Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Uruguay Oncology Collection
Country/Region as subject
Publication year range
1.
Biotechnol Bioeng ; 115(2): 290-299, 2018 02.
Article in English | MEDLINE | ID: mdl-28976545

ABSTRACT

Affitins are highly stable engineered affinity proteins, originally derived from Sac7d and Sso7d, two 7 kDa DNA-binding polypeptides from Sulfolobus genera. Their efficiency as reagents for intracellular targeting, enzyme inhibition, affinity purification, immunolocalization, and various other applications has been demonstrated. Recently, we have characterized the 7 kDa DNA-binding family, and Aho7c originating from Acidianus hospitalis was shown to be its smallest member with thermostability comparable to those of Sac7d and Sso7d. Here, after four rounds of selection by ribosome display against the human recombinant Epithelial Cell Adhesion Molecule (hrEpCAM), we obtained novel Aho7c-based Affitins. The binders were expressed in soluble form in Escherichia coli, displayed high stability (up to 74°C; pH 0-12) and were shown to be specific for the hrEpCAM extracellular domain with picomolar affinities (KD = 110 pM). Thus, we propose Aho7c as a good candidate for the creation of Affitins with a 10% smaller size than the Sac7d-based ones (60 vs. 66 amino acids).


Subject(s)
Epithelial Cell Adhesion Molecule/metabolism , Protein Engineering/methods , Recombinant Proteins/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Circular Dichroism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cell Adhesion Molecule/genetics , Humans , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
2.
J Pept Sci ; 22(11-12): 711-722, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27933724

ABSTRACT

Because of resistance development by cancer cells against current anticancer drugs, there is a considerable interest in developing novel antitumor agents. We have previously demonstrated that CIGB-552, a novel cell-penetrating synthetic peptide, was effective in reducing tumor size and increasing lifespan in tumor-bearing mice. Studies of protein-peptide interactions have shown that COMMD1 protein is a major mediator of CIGB-552 antitumor activity. Furthermore, a typical serine-protease degradation pattern for CIGB-552 in BALB/c mice serum was identified, yielding peptides which differ from CIGB-552 in size and physical properties. In the present study, we show the results obtained from a comparative analysis between CIGB-552 and its main metabolites regarding physicochemical properties, cellular internalization, and their capability to elicit apoptosis in MCF-7 cells. None of the analyzed metabolites proved to be as effective as CIGB-552 in promoting apoptosis in MCF-7. Taking into account these results, it seemed important to examine their cell-penetrating capacity and interaction with COMMD1. We show that internalization, a lipid binding-dependent process, is impaired as well as metabolite-COMMD1 interaction, key component of the apoptotic mechanism. Altogether, our results suggest that features conferred by the amino acid sequence are decisive for CIGB-552 biological activity, turning it into the minimal functional unit. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Amino Acids/chemistry , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biotransformation , Caspase 7/genetics , Caspase 7/metabolism , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Gene Expression , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship
3.
Biochim Biophys Acta ; 1844(4): 837-49, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24590112

ABSTRACT

MARCKS (Myristoylated Alanine-Rich C Kinase substrate) is a natively unfolded protein that interacts with actin, Ca(2+)-Calmodulin, and some plasma membrane lipids. Such interactions occur at a highly conserved region that is specifically phosphorylated by PKC: the Effector Domain. There are two other conserved domains, MH1 (including a myristoylation site) and MH2, also located in the amino terminal region and whose structure and putative protein binding capabilities are currently unknown. MH2 sequence contains a serine that we described as being phosphorylated only in differentiating neurons (S25 in chick). Here, Circular Dichroism (CD) and Nuclear Magnetic Resonance (NMR) spectroscopy were used to characterize the phosphorylated and unphosphorylated forms of a peptide with the MARCKS sequence surrounding S25. The peptide phosphorylated at this residue is recognized by monoclonal antibody 3C3 (mAb 3C3). CD and NMR data indicated that S25 phosphorylation does not cause extensive modifications in the peptide structure. However, the sharper lines, the absence of multiple spin systems and relaxation dispersion data observed for the phosphorylated peptide suggested a more ordered structure. Surface Plasmon Resonance was employed to compare the binding properties of mAb 3C3 to MARCKS protein and peptide. SPR showed that mAb 3C3 binds to the whole protein and the peptide with a similar affinity, albeit different kinetics. The slightly ordered structure of the phosphorylated peptide might be at the origin of its ability to interact with mAb 3C3 antibody, but this binding did not noticeably modify the peptide structure.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/chemistry , Peptides/chemistry , Phosphoproteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Brain Chemistry , Chick Embryo , Circular Dichroism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Myristoylated Alanine-Rich C Kinase Substrate , Peptides/chemical synthesis , Peptides/metabolism , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Structure, Secondary , Surface Plasmon Resonance
4.
Mol Microbiol ; 94(2): 340-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25145397

ABSTRACT

Several Leptospira species cause leptospirosis, the most extended zoonosis worldwide. In bacteria, two-component systems constitute key signalling pathways, some of which are involved in pathogenesis. The physiological roles of two-component systems in Leptospira are largely unknown, despite identifying several dozens within their genomes. Biochemical confirmation of an operative phosphorelaying two-component system has been obtained so far only for the Hklep/Rrlep pair. It is known that hklep/rrlep knockout strains of Leptospira biflexa result in haem auxotrophy, although their de novo biosynthesis machinery remains fully functional. Haem is essential for Leptospira, but information about Hklep/Rrlep effector function(s) and target(s) is still lacking. We are now reporting a thorough molecular characterization of this system, which we rename HemK/HemR. The DNA HemR-binding motif was determined, and found within the genomes of saprophyte and pathogenic Leptospira. In this way, putative HemR-regulated genes were pinpointed, including haem catabolism-related (hmuO - haem oxygenase) and biosynthesis-related (the hemA/C/D/B/L/E/N/G operon). Specific HemR binding to these two promoters was quantified, and a dual function was observed in vivo, inversely repressing the hmuO, while activating the hemA operon transcription. The crystal structure of HemR receiver domain was determined, leading to a mechanistic model for its dual regulatory role.


Subject(s)
Gene Expression Regulation, Bacterial , Heme/metabolism , Leptospira/genetics , Leptospira/metabolism , Metabolic Networks and Pathways/genetics , Transcription Factors/metabolism , Binding Sites , Crystallography, X-Ray , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Models, Molecular , Operon , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Regulon , Transcription Factors/chemistry , Transcription Factors/genetics
5.
Sci Rep ; 14(1): 12190, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806566

ABSTRACT

The Bovine Leukemia Virus (BLV) Envelope (Env) glycoprotein complex is instrumental in viral infectivity and shapes the host's immune response. This study presents the production and characterization of a soluble furin-mutated BLV Env ectodomain (sBLV-EnvFm) expressed in a stable S2 insect cell line. We purified a 63 kDa soluble protein, corresponding to the monomeric sBLV-EnvFm, which predominantly presented oligomannose and paucimannose N-glycans, with a high content of core fucose structures. Our results demonstrate that our recombinant protein can be recognized from specific antibodies in BLV infected cattle, suggesting its potential as a powerful diagnostic tool. Moreover, the robust humoral immune response it elicited in mice shows its potential contribution to the development of subunit-based vaccines against BLV.


Subject(s)
Antibodies, Viral , Leukemia Virus, Bovine , Recombinant Proteins , Viral Envelope Proteins , Animals , Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/immunology , Cattle , Recombinant Proteins/genetics , Mice , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Antibodies, Viral/immunology , Enzootic Bovine Leukosis/virology , Cell Line , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, env/immunology
6.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 3): 388-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23519414

ABSTRACT

Despite being the most abundant class of immunoglobulins in humans and playing central roles in the adaptive immune response, high-resolution structural data are still lacking for the antigen-binding region of human isotype A antibodies (IgAs). The crystal structures of a human Fab fragment of IgA1 in three different crystal forms are now reported. The three-dimensional organization is similar to those of other Fab classes, but FabA1 seems to be more rigid, being constrained by a hydrophobic core in the interface between the variable and constant domains of the heavy chain (VH-CH1) as well as by a disulfide bridge that connects the light and heavy chains, influencing the relative heavy/light-chain orientation. The crystal structure of the same antibody but with a G-isotype CH1 which is reported to display different antigen affinity has also been solved. The differential structural features reveal plausible mechanisms for constant/variable-domain long-distance effects whereby antibody class switching could alter antigen affinity.


Subject(s)
Antigen-Antibody Reactions , Antigens/chemistry , Binding Sites, Antibody , Immunoglobulin A/chemistry , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Fab Fragments/chemistry , Serine Endopeptidases/chemistry , Antigen-Antibody Reactions/physiology , Antigens/physiology , Clostridium/enzymology , Crystallography, X-Ray , Humans , Immunoglobulin A/physiology , Immunoglobulin Constant Regions/physiology , Immunoglobulin Fab Fragments/physiology , Neisseria gonorrhoeae/enzymology , Protein Structure, Tertiary , Serine Endopeptidases/physiology
7.
Cancer Immunol Immunother ; 62(6): 1107-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23604173

ABSTRACT

The Tn antigen (GalNAcα-O-Ser/Thr) is a well-established tumor-associated marker which represents a good target for the design of anti-tumor vaccines. Several studies have established that the binding of some anti-Tn antibodies could be affected by the density of Tn determinant or/and by the amino acid residues neighboring O-glycosylation sites. In the present study, using synthetic Tn-based vaccines, we have generated a panel of anti-Tn monoclonal antibodies. Analysis of their binding to various synthetic glycopeptides, modifying the amino acid carrier of the GalNAc(*) (Ser* vs Thr*), showed subtle differences in their fine specificities. We found that the recognition of these glycopeptides by some of these MAbs was strongly affected by the Tn backbone, such as a S*S*S* specific MAb (15G9) which failed to recognize a S*T*T* or a T*T*T* structure. Different binding patterns of these antibodies were also observed in FACS and Western blot analysis using three human cancer cell lines (MCF-7, LS174T and Jurkat). Importantly, an immunohistochemical analysis of human tumors (72 breast cancer and 44 colon cancer) showed the existence of different recognition profiles among the five antibodies evaluated, demonstrating that the aglyconic part of the Tn structure (Ser vs Thr) plays a key role in the anti-Tn specificity for breast and colon cancer detection. This new structural feature of the Tn antigen could be of important clinical value, notably due to the increasing interest of this antigen in anticancer vaccine design as well as for the development of anti-Tn antibodies for in vivo diagnostic and therapeutic strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Glycopeptides/immunology , Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity/immunology , Antigens, Tumor-Associated, Carbohydrate/chemistry , Antigens, Tumor-Associated, Carbohydrate/metabolism , Biomarkers, Tumor , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Glycopeptides/chemistry , Glycopeptides/metabolism , Humans , Male , Mice , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding/immunology
8.
Vet Res ; 44: 19, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23506507

ABSTRACT

It is widely accepted that the majority of cancers result from multiple cellular events leading to malignancy after a prolonged period of clinical latency, and that the immune system plays a critical role in the control of cancer progression. Bovine leukemia virus (BLV) is an oncogenic member of the Retroviridae family. Complete genomic sequences of BLV strains isolated from peripheral blood mononuclear cells (PBMC) from cattle have been previously reported. However, a detailed characterization of the complete genome of BLV strains directly isolated from bovine tumors is much needed in order to contribute to the understanding of the mechanisms of leukemogenesis induced by BLV in cattle. In this study, we performed a molecular characterization of BLV complete genomes from bovine B-cell lymphosarcoma isolates. A nucleotide substitution was found in the glucocorticoid response element (GRE) site of the 5' long terminal repeat (5'LTR) of the BLV isolates. All amino acid substitutions in Tax previously found to be related to stimulate high transcriptional activity of 5'LTR were not found in these studies. Amino acid substitutions were found in the nucleocapsid, gp51 and G4 proteins. Premature stop-codons in R3 were observed. Few mutations or amino acid substitutions may be needed to allow BLV provirus to achieve silencing. Substitutions that favor suppression of viral expression in malignant B cells might be a strategy to circumvent effective immune attack.


Subject(s)
Enzootic Bovine Leukosis/virology , Genome, Viral , Leukemia Virus, Bovine/genetics , Lymphoma, B-Cell/veterinary , Amino Acid Sequence , Animals , Base Sequence , Cattle , Leukemia Virus, Bovine/chemistry , Leukemia Virus, Bovine/metabolism , Lymphoma, B-Cell/virology , Molecular Sequence Data , Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary
9.
Biochim Biophys Acta ; 1814(9): 1154-61, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21570497

ABSTRACT

Trans-sialidases are surface-located proteins in Trypanosoma cruzi that participate in key parasite-host interactions and parasite virulence. These proteins are encoded by a large multigenic family, with tandem-repeated and individual genes dispersed throughout the genome. While a large number of genes encode for catalytically active enzyme isoforms, many others display mutations that involve catalytic residues. The latter ultimately code for catalytically inactive proteins with very high similarity to their active paralogs. These inactive members have been shown to be lectins, able to bind sialic acid and galactose in vitro, although their cellular functions are yet to be fully established. We now report structural and biochemical evidence extending the current molecular understanding of these lectins. We have solved the crystal structure of one such catalytically inactive trans-sialidase-like protein, after soaking with a specific carbohydrate ligand, sialyl-α2,3-lactose. Instead of the expected trisaccharide, the binding pocket was observed occupied by α-lactose, strongly suggesting that the protein retains residual hydrolytic activity. This hypothesis was validated by enzyme kinetics assays, in comparison to fully active wild-type trans-sialidase. Surface plasmon resonance also confirmed that these trans-sialidase-like lectins are not only able to bind small oligosaccharides, but also sialylated glycoproteins, which is relevant in the physiologic scenario of parasite infection. Inactive trans-sialidase proteins appear thus to be ß-methyl-galactosyl-specific lectins, evolved within an exo-sialidase scaffold, thus explaining why their lectin activity is triggered by the presence of terminal sialic acid.


Subject(s)
Carbohydrates/chemistry , Glycoproteins/chemistry , Lectins/chemistry , Neuraminidase/chemistry , Trypanosoma cruzi/enzymology , Crystallography, X-Ray , Glycoproteins/physiology , Hydrolysis , Lactose/chemistry , Models, Molecular , Neuraminidase/physiology , Protein Structure, Tertiary , alpha-Fetoproteins/chemistry
10.
Sci Adv ; 7(23)2021 06.
Article in English | MEDLINE | ID: mdl-33958322

ABSTRACT

The emergence of SARS-CoV-2 antibody escape mutations highlights the urgent need for broadly neutralizing therapeutics. We previously identified a human monoclonal antibody, 47D11, capable of cross-neutralizing SARS-CoV-2 and SARS-CoV and protecting against the associated respiratory disease in an animal model. Here, we report cryo-EM structures of both trimeric spike ectodomains in complex with the 47D11 Fab. 47D11 binds to the closed receptor-binding domain, distal to the ACE2 binding site. The CDRL3 stabilizes the N343 glycan in an upright conformation, exposing a mutationally constrained hydrophobic pocket, into which the CDRH3 loop inserts two aromatic residues. 47D11 stabilizes a partially open conformation of the SARS-CoV-2 spike, suggesting that it could be used effectively in combination with other antibodies targeting the exposed receptor-binding motif. Together, these results reveal a cross-protective epitope on the SARS-CoV-2 spike and provide a structural roadmap for the development of 47D11 as a prophylactic or postexposure therapy for COVID-19.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , SARS-CoV-2 , Severe acute respiratory syndrome-related coronavirus , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Humans , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe acute respiratory syndrome-related coronavirus/immunology , SARS-CoV-2/chemistry , SARS-CoV-2/immunology , Structure-Activity Relationship
11.
Inorg Chem ; 49(22): 10668-79, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20964419

ABSTRACT

The aggregation of alpha-synuclein (AS) is a critical step in the etiology of Parkinson's disease (PD). A central, unresolved question in the pathophysiology of PD relates to the role of AS-metal interactions in amyloid fibril formation and neurodegeneration. Our previous works established a hierarchy in alpha-synuclein-metal ion interactions, where Cu(II) binds specifically to the protein and triggers its aggregation under conditions that might be relevant for the development of PD. Two independent, non-interacting copper-binding sites were identified at the N-terminal region of AS, with significant difference in their affinities for the metal ion. In this work we have solved unknown details related to the structural binding specificity and aggregation enhancement mediated by Cu(II). The high-resolution structural characterization of the highest affinity N-terminus AS-Cu(II) complex is reported here. Through the measurement of AS aggregation kinetics we proved conclusively that the copper-enhanced AS amyloid formation is a direct consequence of the formation of the AS-Cu(II) complex at the highest affinity binding site. The kinetic behavior was not influenced by the His residue at position 50, arguing against an active role for this residue in the structural and biological events involved in the mechanism of copper-mediated AS aggregation. These new findings are central to elucidate the mechanism through which the metal ion participates in the fibrillization of AS and represent relevant progress in the understanding of the bioinorganic chemistry of PD.


Subject(s)
Amyloid/chemistry , Copper/chemistry , Parkinson Disease , alpha-Synuclein/chemistry , Amyloid/metabolism , Binding Sites , Chemistry, Bioinorganic , Copper/metabolism , Humans , Models, Molecular , alpha-Synuclein/metabolism
12.
Arch Virol ; 155(4): 481-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20169372

ABSTRACT

Bovine leukaemia virus (BLV) is an oncogenic member of the genus Deltaretrovirus of the family Retroviridae. Recent studies revealed that BLV strains can be classified into six different genotypes and raised the possibility that another genotype may exist. In order to gain insight into the degree of genetic variability of BLV strains circulating in the South American region, a phylogenetic analysis was performed using gp51 env gene sequences. The results of these studies revealed the presence of seven BLV genotypes in this geographic region and the suitability of partial gp51 env gene sequences for phylogenetic inference. A significant number of amino acid substitutions found in BLV strains isolated in South America map to the second neutralization domain of gp51. A 3D molecular model of BLV gp51 revealed that these substitutions are located on the surface of the molecule. This may provide a selective advantage to overcome immune host neutralization.


Subject(s)
Enzootic Bovine Leukosis/virology , Leukemia Virus, Bovine/classification , Leukemia Virus, Bovine/isolation & purification , Polymorphism, Genetic , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Cattle , Cluster Analysis , Epitopes/genetics , Gene Products, env/genetics , Genotype , Leukemia Virus, Bovine/genetics , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology , South America
13.
Mol Microbiol ; 70(6): 1408-23, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19019160

ABSTRACT

Protein kinase G of Mycobacterium tuberculosis has been implicated in virulence and in regulation of glutamate metabolism. Here we show that this kinase undergoes a pattern of autophosphorylation that is distinct from that of other M. tuberculosis protein kinases characterized to date and we identify GarA as a substrate for phosphorylation by PknG. Autophosphorylation of PknG has little effect on kinase activity but promotes binding to GarA, an interaction that is also detected in living mycobacteria. PknG phosphorylates GarA at threonine 21, adjacent to the residue phosphorylated by PknB (T22), and these two phosphorylation events are mutually exclusive. Like the homologue OdhI from Corynebacterium glutamicum, the unphosphorylated form of GarA is shown to inhibit alpha-ketoglutarate decarboxylase in the TCA cycle. Additionally GarA is found to bind and modulate the activity of a large NAD(+)-specific glutamate dehydrogenase with an unusually low affinity for glutamate. Previous reports of a defect in glutamate metabolism caused by pknG deletion may thus be explained by the effect of unphosphorylated GarA on these two enzyme activities, which may also contribute to the attenuation of virulence.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Glutamic Acid/metabolism , Mycobacterium tuberculosis/enzymology , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Glutamate Dehydrogenase/metabolism , Molecular Sequence Data , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Phosphorylation
14.
Bioelectrochemistry ; 70(2): 394-400, 2007 May.
Article in English | MEDLINE | ID: mdl-16839825

ABSTRACT

The capability of adsorption of different electroactive cationic Re(V)-amine complexes onto myoglobin-containing electrodes has been investigated. The goal of this work was the development of an Au/thiol/myo electrode and, after incubation of such ensemble in the presence of three different Re(V)-amine complexes, the evaluation of the extent of surface coverage by the complexes (as a way to evaluate the interaction complex-protein) using electrochemical techniques. Our results showed that a protein-containing electrode could therefore be used for the detection of the interaction of small electroactive cationic complexes and the biomolecule. The extent of the coverage of the myoglobin electrode by the complex depends on the number of free tails from the ligands and the total charge of the complex.


Subject(s)
Biosensing Techniques/instrumentation , Cations/analysis , Cations/chemistry , Electrochemistry/instrumentation , Myoglobin/chemistry , Rhenium/analysis , Rhenium/chemistry , Adsorption , Biosensing Techniques/methods , Electrochemistry/methods , Electrodes , Macromolecular Substances/analysis , Macromolecular Substances/chemistry , Metals/chemistry
15.
Sci Rep ; 6: 37274, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853299

ABSTRACT

The "7 kDa DNA-binding" family, also known as the Sul7d family, is composed of chromatin proteins from the Sulfolobales archaeal order. Among them, Sac7d and Sso7d have been the focus of several studies with some characterization of their properties. Here, we studied eleven other proteins alongside Sac7d and Sso7d under the same conditions. The dissociation constants of the purified proteins for binding to double-stranded DNA (dsDNA) were determined in phosphate-buffered saline at 25 °C and were in the range from 11 µM to 22 µM with a preference for G/C rich sequences. In accordance with the extremophilic origin of their hosts, the proteins were found highly stable from pH 0 to pH 12 and at temperatures from 85.5 °C to 100 °C. Thus, these results validate eight putative "7 kDa DNA-binding" family proteins and show that they behave similarly regarding both their function and their stability among various genera and species. As Sac7d and Sso7d have found numerous uses as molecular biology reagents and artificial affinity proteins, this study also sheds light on even more attractive proteins that will facilitate engineering of novel highly robust reagents.


Subject(s)
Archaeal Proteins/chemistry , DNA, Archaeal/chemistry , DNA-Binding Proteins/chemistry , Sulfolobus/chemistry
16.
Elife ; 52016 12 12.
Article in English | MEDLINE | ID: mdl-27938660

ABSTRACT

Two-component systems (TCS) are protein machineries that enable cells to respond to input signals. Histidine kinases (HK) are the sensory component, transferring information toward downstream response regulators (RR). HKs transfer phosphoryl groups to their specific RRs, but also dephosphorylate them, overall ensuring proper signaling. The mechanisms by which HKs discriminate between such disparate directions, are yet unknown. We now disclose crystal structures of the HK:RR complex DesK:DesR from Bacillus subtilis, comprising snapshots of the phosphotransfer and the dephosphorylation reactions. The HK dictates the reactional outcome through conformational rearrangements that include the reactive histidine. The phosphotransfer center is asymmetric, poised for dissociative nucleophilic substitution. The structural bases of HK phosphatase/phosphotransferase control are uncovered, and the unexpected discovery of a dissociative reactional center, sheds light on the evolution of TCS phosphotransfer reversibility. Our findings should be applicable to a broad range of signaling systems and instrumental in synthetic TCS rewiring.


Subject(s)
Bacillus subtilis/enzymology , Histidine Kinase/chemistry , Histidine Kinase/metabolism , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/metabolism , Crystallography, X-Ray , Models, Molecular , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational
17.
J Inorg Biochem ; 99(3): 828-40, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15708805

ABSTRACT

myo-Inositol hexakisphosphate (InsP6) is an ubiquitous and abundant molecule in the cytosol and nucleus of eukaryotic cells whose biological functions are incompletely known. A major hurdle for studying the biology of InsP6 has been a deficiency of a full understanding of the chemistry of its interaction with divalent and trivalent cations. This deficiency has limited our appreciation of how it remains in solution within cells, and the likely degree to which it might interact in vivo with physiologically important cations such as Ca2+ and Fe3+. We report here the initial part of the description of the InsP6-multivalent cation chemistry, including its solution equilibria studied by high resolution potentiometry and (for the Fe(III)/Fe(II) couple) cyclic voltammetry. InsP6 forms anionic complexes of high affinities and 1:1 stoichiometry with Mg(II), Ca(II), Mn(II), Fe(II), Co(II), Ni(II), Cu(II), Zn(II) and Cd(II). Of particular importance is the observation that, in the exceptional case of Mg(II), InsP6 forms the species [Mg5(H2L)] (L representing fully deprotonated InsP6); this soluble neutral species is predicted to be the predominant form of InsP6 under nuclear or cytosolic conditions in animal cells. Contrary to previous suggestions, InsP6 is predicted not to interact with cytosolic calcium even when calcium is increased during signalling events. In vitro, InsP6 also forms high affinity 1:1 complexes with Fe(III) and Al(III). However, our data predict that in the biological context of excess free Mg(II), neither Fe(III) nor Fe(II) are complexed by InsP6.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Inositol Phosphates/chemistry , Magnesium/chemistry , Animals , Binding, Competitive , Calcium/chemistry , Cations , Electrochemistry , Hydrogen-Ion Concentration , Inositol Phosphates/metabolism , Iron/chemistry , Oxidation-Reduction , Predictive Value of Tests , Protons , Solutions
18.
Sci Rep ; 5: 8819, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25743628

ABSTRACT

The bacterial protein tyrosine phosphatase PtpA is a key virulence factor released by Mycobacterium tuberculosis in the cytosol of infected macrophages. So far only two unrelated macrophage components (VPS33B, GSK3α) have been identified as PtpA substrates. As tyrosine phosphatases are capable of using multiple substrates, we developed an improved methodology to pull down novel PtpA substrates from an enriched P-Y macrophage extract using the mutant PtpA D126A. This methodology reduced non-specific protein interactions allowing the identification of four novel putative PtpA substrates by MALDI-TOF-MS and nano LC-MS: three mitochondrial proteins - the trifunctional enzyme (TFP), the ATP synthase, and the sulfide quinone oxidoreductase - and the cytosolic 6-phosphofructokinase. All these proteins play a relevant role in cell energy metabolism. Using surface plasmon resonance, PtpA was found to bind immunopurified human TFP through its catalytic site since TFP-PtpA association was inhibited by a specific phosphatase inhibitor. Moreover, PtpA wt was capable of dephosphorylating immunopurified human TFP in vitro supporting that TFP may be a bona fide PtpA susbtrate. Overall, these results suggest a novel scenario where PtpA-mediated dephosphorylation may affect pathways involved in cell energy metabolism, particularly the beta oxidation of fatty acids through modulation of TFP activity and/or cell distribution.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Protein Tyrosine Phosphatases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line , Circular Dichroism , Humans , Macrophages/immunology , Mass Spectrometry , Mutation , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
19.
J Inorg Biochem ; 98(2): 238-44, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14729304

ABSTRACT

A reliable method based on electrochemical measurements for the evaluation of protein interaction with small electroactive coordination compounds is proposed. Protein binding capacity of five different cationic Re(V) complexes was evaluated by means of voltammetric techniques in the absence and the presence of bovine serum albumin. The percentage of interaction between the complex and the protein was estimated from the quantitative decrease of the diffusion coefficient for the sole complex due to the addition of the protein, and therefore the formation of the protein-complex molecular ensemble. The proposed methodology was checked using cisplatin as a molecular complex probe.


Subject(s)
Electrochemistry/methods , Proteins/metabolism , Rhenium/metabolism , Animals , Cattle , Cisplatin/metabolism , Rhenium/chemistry , Serum Albumin/metabolism , Serum Albumin/pharmacology
20.
Front Microbiol ; 5: 67, 2014.
Article in English | MEDLINE | ID: mdl-24616717

ABSTRACT

Recombinant protein expression has become an invaluable tool for academic and biotechnological projects. With the use of high-throughput screening technologies for soluble protein production, uncountable target proteins have been produced in a soluble and homogeneous state enabling the realization of further studies. Evaluation of hundreds conditions requires the use of high-throughput cloning and screening methods. Here we describe a new versatile vector suite dedicated to the expression improvement of recombinant proteins (RP) with solubility problems. This vector suite allows the parallel cloning of the same PCR product into the 12 different expression vectors evaluating protein expression under different promoter strength, different fusion tags as well as different solubility enhancer proteins. Additionally, we propose the use of a new fusion protein which appears to be a useful solubility enhancer. Above all we propose in this work an economic and useful vector suite to fast track the solubility of different RP. We also propose a new solubility enhancer protein that can be included in the evaluation of the expression of RP that are insoluble in classical expression conditions.

SELECTION OF CITATIONS
SEARCH DETAIL