Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Nanoscale ; 16(8): 4308-4316, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38353599

ABSTRACT

Iron-regulated surface determinant B (IsdB) is a surface protein of Staphylococcus aureus that plays essential roles in host cell invasion by mediating both bacterial adhesion and hemic iron acquisition. Single-molecule experiments have recently revealed that the binding of IsdB to vitronectin and integrins is dramatically strengthened under mechanical stress conditions, promoting staphylococcal adhesion. Here we conducted atomic force spectroscopy (AFS) measurements of the interaction between IsdB and hemoglobin (Hb), in both its oxidized (metHb) and reduced forms (HbCO). While the former represents the natural substrate for IsdB, the latter is resistant to heme extraction. For the unbinding between IsdB and HbCO, we obtained a linear trend in the Bell-Evans plot, indicative of a weakening of the interaction upon mechanical stress. For the unbinding between IsdB and metHb, we found similar behavior at low loading rates. Remarkably, a non-linear trend of the complex interaction force was detected at higher force-pulling rates. Such behavior may provide some cues to the ability of IsdB to form stress-dependent bonds also with Hb, possibly enabling a more efficient heme transfer through stabilization of the transient (in vivo) IsdB-Hb complex.


Subject(s)
Bacterial Proteins , Iron , Bacterial Proteins/metabolism , Iron/metabolism , Hemoglobins/chemistry , Heme/chemistry , Heme/metabolism , Membrane Proteins/metabolism , Protein Binding
2.
Biosensors (Basel) ; 14(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38391998

ABSTRACT

MicroRNAs are small ribonucleotides that act as key gene regulators. Their altered expression is often associated with the onset and progression of several human diseases, including cancer. Given their potential use as biomarkers, there is a need to find detection methods for microRNAs suitable for use in clinical setting. Field-effect-transistor-based biosensors (bioFETs) appear to be valid tools to detect microRNAs, since they may reliably quantitate the specific binding between the immobilized probe and free target in solution through an easily detectable electrical signal. We have investigated the detection of human microRNA 155 (miR-155) using an innovative capturing probe constituted by a synthetic peptide nucleic acid (PNA), which has the advantage to form a duplex even at ionic strengths approaching the physiological conditions. With the aim to develop an optimized BioFET setup, the interaction kinetics between miR-155 and the chosen PNA was preliminarily investigated by using surface plasmon resonance (SPR). By exploiting both these results and our custom-made bioFET system, we were able to attain a low-cost, real-time, label-free and highly specific detection of miR-155 in the nano-molar range.


Subject(s)
Biosensing Techniques , MicroRNAs , Nucleic Acids , Peptide Nucleic Acids , Humans , Surface Plasmon Resonance , Biosensing Techniques/methods , Peptides
3.
ACS Omega ; 8(42): 38941-38949, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37901511

ABSTRACT

miRNAs are short noncoding RNA single strands, with a crucial role in several biological processes. miRNAs are dysregulated in several human diseases, and their detection is an important goal for diagnosis and screening. Innovative biosensors for miRNAs are commonly based on the hybridization process between a miRNA and its corresponding complementary strand (or suitable aptamers) immobilized onto an electrode surface forming a duplex. A detailed description of the hybridization kinetics in working conditions deserves a great deal of interest for the optimization of the biosensing process. Surface plasmon resonance (SPR) and atomic force spectroscopy (AFS) were applied to investigate the hybridization process between miR-155, a multifunctional miRNA that constitutes an important marker overexpressed in several diseases, and its complementary strand (antimiR-155), immobilized on the gold-coated surface of a commercial electrode. Under well-adjusted pH, ionic strength, surface coverage, and concentration, we found that miR-155 has a high affinity for antimiR-155 with kinetics well described by the 1:1 Langmuir model. Both techniques provided an association rate of about 104 M-1 s-1, while a dissociation rate of 10-5 and 10-4 s-1 was assessed by SPR and AFS, respectively. These results allowed us to establish optimized measurement running times for applications in biosensing. An analysis of AFS data also led us to evaluate the binding free energy for the duplex, which was found to be close to that of free molecules in solution. These results could guide in the implementation of fine-tuned working conditions of a biosensor for detecting miRNAs based on correspondent complementary strands.

4.
Food Chem ; 404(Pt B): 134748, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36327502

ABSTRACT

Carotenoids play an important role in the stability, freshness, and nutritional value of extra-virgin olive oil (EVOO). However, the carotenoid content in EVOO changes over time as a function of olive ripening and degrading events. A reliable quality marker is the ratio between the two most abundant carotenoids, namely lutein and ß-carotene, since the second degrades more rapidly. Thus, to obtain a fast quantification of the lutein/ß-carotene ratio in olive oil could deserve a certain interest. Resonant Raman spectroscopy is a rapid and non-destructive technique, widely applied for food chemical characterization. In this work, using high-performance liquid chromatography and UV-vis absorption spectroscopy as calibration techniques, we present a reliable method to assess the lutein/ß-carotene ratio in EVOO using a single Raman spectrum. The novel approach deserves several methodological and applicative interests, since it would allow rapid, on-site screening of EVOO quality and authenticity, especially if implemented as a portable system.


Subject(s)
Lutein , beta Carotene , Olive Oil/chemistry , beta Carotene/analysis , Spectrum Analysis, Raman , Carotenoids/analysis
5.
Int J Mol Sci ; 23(20)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36293489

ABSTRACT

The conformational heterogeneity of the p53 tumor suppressor, the wild-type (p53wt) and mutated forms, was investigated by a computational approach, including the modeling and all atoms of the molecular dynamics (MD) simulations. Four different punctual mutations (p53R175H, p53R248Q, p53R273H, and p53R282W) which are known to affect the DNA binding and belong to the most frequent hot-spot mutations in human cancers, were taken into consideration. The MD trajectories of the wild-type and mutated p53 forms were analyzed by essential dynamics to extract the relevant collective motions and by the frustration method to evaluate the degeneracy of the energy landscape. We found that p53 is characterized by wide collective motions and its energy landscape exhibits a rather high frustration level, especially in the regions involved in the binding to physiological ligands. Punctual mutations give rise to a modulation of both the collective motions and the frustration of p53, with different effects depending on the mutation. The regions of p53wt and of the mutated forms characterized by a high frustration level are also largely involved in the collective motions. Such a correlation is discussed also in connection with the intrinsic disordered character of p53 and with its central functional role.


Subject(s)
Frustration , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Ligands , Mutation , Molecular Dynamics Simulation , DNA/genetics
6.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36142640

ABSTRACT

This study investigated the interaction between Human Serum Albumin (HSA) and microRNA 155 (miR-155) through spectroscopic, nanoscopic and computational methods. Atomic force spectroscopy together with static and time-resolved fluorescence demonstrated the formation of an HSA/miR-155 complex characterized by a moderate affinity constant (KA in the order of 104 M-1). Förster Resonance Energy Transfer (FRET) experiments allowed us to measure a distance of (3.9 ± 0.2) nm between the lone HSA Trp214 and an acceptor dye bound to miR-155 within such a complex. This structural parameter, combined with computational docking and binding free energy calculations, led us to identify two possible models for the structure of the complex, both characterized by a topography in which miR-155 is located within two positively charged pockets of HSA. These results align with the interaction found for HSA and miR-4749, reinforcing the thesis that native HSA is a suitable miRNA carrier under physiological conditions for delivering to appropriate targets.


Subject(s)
MicroRNAs , Serum Albumin, Human , Binding Sites , Circular Dichroism , Computer Simulation , Fluorescence Resonance Energy Transfer/methods , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Docking Simulation , Protein Binding , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Thermodynamics
7.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163220

ABSTRACT

The interaction of Human Serum Albumin (HSA) with the microRNA, miR4749, was investigated by Atomic Force Spectrscopy (AFS), static and time-resolved fluorescence spectroscopy and by computational methods. The formation of a HSA/miR4749 complex with an affinity of about 104 M-1 has been assessed through a Stern-Volmer analysis of steady-state fluorescence quenching of the lone Trp residue (Trp214) emission of HSA. Förster Resonance Energy Transfer (FRET) measurements of fluorescence lifetime of the HSA/miR4749 complex were carried out in the absence and in the presence of an acceptor chromophore linked to miR4749. This allowed us to determine a distance of 4.3 ± 0.5 nm between the lone Trp of HSA and the dye bound to miR4749 5p-end. Such a distance was exploited for a screening of the possible binding sites between HSA and miR4749, as predicted by computational docking. Such an approach, further refined by binding free energy calculations, led us to the identification of a consistent model for the structure of the HSA/miR4749 complex in which a positively charged HSA pocket accommodates the negatively charged miRNA molecule. These results designate native HSA as a suitable miRNA carrier under physiological conditions for delivering to appropriate targets.


Subject(s)
MicroRNAs/chemistry , MicroRNAs/genetics , Serum Albumin, Human/chemistry , Binding Sites/drug effects , Circular Dichroism/methods , Computational Biology/methods , Fluorescence , Fluorescence Resonance Energy Transfer/methods , Humans , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Protein Binding/physiology , Serum Albumin, Human/metabolism , Serum Albumin, Human/ultrastructure , Spectrometry, Fluorescence/methods , Thermodynamics
8.
Cancers (Basel) ; 13(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34885216

ABSTRACT

MicroRNAs (miRNAs) are linear single-stranded non-coding RNAs oligonucleotides, widely distributed in cells, playing a key role as regulators of gene expression at post-transcriptional level. Circular RNAs (circRNAs) are single-stranded RNA oligonucleotides forming a covalently closed continuous loop, which confers them a high structural stability and which may code for proteins or act as gene regulators. Abnormal levels or dysregulation of miRNA or circRNA are linked to several cancerous pathologies, so that they are receiving a large attention as diagnostic and prognostic tools. Some miRNAs and circRNAs are strongly involved in the regulatory networks of the transcription factor p53, which plays a pivotal role as tumor suppressor. Overexpression of miRNAs and/or circRNAs, as registered in a number of cancers, is associated to a concomitant inhibition of the p53 onco-suppressive function. Among other mechanisms, it was recently suggested that a functional inhibition of p53 could arise from a direct interaction between p53 and oncogenic miRNAs or circRNAs; a mechanism that might be reminiscent of the p53 inhibition by some E3 ubiquitin ligase such as MDM2 and COP1. Such evidence might deserve important implications for restoring the p53 anticancer functionality, and pave the way to intriguing perspectives for novel therapeutic strategies. In the present paper, the experimental evidence of the interaction between p53 and miRNAs and/or circRNAs is reviewed and discussed in connection with the development of new anticancer approaches.

9.
J Phys Chem B ; 125(36): 10166-10173, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34493041

ABSTRACT

Trp146 of the p53 DNA-binding domain (DBD) was investigated by static and time-resolved fluorescence combined with molecular dynamics (MD) simulations at different temperatures (25, 30, 37, and 45 °C). Static emission spectra exhibit an intensity maximum at 30 °C without any substantial peak shift, while the time-resolved fluorescence displays a peculiar stretched exponential decay, indicative of a structural disorder, at all of the investigated temperatures. The stretched exponential parameter was found to increase at 37 °C. An analysis of the MD simulation trajectories evidenced the occurrence of jumps in the temporal evolution of the distances between Trp146 and residues Arg110, Asp228, Cys229, and Gln144, which are mainly responsible for Trp146 fluorescence quenching. The times that these quenchers spend close to or far from Trp146 can provide an explanation for the static fluorescence behavior. Further essential dynamics analysis of the MD trajectories indicates a significant restriction of protein global motions above 37 °C. These results are consistent with a decrease in the structural heterogeneity of DBD as the temperature increases. The results are also discussed in view of understanding how temperature can modulate the p53 capability to binding partners, including DNA.


Subject(s)
DNA , Molecular Dynamics Simulation , Protein Domains , Spectrometry, Fluorescence , Temperature
10.
J Pept Sci ; 27(11): e3357, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34151482

ABSTRACT

The p28 peptide derived from Pseudomonas aeruginosa azurin shows an anticancer activity after binding to p53 protein and is currently in Phase I of clinical trials. We have studied its structure in water and in a biomimetic media and show that the peptide is unstructured in water but when studied in a biomimetic medium assumes a structure very similar to the one observed in azurin, suggesting a high propensity of this peptide to maintain this secondary structure. Analysis of p28 sequences from different bacterial species indicates conservation of the secondary structure despite amino acid replacement in different positions, suggesting that others, similar peptides could be tested for binding to p53.


Subject(s)
Antineoplastic Agents , Azurin , Antineoplastic Agents/pharmacology , Biomimetics , Peptide Fragments , Peptides , Pseudomonas aeruginosa
11.
Sensors (Basel) ; 20(24)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327383

ABSTRACT

The tumor suppressor p53 protein plays a crucial role in many biological processes. The presence of abnormal concentrations of wild-type p53, or some of its mutants, can be indicative of a pathological cancer state. p53 represents therefore a valuable biomarker for tumor screening approaches and development of suitable biosensors for its detection deserves a high interest in early diagnostics. Here, we revisit our experimental approaches, combining Surface Enhanced Raman Spectroscopy (SERS) and nanotechnological materials, for ultrasensitive detection of wild-type and mutated p53, in the perspective to develop biosensors to be used in clinical diagnostics. The Raman marker is provided by a small molecule (4-ATP) acting as a bridge between gold nanoparticles (NPs) and a protein biomolecule. The Azurin copper protein and specific antibodies of p53 were used as a capture element for p53 (wild-type and its mutants). The developed approaches allowed us to reach a detection level of p53 down to 10-17 M in both buffer and serum. The implementation of the method in a biosensor device, together with some possible developments are discussed.


Subject(s)
Metal Nanoparticles , Neoplasms , Spectrum Analysis, Raman , Gold , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
12.
Sensors (Basel) ; 20(21)2020 Nov 08.
Article in English | MEDLINE | ID: mdl-33171594

ABSTRACT

The concentration of wild-type tumour suppressor p53wt in cells and blood has a clinical significance for early diagnosis of some types of cancer. We developed a disposable, label-free, field-effect transistor-based immunosensor (BioFET), able to detect p53wt in physiological buffer solutions, over a wide concentration range. Microfabricated, high-purity gold electrodes were used as single-use extended gates (EG), which avoid direct interaction between the transistor gate and the biological solution. Debye screening, which normally hampers target charge effect on the FET gate potential and, consequently, on the registered FET drain-source current, at physiological ionic strength, was overcome by incorporating a biomolecule-permeable polymer layer on the EG electrode surface. Determination of an unknown p53wt concentration was obtained by calibrating the variation of the FET threshold voltage versus the target molecule concentration in buffer solution, with a sensitivity of 1.5 ± 0.2 mV/decade. The BioFET specificity was assessed by control experiments with proteins that may unspecifically bind at the EG surface, while 100pM p53wt concentration was established as limit of detection. This work paves the way for fast and highly sensitive tools for p53wt detection in physiological fluids, which deserve much interest in early cancer diagnosis and prognosis.


Subject(s)
Biosensing Techniques , Immunoassay , Tumor Suppressor Protein p53/analysis , Buffers , Electrodes , Gold , Humans , Transistors, Electronic
13.
J Phys Chem B ; 124(44): 9820-9828, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33103427

ABSTRACT

Time-resolved fluorescence emission was combined with molecular dynamics (MD) simulations to investigate the DNA-binding domain (DBD) of the tumor suppressor p53 alone and its complex with the anticancer peptide p28 (DBD/p28). The fluorescence emission decay of the lone Trp residue, from both DBD and DBD/p28, was well-described by a stretched exponential function. Such a behavior was ascribed to heterogeneity in the Trp relaxation behavior, likely due to the coexistence of different conformational states. The increase of the stretching parameter, on passing from DBD to DBD/p28, indicates a reduced heterogeneity in the Trp146 environment for DBD/p28. Moreover, the effects of p28 on the global dynamics of DBD were analyzed by the essential dynamics method on 30 ns long MD trajectories of both DBD and DBD/p28. We found the establishment of wide-amplitude anharmonic modes throughout the DBD molecule, with a particularly high amplitude being detected in the DNA-binding region. These modes are significantly reduced when DBD is bound to p28, consistently with a structure stabilization. In summary, the results indicate that p28 binding has a strong effect on both the local and global heterogeneity of DBD, thus providing some hints to the understanding of its anticancer activity.


Subject(s)
Molecular Dynamics Simulation , Peptides , Peptide Fragments , Protein Domains
14.
Biomolecules ; 10(2)2020 02 22.
Article in English | MEDLINE | ID: mdl-32098369

ABSTRACT

The interactions between the DNA binding domain (DBD) of the tumor suppressor p53 and miR4749, characterized by a high sequence similarity with the DNA Response Element (RE) of p53, was investigated by fluorescence spectroscopy combined with computational modeling and docking. Fluorescence quenching experiments witnessed the formation of a specific complex between DBD and miR4749 with an affinity of about 105 M. Förster Resonance Energy Transfer (FRET) allowed us to measure a distance of 3.9 ± 0.3 nm, between the lone tryptophan of DBD and an acceptor dye suitably bound to miR4749. Such information, combined with a computational modeling approach, allowed us to predict possible structures for the DBD-miR4749 complex. A successive docking refinement, complemented with binding free energy calculations, led us to single out a best model for the DBD-miR4749 complex. We found that the interaction of miR4749 involves the DBD L3 loop and the H1 helix, close to the Zn-finger motif; with this suggesting that miR4749 could directly inhibit the p53 interaction with DNA. These results might inspire new therapeutic strategies finalized to restore the p53 functional activity.


Subject(s)
MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , DNA/chemistry , DNA/genetics , Fluorescence , Fluorescence Resonance Energy Transfer/methods , Humans , MicroRNAs/genetics , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Spectrometry, Fluorescence/methods , Tumor Suppressor Protein p53/genetics
15.
Sci Rep ; 9(1): 18629, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819099

ABSTRACT

Among multidrug-resistant bacteria, methicillin-resistant Staphylococcus aureus is emerging as one of the most threatening pathogens. S. aureus exploits different mechanisms for its iron supply, but the preferred one is acquisition of organic iron through the expression of hemoglobin (Hb) receptors. One of these, IsdB, belonging to the Isd (Iron-Regulated Surface Determinant) system, was shown to be essential for bacterial growth and virulence. Therefore, interaction of IsdB with Hb represents a promising target for the rational design of a new class of antibacterial molecules. However, despite recent investigations, many structural and mechanistic details of complex formation and heme extraction process are still elusive. By combining site-directed mutagenesis, absorption spectroscopy, surface plasmon resonance and molecular dynamics simulations, we tackled most of the so far unanswered questions: (i) the exact complex stoichiometry, (ii) the microscopic kinetic rates of complex formation, (iii) the IsdB selectivity for binding to, and extracting heme from, α and ß subunits of Hb, iv) the role of specific amino acid residues and structural regions in driving complex formation and heme transfer, and (v) the structural/dynamic effect played by the hemophore on Hb.


Subject(s)
Cation Transport Proteins/genetics , Hemoglobins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/genetics , Drug Resistance, Multiple/genetics , Heme/genetics , Humans , Iron/metabolism , Kinetics , Mutagenesis, Site-Directed , Staphylococcal Infections/microbiology
16.
Int J Mol Sci ; 20(12)2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31238511

ABSTRACT

Raman spectroscopy, which is a suitable tool to elucidate the structural properties of intrinsically disordered proteins, was applied to investigate the changes in both the structure and the conformational heterogeneity of the DNA-binding domain (DBD) belonging to the intrinsically disordered protein p53 upon its binding to Azurin, an electron-transfer anticancer protein from Pseudomonas aeruginosa. The Raman spectra of the DBD and Azurin, isolated in solution or forming a complex, were analyzed by a combined analysis based on peak inspection, band convolution, and principal component analysis (PCA). In particular, our attention was focused on the Raman peaks of Tyrosine and Tryptophan residues, which are diagnostic markers of protein side chain environment, and on the Amide I band, of which the deconvolution allows us to extract information about α-helix, ß-sheet, and random coil contents. The results show an increase of the secondary structure content of DBD concomitantly with a decrease of its conformational heterogeneity upon its binding to Azurin. These findings suggest an Azurin-induced conformational change of DBD structure with possible implications for p53 functionality.


Subject(s)
Azurin/chemistry , DNA/chemistry , Protein Interaction Domains and Motifs , Spectrum Analysis, Raman , Tumor Suppressor Protein p53/chemistry , Azurin/metabolism , Binding Sites , DNA/metabolism , Humans , Models, Molecular , Molecular Conformation , Protein Binding , Tumor Suppressor Protein p53/metabolism
17.
Arch Biochem Biophys ; 671: 35-41, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31181181

ABSTRACT

miRNA-21-3p is overexpressed in a number of cancers and contributes to their development with a concomitant inhibition of the p53 onco-suppressive function. While a direct interaction of p53 with some miRNA precursors (namely pri-miRNAs and pre-miRNAs) was found, no interaction with mature micro RNA has been so far evidenced. It could therefore be very interesting to investigate if a direct interaction of miR-21-3p and p53 is occurring with possible impairment of the p53 onco-suppressive function. Fluorescence and Atomic Force Spectroscopy (AFS) were applied to study the interaction of p53 DNA Binding Domain (DBD) and miRNA-21-3p. Förster resonance energy transfer (FRET) was used to measure the distance between the DBD lone tryptophan (FRET donor) and a dye (FRET acceptor) bound to miRNA-21-3p. AFS and Fluorescence evidenced a direct interaction between miRNA-21-3p and DBD; with the formed complex being characterized by an affinity of 105 M, with a lifetime in the order of seconds. FRET allowed to determine an average distance of 4.0 nm between the DBD lone Trp146 and miRNA-21-3p; consistently with the involvement of the DBD L3 loop and/or the H1 helix in the complex formation, directly involved in the oligomerization and DNA binding. This may suggest that a functional inhibition of p53 could arise from its interaction with the oncogenic miRNA. Evidence of DBD-miRNA-21-3p complex formation may deserve some interest for inspiring novel therapeutic strategies.


Subject(s)
MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , Fluorescence , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Humans , Protein Binding , Protein Domains , Spectrum Analysis , Tryptophan/chemistry , Tumor Suppressor Protein p53/chemistry
18.
Biochim Biophys Acta Gen Subj ; 1863(2): 342-350, 2019 02.
Article in English | MEDLINE | ID: mdl-30419285

ABSTRACT

BACKGROUND: The p28 peptide, derived from the blue copper protein Azurin, exerts an anticancer action due to interaction with the tumor suppressor p53, likely interfering with its down-regulators. Knowledge of both the kinetics and topological details of the interaction, could greatly help to understand the peptide anticancer mechanism. METHODS: Fluorescence and Förster resonance energy transfer (FRET) were used to determine both the binding affinity and the distance between the lone tryptophan (FRET donor) of DNA Binding Domain (DBD) of p53 and the Iaedens dye (FRET acceptor) bound to the p28 peptide. Docking, Molecular Dynamic simulations and free energy binding calculations were used to single out the best complex model, compatible with the distance measured by FRET. RESULTS: Tryptophan fluorescence quenching provided a 105 M-1 binding affinity for the complex. Both FRET donor fluorescence quenching and acceptor enhancement are consistent with a donor-acceptor distance of about 2.6 nm. Docking and molecular dynamics simulations allowed us to select the best complex, enlightening the contact regions between p28 and DBD. CONCLUSIONS: p28 binds to DBD partially engaging the L1 loop, at the same region of the p53 down-regulator COP1, leaving however the DNA binding site available for functional interactions. GENERAL SIGNIFICANCE: Elucidation of the DBD-p28 complex gets insights into the functional role of p28 in regulating the p53 anticancer activity, also offering new perspectives to design new drugs able to protect the p53 anticancer function.


Subject(s)
Antineoplastic Agents/chemistry , Cell-Penetrating Peptides/chemistry , DNA/chemistry , Fluorescence Resonance Energy Transfer , Molecular Docking Simulation , Molecular Dynamics Simulation , Tumor Suppressor Protein p53/chemistry , Antineoplastic Agents/metabolism , Cell-Penetrating Peptides/metabolism , DNA/metabolism , Fluorescence , Humans , Tryptophan/chemistry , Tryptophan/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Anal Chim Acta ; 1029: 86-96, 2018 Oct 31.
Article in English | MEDLINE | ID: mdl-29907296

ABSTRACT

p53 is a powerful transcription factor playing a pivotal role in the prevention of cancer development and in maintaining genome integrity. This oncosuppressor is found to be functionally inactivated by mutations in many human tumors. Accordingly, wild type p53 and its oncogenic mutants represent valuable cancer biomarkers for diagnostic and prognostic purposes. We developed a highly sensitive biosensor, based on Surface Enhanced Raman Spectroscopy, for detection of wild type p53 and of p53R175H, which is one of the most frequent tumor-associated mutants of p53. Our approach combines the huge Raman signal enhancement, mainly arising from the plasmonic resonance effect on molecules close to gold nanoparticles, with the antigen-antibody biorecognition specificity. By following the enhanced signal of a specific Raman marker, intrinsic to the nanoparticle-antibody bioconjugation, we were able to push the antigen detection level down to the attomolar range in buffer and to the femtomolar range in spiked human serum. The method demonstrated a high reproducibility and a remarkable selectivity in discriminating between wild type p53 and p53R175H mutant, in both buffer and serum. A calibration plot was built and validated by ELISA for a reliable quantification of p53. These findings entitle our SERS-based immunosensor as a powerful and reliable tool for a non-invasive screening in human serum targeting p53 network. The approach could be easily extended to ultrasensitive detection of other markers of general interest, with feasible implementations into multiplex assays, functioning as lab-on-chip devices for several applications.


Subject(s)
Biosensing Techniques/methods , Immunoassay/methods , Limit of Detection , Mutant Proteins/analysis , Mutation , Spectrum Analysis, Raman , Tumor Suppressor Protein p53/analysis , Gold/chemistry , Humans , Mutant Proteins/blood , Mutant Proteins/genetics , Tumor Suppressor Protein p53/blood , Tumor Suppressor Protein p53/genetics
20.
Int J Nanomedicine ; 13: 251-259, 2018.
Article in English | MEDLINE | ID: mdl-29379285

ABSTRACT

p53 plays an important role in the safeguard of the genome but it is frequently downregulated mainly by E3 ubiquitin ligases among which COP1 plays an important role. The overexpression of COP1 has been reported to occur in several tumors and may be indicative of its overall oncogenic effect, which in turn might be originated by a direct interaction of COP1 with p53. Such an interaction may constitute a rewarding target for anticancer drug design strategies; therefore, a deeper understanding of its underlying molecular mechanism and kinetics is needed. The formation of a single p53-COP1 bimolecular complex was visualized by atomic force microscopy imaging on a mica substrate. The kinetic characterization of the complex, performed by atomic force spectroscopy and surface plasmon resonance, provided a KD value of ∼10-8 M and a relative long lifetime in the order of minutes, both at the single-molecule level and in bulk solution. The surprisingly high affinity value and low dissociation rate of the p53-COP1 bimolecular complex, which is even stronger than the p53-MDM2 complex, should be considered a benchmark for designing, development and optimization of suitable drugs able to antagonize the complex formation with the aim of preventing the inhibitory effect of COP1 on the p53 oncosuppressive function.


Subject(s)
Microscopy, Atomic Force/methods , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Humans , Kinetics , Microscopy, Atomic Force/instrumentation , Molecular Imaging/methods , Multiprotein Complexes/analysis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Surface Plasmon Resonance/methods , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...