Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Mol Sci ; 23(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35328554

ABSTRACT

Salp15 is one of the proteins in the saliva of the tick Ixodes scapularis. Together with other biomolecules injected into the mammalian host at the biting site, it helps the tick to sustain its blood meal for days. Salp15 interferes with the cellular immune response of the mammalian host by inhibiting the activation of CD4+ T-lymphocytes. This function is co-opted by pathogens that use the tick as a vector and invade the host when the tick bites, such as Borrelia burgdorferi, the causative agent of Lyme borreliosis. Because of the immunity-suppressing role of Salp15, it has been proposed as a candidate for therapeutic applications in disorders of the immune system. The protein is produced as a 135-residue long polypeptide and secreted without its N-terminal signal 1-21 sequence. Detailed structural studies on Salp15 are lacking because of the difficulty in producing large amounts of the folded protein. We report the production of Salp15 and its structural analysis by NMR. The protein is monomeric and contains a flexible N-terminal region followed by a folded domain with mixed α + ß secondary structures. Our results are consistent with a three-dimensional structural model derived from AlphaFold, which predicts the formation of three disulfide bridges and a free C-terminal cysteine.


Subject(s)
Borrelia burgdorferi , Ixodes , Lyme Disease , Animals , Ixodes/metabolism , Mammals , Saliva , Salivary Proteins and Peptides/metabolism
2.
J Biol Chem ; 294(11): 3947-3956, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30655288

ABSTRACT

Human DNA polymerase δ is essential for DNA replication and acts in conjunction with the processivity factor proliferating cell nuclear antigen (PCNA). In addition to its catalytic subunit (p125), pol δ comprises three regulatory subunits (p50, p68, and p12). PCNA interacts with all of these subunits, but only the interaction with p68 has been structurally characterized. Here, we report solution NMR-, isothermal calorimetry-, and X-ray crystallography-based analyses of the p12-PCNA interaction, which takes part in the modulation of the rate and fidelity of DNA synthesis by pol δ. We show that p12 binds with micromolar affinity to the classical PIP-binding pocket of PCNA via a highly atypical PIP box located at the p12 N terminus. Unlike the canonical PIP box of p68, the PIP box of p12 lacks the conserved glutamine; binds through a 2-fork plug made of an isoleucine and a tyrosine residue at +3 and +8 positions, respectively; and is stabilized by an aspartate at +6 position, which creates a network of intramolecular hydrogen bonds. These findings add to growing evidence that PCNA can bind a diverse range of protein sequences that may be broadly grouped as PIP-like motifs as has been previously suggested.


Subject(s)
DNA Polymerase III/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Motifs , Calorimetry , Catalytic Domain , DNA Polymerase III/chemistry , DNA Polymerase III/isolation & purification , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/isolation & purification
3.
Nucleic Acids Res ; 46(18): 9816-9828, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30102405

ABSTRACT

p15PAF is an oncogenic intrinsically disordered protein that regulates DNA replication and lesion bypass by interacting with the human sliding clamp PCNA. In the absence of DNA, p15PAF traverses the PCNA ring via an extended PIP-box that contacts the sliding surface. Here, we probed the atomic-scale structure of p15PAF-PCNA-DNA ternary complexes. Crystallography and MD simulations show that, when p15PAF occupies two subunits of the PCNA homotrimer, DNA within the ring channel binds the unoccupied subunit. The structure of PCNA-bound p15PAF in the absence and presence of DNA is invariant, and solution NMR confirms that DNA does not displace p15PAF from the ring wall. Thus, p15PAF reduces the available sliding surfaces of PCNA, and may function as a belt that fastens the DNA to the clamp during synthesis by the replicative polymerase (pol δ). This constraint, however, may need to be released for efficient DNA lesion bypass by the translesion synthesis polymerase (pol η). Accordingly, our biochemical data show that p15PAF impairs primer synthesis by pol η-PCNA holoenzyme against both damaged and normal DNA templates. In light of our findings, we discuss the possible mechanistic roles of p15PAF in DNA replication and suppression of DNA lesion bypass.


Subject(s)
Carrier Proteins/chemistry , DNA/chemistry , Intrinsically Disordered Proteins/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Carrier Proteins/genetics , Crystallography, X-Ray , DNA/genetics , DNA Polymerase III/chemistry , DNA Polymerase III/genetics , DNA Replication/genetics , DNA-Binding Proteins , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , Escherichia coli/genetics , Holoenzymes/chemistry , Holoenzymes/genetics , Humans , Intrinsically Disordered Proteins/genetics , Magnetic Resonance Spectroscopy , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Proliferating Cell Nuclear Antigen/genetics
4.
Int J Mol Sci ; 20(24)2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31842432

ABSTRACT

The four member family of "Cyclin and Cystathionine ß-synthase (CBS) domain divalent metal cation transport mediators", CNNMs, are the least-studied mammalian magnesium transport mediators. CNNM4 is abundant in the brain and the intestinal tract, and its abnormal activity causes Jalili Syndrome. Recent findings show that suppression of CNNM4 in mice promotes malignant progression of intestinal polyps and is linked to infertility. The association of CNNM4 with phosphatases of the regenerating liver, PRLs, abrogates its Mg2+-efflux capacity, thus resulting in an increased intracellular Mg2+ concentration that favors tumor growth. Here we present the crystal structures of the two independent intracellular domains of human CNNM4, i.e., the Bateman module and the cyclic nucleotide binding-like domain (cNMP). We also derive a model structure for the full intracellular region in the absence and presence of MgATP and the oncogenic interacting partner, PRL-1. We find that only the Bateman module interacts with ATP and Mg2+, at non-overlapping sites facilitating their positive cooperativity. Furthermore, both domains dimerize autonomously, where the cNMP domain dimer forms a rigid cleft to restrict the Mg2+ induced sliding of the inserting CBS1 motives of the Bateman module, from a twisted to a flat disk shaped dimer.


Subject(s)
Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Magnesium/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Biological Transport , Humans , Magnesium/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Structure-Activity Relationship
5.
Cancer Immunol Immunother ; 67(8): 1251-1260, 2018 08.
Article in English | MEDLINE | ID: mdl-29869168

ABSTRACT

The recruitment of T-cells by bispecific antibodies secreted from adoptively transferred, gene-modified autologous cells has shown satisfactory results in preclinical cancer models. Even so, the approach's translation into the clinic will require incremental improvements to its efficacy and reduction of its toxicity. Here, we characterized a tandem T-cell recruiting bispecific antibody intended to benefit gene-based immunotherapy approaches, which we call the light T-cell engager (LiTE), consisting of an EGFR-specific single-domain VHH antibody fused to a CD3-specific scFv. We generated two LiTEs with the anti-EGFR VHH and the anti-CD3 scFv arranged in both possible orders. Both constructs were well expressed in mammalian cells as highly homogenous monomers in solution with molecular weights of 43 and 41 kDa, respectively. In situ secreted LiTEs bound the cognate antigens of both parental antibodies and triggered the specific cytolysis of EGFR-expressing cancer cells without inducing T-cell activation and cytotoxicity spontaneously or against EGFR-negative cells. Light T-cell engagers are, therefore, suitable for future applications in gene-based immunotherapy approaches.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , ErbB Receptors/immunology , Immunotherapy , Neoplasms/therapy , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Humans , Lymphocyte Activation , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Tumor Cells, Cultured
6.
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
7.
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
8.
Int J Biol Macromol ; 273(Pt 2): 133187, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38880460

ABSTRACT

The coordination of enzymes and regulatory proteins for eukaryotic DNA replication and repair is largely achieved by Proliferating Cell Nuclear Antigen (PCNA), a toroidal homotrimeric protein that embraces the DNA duplex. Many proteins bind PCNA through a conserved sequence known as the PCNA interacting protein motif (PIP). PCNA is further regulated by different post-translational modifications. Phosphorylation at residue Y211 facilitates unlocking stalled replication forks to bypass DNA damage repair processes but increasing nucleotide misincorporation. We explore here how phosphorylation at Y211 affects PCNA recognition of the canonical PIP sequences of the regulatory proteins p21 and p15, which bind with nM and µM affinity, respectively. For that purpose, we have prepared PCNA with p-carboxymethyl-L-phenylalanine (pCMF, a mimetic of phosphorylated tyrosine) at position 211. We have also characterized PCNA binding to the non-canonical PIP sequence of the catalytic subunit of DNA polymerase δ (p125), and to the canonical PIP sequence of the enzyme ubiquitin specific peptidase 29 (USP29) which deubiquitinates PCNA. Our results show that Tyr211 phosphorylation has little effect on the molecular recognition of p21 and p15, and that the PIP sequences of p125 and USP29 bind to the same site on PCNA as other PIP sequences, but with very low affinity.


Subject(s)
Proliferating Cell Nuclear Antigen , Protein Binding , Tyrosine , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/chemistry , Phosphorylation , Tyrosine/metabolism , Tyrosine/chemistry , Humans , Amino Acid Motifs , DNA Polymerase III/metabolism , DNA Polymerase III/chemistry , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/chemistry
9.
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
10.
Int J Biol Macromol ; 242(Pt 1): 124724, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37148949

ABSTRACT

Proteins belonging to the ING family regulate 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-terminal region of the five ING proteins. ING3 facilitates acetylation of histones H2A and H4 by the NuA4-Tip60 MYST histone acetyl transferase complex, and it has been proposed to be an oncoprotein. The crystal structure of the N-terminal domain of ING3 shows that it forms homodimers with an antiparallel coiled-coil fold. The crystal structure of the PHD is similar to those of its four homologs. These structures explain the possible deleterious effects of ING3 mutations detected in tumors. The PHD binds histone H3K4me3 with low-micromolar, and binds the non-methylated histone with a 54-fold reduced affinity. Our structure explains the impact of site directed mutagenesis experiments on histone recognition. These structural features could not be confirmed for the full-length protein as solubility was insufficient for structural studies, but the structure of its folded domains suggest a conserved structural organization for the ING proteins as homodimers and bivalent readers of the histone H3K4me3 mark.


Subject(s)
Histones , Tumor Suppressor Proteins , Histones/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Homeodomain Proteins/metabolism , Chromatin , Protein Binding
11.
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
12.
Nucleic Acids Res ; 37(16): 5405-19, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19584299

ABSTRACT

Sequence-specific endonucleases recognizing long target sequences are emerging as powerful tools for genome engineering. These endonucleases could be used to correct deleterious mutations or to inactivate viruses, in a new approach to molecular medicine. However, such applications are highly demanding in terms of safety. Mutations in the human RAG1 gene cause severe combined immunodeficiency (SCID). Using the I-CreI dimeric LAGLIDADG meganuclease as a scaffold, we describe here the engineering of a series of endonucleases cleaving the human RAG1 gene, including obligate heterodimers and single-chain molecules. We show that a novel single-chain design, in which two different monomers are linked to form a single molecule, can induce high levels of recombination while safeguarding more effectively against potential genotoxicity. We provide here the first demonstration that an engineered meganuclease can induce targeted recombination at an endogenous locus in up to 6% of transfected human cells. These properties rank this new generation of endonucleases among the best molecular scissors available for genome surgery strategies, potentially avoiding the deleterious effects of previous gene therapy approaches.


Subject(s)
DNA Restriction Enzymes/genetics , Gene Targeting , Genes, RAG-1 , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Restriction Enzymes/chemistry , DNA Restriction Enzymes/metabolism , Dimerization , Genetic Engineering , Humans , Recombination, Genetic , Severe Combined Immunodeficiency/genetics
13.
Clin Cancer Res ; 27(11): 3167-3177, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33785484

ABSTRACT

PURPOSE: The induction of 4-1BB signaling by agonistic antibodies can drive the activation and proliferation of effector T cells and thereby enhance a T-cell-mediated antitumor response. Systemic administration of anti-4-1BB-agonistic IgGs, although effective preclinically, has not advanced in clinical development due to their severe hepatotoxicity. EXPERIMENTAL DESIGN: Here, we generated a humanized EGFR-specific 4-1BB-agonistic trimerbody, which replaces the IgG Fc region with a human collagen homotrimerization domain. It was characterized by structural analysis and in vitro functional studies. We also assessed pharmacokinetics, antitumor efficacy, and toxicity in vivo. RESULTS: In the presence of a T-cell receptor signal, the trimerbody provided potent T-cell costimulation that was strictly dependent on 4-1BB hyperclustering at the point of contact with a tumor antigen-displaying cell surface. It exhibits significant antitumor activity in vivo, without hepatotoxicity, in a wide range of human tumors including colorectal and breast cancer cell-derived xenografts, and non-small cell lung cancer patient-derived xenografts associated with increased tumor-infiltrating CD8+ T cells. The combination of the trimerbody with a PD-L1 blocker led to increased IFNγ secretion in vitro and resulted in tumor regression in humanized mice bearing aggressive triple-negative breast cancer. CONCLUSIONS: These results demonstrate the nontoxic broad antitumor activity of humanized Fc-free tumor-specific 4-1BB-agonistic trimerbodies and their synergy with checkpoint blockers, which may provide a way to elicit responses in most patients with cancer while avoiding Fc-mediated adverse reactions.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , ErbB Receptors , Immunotherapy/methods , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/therapeutic use , Animals , Breast Neoplasms/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line , Disease Models, Animal , Female , Lung Neoplasms/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Mice, Transgenic , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
14.
Nat Commun ; 11(1): 1109, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111820

ABSTRACT

In eukaryotes, DNA polymerase δ (Pol δ) bound to the proliferating cell nuclear antigen (PCNA) replicates the lagging strand and cooperates with flap endonuclease 1 (FEN1) to process the Okazaki fragments for their ligation. We present the high-resolution cryo-EM structure of the human processive Pol δ-DNA-PCNA complex in the absence and presence of FEN1. Pol δ is anchored to one of the three PCNA monomers through the C-terminal domain of the catalytic subunit. The catalytic core sits on top of PCNA in an open configuration while the regulatory subunits project laterally. This arrangement allows PCNA to thread and stabilize the DNA exiting the catalytic cleft and recruit FEN1 to one unoccupied monomer in a toolbelt fashion. Alternative holoenzyme conformations reveal important functional interactions that maintain PCNA orientation during synthesis. This work sheds light on the structural basis of Pol δ's activity in replicating the human genome.


Subject(s)
DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Amino Acid Motifs , Catalytic Domain , Cryoelectron Microscopy , DNA/metabolism , DNA Polymerase III/genetics , DNA Replication , Flap Endonucleases/chemistry , Flap Endonucleases/metabolism , Holoenzymes , Humans , Models, Molecular , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Subunits , Structure-Activity Relationship
15.
J Bacteriol ; 191(3): 832-43, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047354

ABSTRACT

The capacity of Staphylococcus aureus to form biofilms on host tissues and implanted medical devices is one of the major virulence traits underlying persistent and chronic infections. The matrix in which S. aureus cells are encased in a biofilm often consists of the polysaccharide intercellular adhesin (PIA) or poly-N-acetyl glucosamine (PNAG). However, surface proteins capable of promoting biofilm development in the absence of PIA/PNAG exopolysaccharide have been described. Here, we used two-dimensional nano-liquid chromatography and mass spectrometry to investigate the composition of a proteinaceous biofilm matrix and identified protein A (spa) as an essential component of the biofilm; protein A induced bacterial aggregation in liquid medium and biofilm formation under standing and flow conditions. Exogenous addition of synthetic protein A or supernatants containing secreted protein A to growth media induced biofilm development, indicating that protein A can promote biofilm development without being covalently anchored to the cell wall. Protein A-mediated biofilm formation was completely inhibited in a dose-dependent manner by addition of serum, purified immunoglobulin G, or anti-protein A-specific antibodies. A murine model of subcutaneous catheter infection unveiled a significant role for protein A in the development of biofilm-associated infections, as the amount of protein A-deficient bacteria recovered from the catheter was significantly lower than that of wild-type bacteria when both strains were used to coinfect the implanted medical device. Our results suggest a novel role for protein A complementary to its known capacity to interact with multiple immunologically important eukaryotic receptors.


Subject(s)
Staphylococcal Protein A/pharmacology , Staphylococcus aureus/physiology , Animals , Antibodies/immunology , Antibodies/pharmacology , Bacterial Adhesion/drug effects , Bacterial Adhesion/genetics , Biofilms/drug effects , Biofilms/growth & development , Blotting, Western , Catheter-Related Infections/microbiology , Chromatography, Liquid , Electrophoresis , Immunoglobulin G/pharmacology , Male , Mice , Microscopy, Fluorescence , Polymerase Chain Reaction , Serum , Staphylococcal Infections/microbiology , Staphylococcal Protein A/immunology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Tandem Mass Spectrometry
16.
Infect Immun ; 77(9): 3978-91, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19581398

ABSTRACT

Staphylococcus aureus can establish chronic infections on implanted medical devices due to its capacity to form biofilms. Analysis of the factors that assemble cells into a biofilm has revealed the occurrence of strains that produce either a polysaccharide intercellular adhesin/poly-N-acetylglucosamine (PIA/PNAG) exopolysaccharide- or a protein-dependent biofilm. Examination of the influence of matrix nature on the biofilm capacities of embedded bacteria has remained elusive, because a natural strain that readily converts between a polysaccharide- and a protein-based biofilm has not been studied. Here, we have investigated the clinical methicillin (meticillin)-resistant Staphylococcus aureus strain 132, which is able to alternate between a proteinaceous and an exopolysaccharidic biofilm matrix, depending on environmental conditions. Systematic disruption of each member of the LPXTG surface protein family identified fibronectin-binding proteins (FnBPs) as components of a proteinaceous biofilm formed in Trypticase soy broth-glucose, whereas a PIA/PNAG-dependent biofilm was produced under osmotic stress conditions. The induction of FnBP levels due to a spontaneous agr deficiency present in strain 132 and the activation of a LexA-dependent SOS response or FnBP overexpression from a multicopy plasmid enhanced biofilm development, suggesting a direct relationship between the FnBP levels and the strength of the multicellular phenotype. Scanning electron microscopy revealed that cells growing in the FnBP-mediated biofilm formed highly dense aggregates without any detectable extracellular matrix, whereas cells in a PIA/PNAG-dependent biofilm were embedded in an abundant extracellular material. Finally, studies of the contribution of each type of biofilm matrix to subcutaneous catheter colonization revealed that an FnBP mutant displayed a significantly lower capacity to develop biofilm on implanted catheters than the isogenic PIA/PNAG-deficient mutant.


Subject(s)
Adhesins, Bacterial/physiology , Biofilms , Catheter-Related Infections/etiology , Staphylococcal Infections/etiology , Staphylococcus aureus/physiology , Acetylglucosamine/physiology , Bacterial Proteins/physiology , Humans , Polysaccharides, Bacterial/physiology , SOS Response, Genetics , Trans-Activators/physiology
17.
ACS Chem Biol ; 14(10): 2315-2326, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31479228

ABSTRACT

The proliferating cell nuclear antigen (PCNA)-associated factor p15PAF is a nuclear protein that acts as a regulator of DNA repair during DNA replication. The p15PAF gene is overexpressed in several types of human cancer, and its function is regulated by monoubiquitination of two lysines (K15 and K24) at the protein N-terminal region. We have previously shown that p15PAF is an intrinsically disordered protein which partially folds upon binding to PCNA and independently contacts DNA through its N-terminal tail. Here we present an NMR conformational characterization of p15PAF monoubiquitinated at both K15 and K24 via a disulfide bridge mimicking the isopeptide bond. We show that doubly monoubiquitinated p15PAF is monomeric, intrinsically disordered, and binds to PCNA as nonubiquitinated p15PAF does but interacts with DNA with reduced affinity. Our SAXS-derived conformational ensemble of doubly monoubiquitinated p15PAF shows that the ubiquitin moieties, separated by eight disordered residues, form transient dimers because of the high local effective ubiquitin concentration. This observation and the sequence similarity with histone H3 N-terminal tail suggest that doubly monoubiquitinated p15PAF is a binding target of DNA methyl transferase Dnmt1, as confirmed by calorimetry. Therefore, doubly monoubiquitinated p15PAF directly interacts with PCNA and recruits Dnmt1 for maintenance of DNA methylation during replication.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA-Binding Proteins/metabolism , Ubiquitination , DNA/metabolism , DNA-Binding Proteins/chemistry , Escherichia coli , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Conformation , Protein Domains , Protein Processing, Post-Translational
18.
Front Immunol ; 10: 1791, 2019.
Article in English | MEDLINE | ID: mdl-31417564

ABSTRACT

4-1BB (CD137) is an inducible costimulatory receptor that promotes expansion and survival of activated T cells; and IgG-based 4-1BB-agonistic monoclonal antibodies exhibited potent antitumor activity in clinical trials. However, the clinical development of those antibodies is restricted by major off-tumor toxicities associated with FcγR interactions. We have recently generated an EGFR-targeted 4-1BB-agonistic trimerbody that demonstrated strong antitumor activity and did not induce systemic inflammatory cytokine secretion and hepatotoxicity associated with first-generation 4-1BB agonists. Here, we generate a bispecific 4-1BB-agonistic trimerbody targeting the carcinoembryonic antigen (CEA) that is highly expressed in cancers of diverse origins. The CEA-targeted anti-4-1BB-agonistic trimerbody consists of three 4-1BB-specific single-chain fragment variable antibodies and three anti-CEA single-domain antibodies positioned around a murine collagen XVIII-derived homotrimerization domain. The trimerbody was produced as a homogenous, non-aggregating, soluble protein purifiable by standard affinity chromatographic methods. The purified trimerbody was found to be trimeric in solution, very efficient at recognizing 4-1BB and CEA, and potently costimulating T cells in vitro in the presence of CEA. Therefore, trimerbody-based tumor-targeted 4-1BB costimulation is a broadly applicable and clinically feasible approach to enhance the costimulatory environment of disseminated tumor lesions.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , Carcinoembryonic Antigen/chemistry , Neoplasms/chemistry , Single-Chain Antibodies/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Animals , Antineoplastic Agents, Immunological/immunology , Carcinoembryonic Antigen/immunology , Female , HEK293 Cells , Humans , Mice , Neoplasms/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
19.
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
20.
Sci Rep ; 8(1): 10286, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29980759

ABSTRACT

The high DNA specificity of homing endonucleases makes them a powerful protein scaffold to engineer enzymes for genome manipulation. Understanding their molecular recognition of DNA is an important prerequisite to generate engineered enzymes able to cleave DNA in specific desired genome sites. Protein-DNA recognition studies have been mostly focused on specific direct contacts between amino acid side chains and bases to redesign the binding interface. However, the important role of indirect readout in the central region of the target DNA of the homing endonuclease I-CreI suggested that indirect readout may play a key role in the redesign of protein-DNA interactions. The sequences of the I-CreI central substrate region, 2NN, along with the adjacent 5NNN, are key for substrate cleavage. Here, we analyse the mechanism of target discrimination at the 5NNN region by the I-CreI protein, revealing its critical role in the location and occupancy of the catalytic metal ions, which is crucial for cleavage. Our data highlight the importance of indirect readout for target DNA cleavage, thus aiding I-CreI engineering when targeting new DNA sequences.


Subject(s)
DNA Cleavage , DNA Restriction Enzymes/metabolism , DNA/metabolism , Base Sequence , Binding Sites , Crystallization , DNA/chemistry , DNA Restriction Enzymes/chemistry , Models, Molecular , Protein Conformation , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL