Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters










Publication year range
1.
Q Rev Biophys ; 57: e4, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38597675

ABSTRACT

Solving the mechanism of a chemical reaction requires determining the structures of all the ground states on the pathway and the elusive transition states linking them. 2024 is the centenary of Brønsted's landmark paper that introduced the ß-value and structure-activity studies as the only experimental means to infer the structures of transition states. It involves making systematic small changes in the covalent structure of the reactants and analysing changes in activation and equilibrium-free energies. Protein engineering was introduced for an analogous procedure, Φ-value analysis, to analyse the noncovalent interactions in proteins central to biological chemistry. The methodology was developed first by analysing noncovalent interactions in transition states in enzyme catalysis. The mature procedure was then applied to study transition states in the pathway of protein folding - 'part (b) of the protein folding problem'. This review describes the development of Φ-value analysis of transition states and compares and contrasts the interpretation of ß- and Φ-values and their limitations. Φ-analysis afforded the first description of transition states in protein folding at the level of individual residues. It revealed the nucleation-condensation folding mechanism of protein domains with the transition state as an expanded, distorted native structure, containing little fully formed secondary structure but many weak tertiary interactions. A spectrum of transition states with various degrees of structural polarisation was then uncovered that spanned from nucleation-condensation to the framework mechanism of fully formed secondary structure. Φ-analysis revealed how movement of the expanded transition state on an energy landscape accommodates the transition from framework to nucleation-condensation mechanisms with a malleability of structure as a unifying feature of folding mechanisms. Such movement follows the rubric of analysis of classical covalent chemical mechanisms that began with Brønsted. Φ-values are used to benchmark computer simulation, and Φ and simulation combine to describe folding pathways at atomic resolution.


Subject(s)
Protein Folding , Proteins , Computer Simulation , Proteins/chemistry , Protein Engineering , Biology , Kinetics , Thermodynamics
2.
J Mol Biol ; 433(20): 167088, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34087198

ABSTRACT

I outline how over my career as a protein scientist Machine Learning has impacted my area of science and one of my pastimes, chess, where there are some interesting parallels. In 1968, modelling of three-dimensional structures was initiated based on a known structure as a template, the problem of the pathway of protein folding was posed and bets were taken in the emerging field of Machine Learning on whether computers could outplay humans at chess. Half a century later, Machine Learning has progressed from using computational power combined with human knowledge in solving problems to playing chess without human knowledge being used, where it has produced novel strategies. Protein structures are being solved by Machine Learning based on human-derived knowledge but without templates. There is much promise that programs like AlphaFold based on Machine Learning will be powerful tools for designing entirely novel protein folds and new activities. But, will they produce novel ideas on protein folding pathways and provide new insights into the principles that govern folds?


Subject(s)
Machine Learning , Protein Folding , Proteins/chemistry , Animals , Chymotrypsin/chemistry , Humans , Models, Molecular , Protein Conformation , Software , Trypsin/chemistry
3.
Protein Sci ; 30(9): 1818-1832, 2021 09.
Article in English | MEDLINE | ID: mdl-34089216

ABSTRACT

The Rel proteins of the NF-κB complex comprise one of the most investigated transcription factor families, forming a variety of hetero- or homodimers. Nevertheless, very little is known about the fundamental kinetics of NF-κB complex assembly, or the inter-conversion potential of dimerised Rel subunits. Here, we examined an unexplored aspect of NF-κB dynamics, focusing on the dissociation and reassociation of the canonical p50 and p65 Rel subunits and their ability to form new hetero- or homodimers. We employed a soluble expression system to enable the facile production of NF-κB Rel subunits, and verified these proteins display canonical NF-κB nucleic acid binding properties. Using a combination of biophysical techniques, we demonstrated that, at physiological temperatures, homodimeric Rel complexes routinely exchange subunits with a half-life of less than 10 min. In contrast, we found a dramatic preference for the formation of the p50/p65 heterodimer, which demonstrated a kinetic stability of at least an order of magnitude greater than either homodimer. These results suggest that specific DNA targets of either the p50 or p65 homodimers can only be targeted when these subunits are expressed exclusively, or with the intervention of additional post-translational modifications. Together, this work implies a new model of how cells can modulate NF-κB activity by fine-tuning the relative proportions of the p50 and p65 proteins, as well as their time of expression. This work thus provides a new quantitative interpretation of Rel dimer distribution in the cell, particularly for those who are developing mathematical models of NF-κB activity.


Subject(s)
DNA/chemistry , NF-kappa B p50 Subunit/chemistry , Oligodeoxyribonucleotides/chemistry , Protein Subunits/chemistry , Transcription Factor RelA/chemistry , Binding Sites , Cloning, Molecular , DNA/genetics , DNA/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Models, Molecular , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Oligodeoxyribonucleotides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
4.
ACS Chem Biol ; 15(3): 657-668, 2020 03 20.
Article in English | MEDLINE | ID: mdl-31990523

ABSTRACT

We have previously shown that the thermolabile, cavity-creating p53 cancer mutant Y220C can be reactivated by small-molecule stabilizers. In our ongoing efforts to unearth druggable variants of the p53 mutome, we have now analyzed the effects of other cancer-associated mutations at codon 220 on the structure, stability, and dynamics of the p53 DNA-binding domain (DBD). We found that the oncogenic Y220H, Y220N, and Y220S mutations are also highly destabilizing, suggesting that they are largely unfolded under physiological conditions. A high-resolution crystal structure of the Y220S mutant DBD revealed a mutation-induced surface crevice similar to that of Y220C, whereas the corresponding pocket's accessibility to small molecules was blocked in the structure of the Y220H mutant. Accordingly, a series of carbazole-based small molecules, designed for stabilizing the Y220C mutant, also bound to and stabilized the folded state of the Y220S mutant, albeit with varying affinities due to structural differences in the binding pocket of the two mutants. Some of the compounds also bound to and stabilized the Y220N mutant, but not the Y220H mutant. Our data validate the Y220S and Y220N mutants as druggable targets and provide a framework for the design of Y220S or Y220N-specific compounds as well as compounds with dual Y220C/Y220S specificity for use in personalized cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Carbazoles/chemistry , Mutant Proteins/chemistry , Mutant Proteins/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Crystallization , Drug Screening Assays, Antitumor , Gene Expression Regulation/drug effects , Humans , Models, Molecular , Mutation , Protein Binding , Protein Domains , Protein Stability/drug effects , Structure-Activity Relationship
5.
Chembiochem ; 21(1-2): 5-6, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31769931

ABSTRACT

Looking back, looking forward: In 2000, ChemBioChem debuted. The chemistry of carbohydrates, nucleic acids, peptides, proteins, natural products and other small molecules had reached a level that allowed biological questions to be probed. Today, there is no end in sight to studying biological matter with chemical tools or making use of biological methods to produce chemicals.


Subject(s)
Biological Products/metabolism , Carbohydrates/chemistry , Nucleic Acids/metabolism , Peptides/metabolism , Proteins/metabolism , Biological Products/chemistry , Humans , Nucleic Acids/chemistry , Peptides/chemistry , Proteins/chemistry , Synthetic Biology
6.
Future Med Chem ; 11(19): 2491-2504, 2019 10.
Article in English | MEDLINE | ID: mdl-31633398

ABSTRACT

Aim: The p53 cancer mutation Y220C creates a conformationally unstable protein with a unique elongated surface crevice that can be targeted by molecular chaperones. We report the structure-guided optimization of the carbazole-based stabilizer PK083. Materials & methods: Biophysical, cellular and x-ray crystallographic techniques have been employed to elucidate the mode of action of the carbazole scaffolds. Results: Targeting an unoccupied subsite of the surface crevice with heterocycle-substituted PK083 analogs resulted in a 70-fold affinity increase to single-digit micromolar levels, increased thermal stability and decreased rate of aggregation of the mutant protein. PK9318, one of the most potent binders, restored p53 signaling in the liver cancer cell line HUH-7 with homozygous Y220C mutation. Conclusion: The p53-Y220C mutant is an excellent paradigm for the development of mutant p53 rescue drugs via protein stabilization. Similar rescue strategies may be applicable to other cavity-creating p53 cancer mutations.


Subject(s)
Carbazoles/pharmacology , Molecular Chaperones/metabolism , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Carbazoles/chemical synthesis , Carbazoles/chemistry , Humans , Molecular Chaperones/chemical synthesis , Molecular Chaperones/chemistry , Molecular Structure , Tumor Cells, Cultured , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism
7.
Cancers (Basel) ; 11(8)2019 Aug 10.
Article in English | MEDLINE | ID: mdl-31405179

ABSTRACT

Half of human cancers harbor TP53 mutations that render p53 inactive as a tumor suppressor. In these cancers, reactivation of mutant p53 (mutp53) through restoration of wild-type-like function constitutes a valuable anticancer therapeutic strategy. In order to search for mutp53 reactivators, a small library of tryptophanol-derived oxazoloisoindolinones was synthesized and the potential of these compounds as mutp53 reactivators and anticancer agents was investigated in human tumor cells and xenograft mouse models. By analysis of their anti-proliferative effect on a panel of p53-null NCI-H1299 tumor cells ectopically expressing highly prevalent mutp53, the compound SLMP53-2 was selected based on its potential reactivation of multiple structural mutp53. In mutp53-Y220C-expressing hepatocellular carcinoma (HCC) cells, SLMP53-2-induced growth inhibition was mediated by cell cycle arrest, apoptosis, and endoplasmic reticulum stress response. In these cells, SLMP53-2 restored wild-type-like conformation and DNA-binding ability of mutp53-Y220C by enhancing its interaction with the heat shock protein 70 (Hsp70), leading to the reestablishment of p53 transcriptional activity. Additionally, SLMP53-2 displayed synergistic effect with sorafenib, the only approved therapy for advanced HCC. Notably, it exhibited potent antitumor activity in human HCC xenograft mouse models with a favorable toxicological profile. Collectively, SLMP53-2 is a new mutp53-targeting agent with promising antitumor activity, particularly against HCC.

8.
Eur J Med Chem ; 152: 101-114, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29702446

ABSTRACT

Many cancers have the tumor suppressor p53 inactivated by mutation, making reactivation of mutant p53 with small molecules a promising strategy for the development of novel anticancer therapeutics. The oncogenic p53 mutation Y220C, which accounts for approximately 100,000 cancer cases per year, creates an extended surface crevice in the DNA-binding domain, which destabilizes p53 and causes denaturation and aggregation. Here, we describe the structure-guided design of a novel class of small-molecule Y220C stabilizers and the challenging synthetic routes developed in the process. The synthesized chemical probe MB710, an aminobenzothiazole derivative, binds tightly to the Y220C pocket and stabilizes p53-Y220C in vitro. MB725, an ethylamide analogue of MB710, induced selective viability reduction in several p53-Y220C cancer cell lines while being well tolerated in control cell lines. Reduction of viability correlated with increased and selective transcription of p53 target genes such as BTG2, p21, PUMA, FAS, TNF, and TNFRSF10B, which promote apoptosis and cell cycle arrest, suggesting compound-mediated transcriptional activation of the Y220C mutant. Our data provide a framework for the development of a class of potent, non-toxic compounds for reactivating the Y220C mutant in anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Structure-Activity Relationship , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
9.
J Biol Chem ; 293(12): 4262-4276, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29382728

ABSTRACT

p53 is an important tumor-suppressor protein that is mutated in more than 50% of cancers. Strategies for restoring normal p53 function are complicated by the oncogenic properties of mutant p53 and have not met with clinical success. To counteract mutant p53 activity, a variety of drugs with the potential to reconvert mutant p53 to an active wildtype form have been developed. However, these drugs are associated with various negative effects such as cellular toxicity, nonspecific binding to other proteins, and inability to induce a wildtype p53 response in cancer tissue. Here, we report on the effects of a curcumin analog, HO-3867, on p53 activity in cancer cells from different origins. We found that HO-3867 covalently binds to mutant p53, initiates a wildtype p53-like anticancer genetic response, is exclusively cytotoxic toward cancer cells, and exhibits high anticancer efficacy in tumor models. In conclusion, HO-3867 is a p53 mutant-reactivating drug with high clinical anticancer potential.


Subject(s)
Apoptosis/drug effects , Curcumin/analogs & derivatives , Mutant Proteins/genetics , Mutation , Neoplasms/pathology , Piperidones/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Curcumin/pharmacology , Female , Humans , Mice , Mice, Nude , Mutant Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
10.
Cancer Lett ; 414: 99-106, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29069577

ABSTRACT

The identification of a targeted therapy for patients with triple-negative breast cancer (TNBC) is one of the most urgent needs in breast cancer therapeutics. The p53 gene is mutated in approximately 80% of patients with TNBC, and is a potential therapeutic target for patients with this form of breast cancer. The 2-sulfonylpyrimidine compound, PK11007, preferentially decreases viability in p53-compromised cancer cell lines. We investigated PK11007 as a potential new treatment for TNBC. IC50 values for inhibition of proliferation in a panel of 17 breast cell lines by PK11007 ranged from 2.3 to 42.2 µM. There were significantly lower IC50 values for TNBC than for non-TNBC cell lines (p = 0.03) and for p53-mutated cell lines compared with p53 WT cells (p = 0.003). Response to PK11007 however, was independent of the estrogen receptor (ER) or HER2 status of the cell lines. In addition to inhibiting cell proliferation, PK11007 induced apoptosis in p53 mutant cell lines. Using RNAseq and gene ontology analysis, we found that PK11007 altered the expression of genes enriched in pathways involved in regulated cell death, regulation of apoptosis, signal transduction, protein refolding and locomotion. The observations that PK11007 inhibited cell proliferation, induced apoptosis and altered genes involved in cell death are all consistent with the ability of PK11007 to reactivate mutant p53. Based on our data, we conclude that targeting mutant p53 with PK11007 is a potential approach for treating p53-mutated breast cancer, including the subgroup with TN disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Mutant Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Humans , MCF-7 Cells , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pyrimidines/pharmacology , Sulfones/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Sci Adv ; 3(5): e1700933, 2017 May.
Article in English | MEDLINE | ID: mdl-28508083

ABSTRACT

ATM (ataxia-telangiectasia mutated) is a phosphatidylinositol 3-kinase-related protein kinase (PIKK) best known for its role in DNA damage response. ATM also functions in oxidative stress response, insulin signaling, and neurogenesis. Our electron cryomicroscopy (cryo-EM) suggests that human ATM is in a dynamic equilibrium between closed and open dimers. In the closed state, the PIKK regulatory domain blocks the peptide substrate-binding site, suggesting that this conformation may represent an inactive or basally active enzyme. The active site is held in this closed conformation by interaction with a long helical hairpin in the TRD3 (tetratricopeptide repeats domain 3) domain of the symmetry-related molecule. The open dimer has two protomers with only a limited contact interface, and it lacks the intermolecular interactions that block the peptide-binding site in the closed dimer. This suggests that the open conformation may be more active. The ATM structure shows the detailed topology of the regulator-interacting N-terminal helical solenoid. The ATM conformational dynamics shown by the structures represent an important step in understanding the enzyme regulation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/chemistry , Protein Multimerization , Cryoelectron Microscopy , Humans , Protein Domains , Protein Structure, Quaternary
12.
Proc Natl Acad Sci U S A ; 114(13): E2634-E2643, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28292898

ABSTRACT

Protein aggregation is involved in many diseases. Often, a unique aggregation-prone sequence polymerizes to form regular fibrils. Many oncogenic mutants of the tumor suppressor p53 rapidly aggregate but form amorphous fibrils. A peptide surrounding Ile254 is proposed to be the aggregation-driving sequence in cells. We identified several different aggregating sites from limited proteolysis of harvested aggregates and effects of mutations on kinetics and products of aggregation. We present a model whereby the amorphous nature of the aggregates results from multisite branching of polymerization after slow unfolding of the protein, which may be a common feature of aggregation of large proteins. Greatly lowering the aggregation propensity of any one single site, including the site of Ile254, by mutation did not inhibit aggregation in vitro because aggregation could still occur via the other sites. Inhibition of an individual site is, accordingly, potentially unable to prevent aggregation in vivo. However, cancer cells are specifically killed by peptides designed to inhibit the Ile254 sequence and further aggregation-driving sequences that we have found. Consistent with our proposed mechanism of aggregation, we found that such peptides did not inhibit aggregation of mutant p53 in vitro. The cytotoxicity was not eliminated by knockdown of p53 in 2D cancer cell cultures. The peptides caused rapid cell death, much faster than usually expected for p53-mediated transcription-dependent apoptosis. There may also be non-p53 targets for those peptides in cancer cells, such as p63, or the peptides may alter other interactions of partly denatured p53 with receptors.


Subject(s)
Protein Aggregation, Pathological , Tumor Suppressor Protein p53/metabolism , Humans , Models, Theoretical , Mutation , Neoplasms/genetics , Protein Domains , Tumor Suppressor Protein p53/chemistry
13.
Proc Natl Acad Sci U S A ; 113(47): E7456-E7463, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821763

ABSTRACT

Despite the recent rapid progress in cryo-electron microscopy (cryo-EM), there still exist ample opportunities for improvement in sample preparation. Macromolecular complexes may disassociate or adopt nonrandom orientations against the extended air-water interface that exists for a short time before the sample is frozen. We designed a hollow support structure using 3D DNA origami to protect complexes from the detrimental effects of cryo-EM sample preparation. For a first proof-of-principle, we concentrated on the transcription factor p53, which binds to specific DNA sequences on double-stranded DNA. The support structures spontaneously form monolayers of preoriented particles in a thin film of water, and offer advantages in particle picking and sorting. By controlling the position of the binding sequence on a single helix that spans the hollow support structure, we also sought to control the orientation of individual p53 complexes. Although the latter did not yet yield the desired results, the support structures did provide partial information about the relative orientations of individual p53 complexes. We used this information to calculate a tomographic 3D reconstruction, and refined this structure to a final resolution of ∼15 Å. This structure settles an ongoing debate about the symmetry of the p53 tetramer bound to DNA.


Subject(s)
Cryoelectron Microscopy/methods , DNA/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , DNA/chemistry , Humans , Imaging, Three-Dimensional/methods , Macromolecular Substances/chemistry , Protein Conformation , Protein Multimerization , Water
14.
Proc Natl Acad Sci U S A ; 113(36): E5271-80, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27551077

ABSTRACT

The tumor suppressor p53 has the most frequently mutated gene in human cancers. Many of p53's oncogenic mutants are just destabilized and rapidly aggregate, and are targets for stabilization by drugs. We found certain 2-sulfonylpyrimidines, including one named PK11007, to be mild thiol alkylators with anticancer activity in several cell lines, especially those with mutationally compromised p53. PK11007 acted by two routes: p53 dependent and p53 independent. PK11007 stabilized p53 in vitro via selective alkylation of two surface-exposed cysteines without compromising its DNA binding activity. Unstable p53 was reactivated by PK11007 in some cancer cell lines, leading to up-regulation of p53 target genes such as p21 and PUMA. More generally, there was cell death that was independent of p53 but dependent on glutathione depletion and associated with highly elevated levels of reactive oxygen species and induction of endoplasmic reticulum (ER) stress, as also found for the anticancer agent PRIMA-1(MET)(APR-246). PK11007 may be a lead for anticancer drugs that target cells with nonfunctional p53 or impaired reactive oxygen species (ROS) detoxification in a wide variety of mutant p53 cells.


Subject(s)
Alkylating Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Pyrimidines/administration & dosage , Sulfones/administration & dosage , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Crystallography, X-Ray , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation , Neoplasms/genetics , Reactive Oxygen Species/metabolism
15.
Protein Eng Des Sel ; 29(9): 377-90, 2016 09.
Article in English | MEDLINE | ID: mdl-27503952

ABSTRACT

The p53 tumor suppressor protein performs a critical role in stimulating apoptosis and cell cycle arrest in response to oncogenic stress. The function of p53 can be compromised by mutation, leading to increased risk of cancer; approximately 50% of cancers are associated with mutations in the p53 gene, the majority of which are in the core DNA-binding domain. The Y220C mutation of p53, for example, destabilizes the core domain by 4 kcal/mol, leading to rapid denaturation and aggregation. The associated loss of tumor suppressor functionality is associated with approximately 75 000 new cancer cases every year. Destabilized p53 mutants can be 'rescued' and their function restored; binding of a small molecule into a pocket on the surface of mutant p53 can stabilize its wild-type structure and restore its function. Here, we describe an in silico algorithm for identifying potential rescue pockets, including the algorithm's integration with the Dynameomics molecular dynamics data warehouse and the DIVE visual analytics engine. We discuss the results of the application of the method to the Y220C p53 mutant, entailing finding a putative rescue pocket through MD simulations followed by an in silico search for stabilizing ligands that dock into the putative rescue pocket. The top three compounds from this search were tested experimentally and one of them bound in the pocket, as shown by nuclear magnetic resonance, and weakly stabilized the mutant.


Subject(s)
Algorithms , Computer Simulation , Mutation , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , DNA/metabolism , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Domains , Protein Stability , Temperature , Tumor Suppressor Protein p53/metabolism
16.
ACS Chem Biol ; 11(8): 2265-74, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27267810

ABSTRACT

Many oncogenic mutants of the tumor suppressor p53 are conformationally unstable, including the frequently occurring Y220C mutant. We have previously developed several small-molecule stabilizers of this mutant. One of these molecules, PhiKan083, 1-(9-ethyl-9H-carbazole-3-yl)-N-methylmethanamine, binds to a mutation-induced surface crevice with a KD = 150 µM, thereby increasing the melting temperature of the protein and slowing its rate of aggregation. Incorporation of fluorine atoms into small molecule ligands can substantially improve binding affinity to their protein targets. We have, therefore, harnessed fluorine-protein interactions to improve the affinity of this ligand. Step-wise introduction of fluorines at the carbazole ethyl anchor, which is deeply buried within the binding site in the Y220C-PhiKan083 complex, led to a 5-fold increase in affinity for a 2,2,2-trifluoroethyl anchor (ligand efficiency of 0.3 kcal mol(-1) atom(-1)). High-resolution crystal structures of the Y220C-ligand complexes combined with quantum chemical calculations revealed favorable interactions of the fluorines with protein backbone carbonyl groups (Leu145 and Trp146) and the sulfur of Cys220 at the mutation site. Affinity gains were, however, only achieved upon trifluorination, despite favorable interactions of the mono- and difluorinated anchors with the binding pocket, indicating a trade-off between energetically favorable protein-fluorine interactions and increased desolvation penalties. Taken together, the optimized carbazole scaffold provides a promising starting point for the development of high-affinity ligands to reactivate the tumor suppressor function of the p53 mutant Y220C in cancer cells.


Subject(s)
Drug Design , Fluorine/chemistry , Mutation , Sulfur/chemistry , Tumor Suppressor Protein p53/chemistry , Biophysics , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Quantum Theory , Tumor Suppressor Protein p53/genetics
17.
Annu Rev Biochem ; 85: 375-404, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145840

ABSTRACT

Inactivation of the transcription factor p53, through either direct mutation or aberrations in one of its many regulatory pathways, is a hallmark of virtually every tumor. In recent years, screening for p53 activators and a better understanding of the molecular mechanisms of oncogenic perturbations of p53 function have opened up a host of novel avenues for therapeutic intervention in cancer: from the structure-guided design of chemical chaperones to restore the function of conformationally unstable p53 cancer mutants, to the development of potent antagonists of the negative regulators MDM2 and MDMX and other modulators of the p53 pathway for the treatment of cancers with wild-type p53. Some of these compounds have now moved from proof-of-concept studies into clinical trials, with prospects for further, personalized anticancer medicines. We trace the structural evolution of the p53 pathway, from germ-line surveillance in simple multicellular organisms to its pluripotential role in humans.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Gene Expression Regulation, Neoplastic , Molecular Targeted Therapy , Neoplasms/drug therapy , Tumor Suppressor Protein p53/agonists , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Cell Cycle Proteins , Clinical Trials as Topic , Drug Design , Humans , Molecular Docking Simulation , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Multimerization , Protein Structure, Secondary , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Structure ; 23(12): 2246-2255, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26636255

ABSTRACT

The destabilizing p53 cancer mutation Y220C creates an extended crevice on the surface of the protein that can be targeted by small-molecule stabilizers. Here, we identify different classes of small molecules that bind to this crevice and determine their binding modes by X-ray crystallography. These structures reveal two major conformational states of the pocket and a cryptic, transiently open hydrophobic subpocket that is modulated by Cys220. In one instance, specifically targeting this transient protein state by a pyrrole moiety resulted in a 40-fold increase in binding affinity. Molecular dynamics simulations showed that both open and closed states of this subsite were populated at comparable frequencies along the trajectories. Our data extend the framework for the design of high-affinity Y220C mutant binders for use in personalized anticancer therapy and, more generally, highlight the importance of implementing protein dynamics and hydration patterns in the drug-discovery process.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Dynamics Simulation , Tumor Suppressor Protein p53/chemistry , Amino Acid Sequence , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Mutation , Protein Binding , Protein Stability , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
ACS Chem Biol ; 10(12): 2725-32, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26378745

ABSTRACT

Bioisosteric replacements are widely used in medicinal chemistry to improve physicochemical and ADME properties of molecules while retaining or improving affinity. Here, using the p53 cancer mutant Y220C as a test case, we investigate both computationally and experimentally whether an ethynyl moiety is a suitable bioisostere to replace iodine in ligands that form halogen bonds with the protein backbone. This bioisosteric transformation is synthetically feasible via Sonogashira cross-coupling. In our test case of a particularly strong halogen bond, replacement of the iodine with an ethynyl group resulted in a 13-fold affinity loss. High-resolution crystal structures of the two analogues in complex with the p53-Y220C mutant enabled us to correlate the different affinities with particular features of the binding site and subtle changes in ligand binding mode. In addition, using QM calculations and analyzing the PDB, we provide general guidelines for identifying cases where such a transformation is likely to improve ligand recognition.


Subject(s)
Acetylene/chemistry , Alkynes/chemistry , Computer Simulation , Halogens/chemistry , Models, Chemical , Phenols/chemistry , Alkynes/pharmacology , Binding Sites , Crystallography, X-Ray , Isomerism , Ligands , Molecular Structure , Mutation , Phenols/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
PLoS One ; 10(5): e0125662, 2015.
Article in English | MEDLINE | ID: mdl-25946337

ABSTRACT

The folding and unfolding of protein domains is an apparently cooperative process, but transient intermediates have been detected in some cases. Such (un)folding intermediates are challenging to investigate structurally as they are typically not long-lived and their role in the (un)folding reaction has often been questioned. One of the most well studied (un)folding pathways is that of Drosophila melanogaster Engrailed homeodomain (EnHD): this 61-residue protein forms a three helix bundle in the native state and folds via a helical intermediate. Here we used molecular dynamics simulations to derive sample conformations of EnHD in the native, intermediate, and unfolded states and selected the relevant structural clusters by comparing to small/wide angle X-ray scattering data at four different temperatures. The results are corroborated using residual dipolar couplings determined by NMR spectroscopy. Our results agree well with the previously proposed (un)folding pathway. However, they also suggest that the fully unfolded state is present at a low fraction throughout the investigated temperature interval, and that the (un)folding intermediate is highly populated at the thermal midpoint in line with the view that this intermediate can be regarded to be the denatured state under physiological conditions. Further, the combination of ensemble structural techniques with MD allows for determination of structures and populations of multiple interconverting structures in solution.


Subject(s)
Drosophila melanogaster/metabolism , Homeodomain Proteins/metabolism , Molecular Dynamics Simulation , Protein Folding , Protein Unfolding , Transcription Factors/metabolism , Animals , Cluster Analysis , Drosophila Proteins , Homeodomain Proteins/analysis , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , Transcription Factors/analysis , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...