2): e16600, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21364740

ABSTRACT

Proliferating Cell Nuclear Antigen (PCNA) is an essential factor for DNA replication and repair. PCNA forms a toroidal, ring shaped structure of 90 kDa by the symmetric association of three identical monomers. The ring encircles the DNA and acts as a platform where polymerases and other proteins dock to carry out different DNA metabolic processes. The amino acid sequence of human PCNA is 35% identical to the yeast homolog, and the two proteins have the same 3D crystal structure. In this report, we give evidence that the budding yeast (sc) and human (h) PCNAs have highly similar structures in solution but differ substantially in their stability and dynamics. hPCNA is less resistant to chemical and thermal denaturation and displays lower cooperativity of unfolding as compared to scPCNA. Solvent exchange rates measurements show that the slowest exchanging backbone amides are at the ß-sheet, in the structure core, and not at the helices, which line the central channel. However, all the backbone amides of hPCNA exchange fast, becoming undetectable within hours, while the signals from the core amides of scPCNA persist for longer times. The high dynamics of the α-helices, which face the DNA in the PCNA-loaded form, is likely to have functional implications for the sliding of the PCNA ring on the DNA since a large hole with a flexible wall facilitates the establishment of protein-DNA interactions that are transient and easily broken. The increased dynamics of hPCNA relative to scPCNA may allow it to acquire multiple induced conformations upon binding to its substrates enlarging its binding diversity.


Subject(s)
Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Sequence Homology , Yeasts/metabolism , Amino Acid Sequence , Antigens, Nuclear/chemistry , Antigens, Nuclear/metabolism , Antigens, Nuclear/physiology , Down-Regulation/physiology , Humans , Models, Molecular , Molecular Sequence Data , Proliferating Cell Nuclear Antigen/physiology , Protein Binding , Protein Denaturation , Protein Folding , Protein Stability , Protein Structure, Secondary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Species Specificity , Thermodynamics , Up-Regulation/physiology , Yeasts/chemistry , Yeasts/physiology
18.
Nucleic Acids Res ; 39(2): 729-43, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20846960

ABSTRACT

Homing endonucleases recognize long target DNA sequences generating an accurate double-strand break that promotes gene targeting through homologous recombination. We have modified the homodimeric I-CreI endonuclease through protein engineering to target a specific DNA sequence within the human RAG1 gene. Mutations in RAG1 produce severe combined immunodeficiency (SCID), a monogenic disease leading to defective immune response in the individuals, leaving them vulnerable to infectious diseases. The structures of two engineered heterodimeric variants and one single-chain variant of I-CreI, in complex with a 24-bp oligonucleotide of the human RAG1 gene sequence, show how the DNA binding is achieved through interactions in the major groove. In addition, the introduction of the G19S mutation in the neighborhood of the catalytic site lowers the reaction energy barrier for DNA cleavage without compromising DNA recognition. Gene-targeting experiments in human cell lines show that the designed single-chain molecule preserves its in vivo activity with higher specificity, further enhanced by the G19S mutation. This is the first time that an engineered meganuclease variant targets the human RAG1 locus by stimulating homologous recombination in human cell lines up to 265 bp away from the cleavage site. Our analysis illustrates the key features for à la carte procedure in protein-DNA recognition design, opening new possibilities for SCID patients whose illness can be treated ex vivo.


Subject(s)
DNA Repair , DNA Restriction Enzymes/chemistry , Genes, RAG-1 , Cell Line , DNA/chemistry , DNA Cleavage , DNA Restriction Enzymes/genetics , DNA Restriction Enzymes/metabolism , Gene Targeting , Genetic Loci , Humans , Models, Molecular , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Engineering , Recombination, Genetic
19.
Proc Natl Acad Sci U S A ; 107(21): 9608-13, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20453199

ABSTRACT

Inositol phosphates (InsPs) are signaling molecules with multiple roles in cells. In particular (InsP(6)) is involved in mRNA export and editing or chromatin remodeling among other events. InsP(6) accumulates as mixed salts (phytate) in storage tissues of plants and plays a key role in their physiology. Human diets that are exclusively grain-based provide an excess of InsP(6) that, through chelation of metal ions, may have a detrimental effect on human health. Ins(1,3,4,5,6)P(5) 2-kinase (InsP(5) 2-kinase or Ipk1) catalyses the synthesis of InsP(6) from InsP(5) and ATP, and is the only enzyme that transfers a phosphate group to the axial 2-OH of the myo-inositide. We present the first structure for an InsP(5) 2-kinase in complex with both substrates and products. This enzyme presents a singular structural region for inositide binding that encompasses almost half of the protein. The key residues in substrate binding are identified, with Asp368 being responsible for recognition of the axial 2-OH. This study sheds light on the unique molecular mechanism for the synthesis of the precursor of inositol pyrophosphates.


Subject(s)
Arabidopsis/enzymology , Inositol Phosphates/chemistry , Inositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Water/chemistry , Amino Acid Sequence , Animals , Binding Sites , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity
20.
Article in English | MEDLINE | ID: mdl-20057083

ABSTRACT

Inositol 1,3,4,5,6-pentakisphosphate kinase (IP(5) 2-K) is an enzyme involved in inositol metabolism that synthesizes IP(6) (inositol 1,2,3,4,5,6-hexakisphosphate) from inositol 1,3,4,5,6-pentakisphosphate (IP(5)) and ATP. IP(6) is the major phosphorus reserve in plants, while in mammals it is involved in multiple cellular events such as DNA editing and chromatin remodelling. In addition, IP(6) is the precursor of other highly phosphorylated inositols which also play highly relevant roles. IP(5) 2-K is the only enzyme that phosphorylates the 2-OH axial position of the inositide and understanding its molecular mechanism of substrate specificity is of great interest in cell biology. IP(5) 2-K from Arabidopsis thaliana has been expressed in Escherichia coli as two different fusion proteins and purified. Both protein preparations yielded crystals of different quality, always in the presence of IP(6). The best crystals obtained for X-ray crystallographic analysis belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 58.124, b = 113.591, c = 142.478 A. Several diffraction data sets were collected for the native enzyme and two heavy-atom derivatives using a synchrotron source.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/chemistry , Arabidopsis/enzymology , Arabidopsis Proteins/chemistry , Crystallization , Crystallography, X-Ray
SELECTION OF CITATIONS
SEARCH DETAIL
...