Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 85: 375-404, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27145840

RESUMEN

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.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/agonistas , Animales , Antineoplásicos Alquilantes/síntesis química , Proteínas de Ciclo Celular , Ensayos Clínicos como Asunto , Diseño de Fármacos , Humanos , Simulación del Acoplamiento Molecular , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Q Rev Biophys ; 57: e4, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38597675

RESUMEN

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.


Asunto(s)
Pliegue de Proteína , Proteínas , Simulación por Computador , Proteínas/química , Ingeniería de Proteínas , Biología , Cinética , Termodinámica
3.
Chembiochem ; 21(1-2): 5-6, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31769931

RESUMEN

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.


Asunto(s)
Productos Biológicos/metabolismo , Carbohidratos/química , Ácidos Nucleicos/metabolismo , Péptidos/metabolismo , Proteínas/metabolismo , Productos Biológicos/química , Humanos , Ácidos Nucleicos/química , Péptidos/química , Proteínas/química , Biología Sintética
4.
Nat Rev Mol Cell Biol ; 9(8): 650-4, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18578032

RESUMEN

Every breakthrough that opens new vistas also removes the ground from under the feet of other scientists. The scientific joy of those who have seen the new light is accompanied by the dismay of those whose way of life has been changed for ever. The publication of the first structures of proteins at atomic resolution 50 years ago astounded and inspired scientists in every field, but caused others to flee or scoff. That advance and every subsequent paradigm-shifting breakthrough in protein science have met with some resistance before universal acceptance. I relate these events and their impact on the field of protein folding.


Asunto(s)
Conformación Proteica , Pliegue de Proteína , Animales , Simulación por Computador , Genómica/tendencias , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Imagenología Tridimensional , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular/métodos , Ingeniería de Proteínas/historia , Ingeniería de Proteínas/tendencias , Proteínas Recombinantes/química
5.
Proc Natl Acad Sci U S A ; 114(13): E2634-E2643, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28292898

RESUMEN

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.


Asunto(s)
Agregación Patológica de Proteínas , Proteína p53 Supresora de Tumor/metabolismo , Humanos , Modelos Teóricos , Mutación , Neoplasias/genética , Dominios Proteicos , Proteína p53 Supresora de Tumor/química
6.
J Biol Chem ; 293(12): 4262-4276, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29382728

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Curcumina/análogos & derivados , Proteínas Mutantes/genética , Mutación , Neoplasias/patología , Piperidonas/farmacología , Proteína p53 Supresora de Tumor/genética , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , Proteínas Mutantes/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Proc Natl Acad Sci U S A ; 113(36): E5271-80, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27551077

RESUMEN

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.


Asunto(s)
Alquilantes/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , Pirimidinas/administración & dosificación , Sulfonas/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Cristalografía por Rayos X , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mutación , Neoplasias/genética , Especies Reactivas de Oxígeno/metabolismo
8.
Proc Natl Acad Sci U S A ; 113(47): E7456-E7463, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821763

RESUMEN

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.


Asunto(s)
Microscopía por Crioelectrón/métodos , ADN/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , ADN/química , Humanos , Imagenología Tridimensional/métodos , Sustancias Macromoleculares/química , Conformación Proteica , Multimerización de Proteína , Agua
9.
Proc Natl Acad Sci U S A ; 112(8): 2437-42, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675526

RESUMEN

Many oncogenic mutations inactivate the tumor suppressor p53 by destabilizing it, leading to its rapid aggregation. Small molecule drugs are being developed to stabilize such mutants. The kinetics of aggregation of p53 is deceptively simple. The initial steps in the micromolar concentration range follow apparent sigmoidal sequential first-order kinetics, with rate constants k1 and k2. However, the aggregation kinetics of a panel of mutants prepared for Φ-value analysis has now revealed a bimolecular reaction hidden beneath the observed first-order kinetics. Φu measures the degree of local unfolding on a scale of 0-1. A number of sequential Φu-values of ∼1 for k1 and k2 over the molecule implied more than one protein molecule must be reacting, which was confirmed by finding a clear concentration dependence at submicromolar protein. Numerical simulations showed that the kinetics of the more complex mechanism is difficult, if not impossible, to distinguish experimentally from simple first order under many reaction conditions. Stabilization of mutants by small molecules will be enhanced because they decrease both k1 and k2. The regions with high Φu-values point to the areas where stabilization of mutant proteins would have the greatest effect.


Asunto(s)
Agregado de Proteínas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Benzotiazoles , Fenómenos Biofísicos , Simulación por Computador , Cinética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estructura Terciaria de Proteína , Tiazoles/metabolismo
10.
Proc Natl Acad Sci U S A ; 112(8): 2443-8, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675527

RESUMEN

Destabilized mutant p53s coaggregate with WT p53, p63, and p73 in cancer cell lines. We found that stoichiometric amounts of aggregation-prone mutants induced only small amounts of WT p53 to coaggregate, and preformed aggregates did not significantly seed the aggregation of bulk protein. Similarly, p53 mutants trapped only small amounts of p63 and p73 into their p53 aggregates. Tetrameric full-length protein aggregated at similar rates and kinetics to isolated core domains, but there was some induced aggregation of WT by mutants in hetero-tetramers. p53 aggregation thus differs from the usual formation of amyloid fibril or prion aggregates where tiny amounts of preformed aggregate rapidly seed further aggregation. The proposed aggregation mechanism of p53 of rate-determining sequential unfolding and combination of two molecules accounts for the difference. A molecule of fast-unfolding mutant preferentially reacts with another molecule of mutant and only occasionally traps a slower unfolding WT molecule. The mutant population rapidly self-aggregates before much WT protein is depleted. Subsequently, WT protein self-aggregates at its normal rate. However, the continual production of mutant p53 in a cancer cell would gradually trap more and more WT and other proteins, accounting for the observations of coaggregates in vivo. The mechanism corresponds more to trapping by cross-reaction and coaggregation rather than classical seeding and growth.


Asunto(s)
Agregado de Proteínas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Benzotiazoles , Simulación por Computador , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Cinética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Tiazoles/metabolismo , Factores de Tiempo , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo
11.
Proc Natl Acad Sci U S A ; 110(44): 17814-9, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24127580

RESUMEN

MDM2 and MDMX are homologous proteins that bind to p53 and regulate its activity. Both contain three folded domains and ~70% intrinsically disordered regions. Previous detailed structural and biophysical studies have concentrated on the isolated folded domains. The N-terminal domains of both exhibit high affinity for the disordered N-terminal of p53 (p53TAD) and inhibit its transactivation function. Here, we have studied full-length MDMX and found a ~100-fold weaker affinity for p53TAD than does its isolated N-terminal domain. We found from NMR spectroscopy and binding studies that MDMX (but not MDM2) contains a conserved, disordered self-inhibitory element that competes intramolecularly for binding with p53TAD. This motif, which we call the WWW element, is centered around residues Trp200 and Trp201. Deletion or mutation of the element increased binding affinity of MDMX to that of the isolated N-terminal domain level. The self-inhibition of MDMX implies a regulatory, allosteric mechanism of its activity. MDMX rests in a latent state in which its binding activity with p53TAD is masked by autoinhibition. Activation of MDMX would require binding to a regulatory protein. The inhibitory function of the WWW element may explain the oncogenic effects of an alternative splicing variant of MDMX that does not contain the WWW element and is found in some aggressive cancers.


Asunto(s)
Proteínas Nucleares/genética , Oncogenes/genética , Proteínas Proto-Oncogénicas/genética , Elementos Reguladores de la Transcripción/genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Calorimetría , Proteínas de Ciclo Celular , Cromatografía en Gel , Humanos , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Proteína p53 Supresora de Tumor/genética , Ubiquitinación , Ultracentrifugación
12.
Nucleic Acids Res ; 41(12): 6034-44, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23630318

RESUMEN

The p53 cancer mutant Y220C is an excellent paradigm for rescuing the function of conformationally unstable p53 mutants because it has a unique surface crevice that can be targeted by small-molecule stabilizers. Here, we have identified a compound, PK7088, which is active in vitro: PK7088 bound to the mutant with a dissociation constant of 140 µM and raised its melting temperature, and we have determined the binding mode of a close structural analogue by X-ray crystallography. We showed that PK7088 is biologically active in cancer cells carrying the Y220C mutant by a battery of tests. PK7088 increased the amount of folded mutant protein with wild-type conformation, as monitored by immunofluorescence, and restored its transcriptional functions. It induced p53-Y220C-dependent growth inhibition, cell-cycle arrest and apoptosis. Most notably, PK7088 increased the expression levels of p21 and the proapoptotic NOXA protein. PK7088 worked synergistically with Nutlin-3 on up-regulating p21 expression, whereas Nutlin-3 on its own had no effect, consistent with its mechanism of action. PK7088 also restored non-transcriptional apoptotic functions of p53 by triggering nuclear export of BAX to the mitochondria. We suggest a set of criteria for assigning activation of p53.


Asunto(s)
Antineoplásicos/farmacología , Mutación , Pirazoles/farmacología , Pirroles/farmacología , Proteína p53 Supresora de Tumor/efectos de los fármacos , Antineoplásicos/química , Apoptosis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Genes p53 , Humanos , Conformación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Pirazoles/química , Pirroles/química , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(34): 13590-5, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869710

RESUMEN

Aggregation of p53 is initiated by first-order processes that generate an aggregation-prone state with parallel pathways of major or partial unfolding. Here, we elaborate the mechanism and explore its consequences, beginning with the core domain and extending to the full-length p53 mutant Y220C. Production of large light-scattering particles was slower than formation of the Thioflavin T-binding state and simultaneous depletion of monomer. EDTA removes Zn(2+) to generate apo-p53, which aggregated faster than holo-p53. Apo-Y220C also aggregated by both partial and major unfolding. Apo-p53 was not an obligatory intermediate in the aggregation of holo-p53, but affords a parallel pathway that may be relevant to oncogenic mutants with impaired Zn(2+) binding. Full-length tetrameric Y220C formed the Thioflavin T-binding state with similar rate constants to those of core domain, consistent with a unimolecular initiation that is unaffected by neighboring subunits, but very slowly formed small light-scattering particles. Apo-Y220C and aggregated holo-Y220C had little, if any, seeding effect on the initial polymerization of holo-Y220C (measured by Thioflavin T binding), consistent with initiation being a unimolecular process. But apo-Y220C and aggregated holo-Y220C accelerated somewhat the subsequent formation of light-scattering particles from holo-protein, implying coaggregation. The implications for cancer cells containing wild-type and unstable mutant alleles are that aggregation of wild-type p53 (or homologs) might not be seeded by aggregated mutant, but it could coaggregate with p53 or other cellular proteins that have undergone the first steps of aggregation and speed up the formation of microscopically observable aggregates.


Asunto(s)
Mutación , Tiazoles/química , Proteína p53 Supresora de Tumor/química , Benzotiazoles , Sitios de Unión , Reactivos de Enlaces Cruzados/química , Ácido Edético/química , Humanos , Cinética , Luz , Desnaturalización Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Dispersión de Radiación , Temperatura
14.
Proc Natl Acad Sci U S A ; 109(34): 13584-9, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869713

RESUMEN

Aggregation of destabilized mutants of the tumor suppressor p53 is a major route for its loss of activity. In order to assay drugs that inhibit aggregation of p53, we established the basic kinetics of aggregation of its core domain, using the mutant Y220C that has a mutation-induced, druggable cavity. Aggregation monitored by light scattering followed lag kinetics. Electron microscopy revealed the formation of small aggregates that subsequently grew to larger amorphous aggregates. The kinetics of aggregation produced surprising results: progress curves followed either by the binding of Thioflavin T or the fluorescence of the protein at 340 nm fitted well to simple two-step sequential first-order lag kinetics with rate constants k(1) and k(2) that were independent of protein concentration, and not to classical nucleation-growth. We suggest a mechanism of first-order formation of an aggregation competent state as being rate determining followed by rapid polymerization with the higher order kinetics. By measuring the inhibition kinetics of k(1) and k(2), we resolved that the process with the higher rate constant followed that of the lower. Further, there was only partial inhibition of k(1) and k(2), which showed two parallel pathways of aggregation, one via a state that requires unfolding of the protein and the other of partial unfolding with the ligand still bound. Inhibition kinetics of ligands provides a useful tool for probing an aggregation mechanism.


Asunto(s)
Mutación , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Amiloide/química , Benzotiazoles , Humanos , Cinética , Ligandos , Luz , Microscopía Electrónica/métodos , Neoplasias/metabolismo , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Pliegue de Proteína , Dispersión de Radiación , Tiazoles/química
15.
Proc Natl Acad Sci U S A ; 109(39): 15752-6, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22972749

RESUMEN

The tumor suppressor p53 is a homotetramer of 4 × 393 residues. Its core domain and tetramerization domain are linked and flanked by intrinsically disordered sequences, which hinder its full structural characterization. There is an outstanding problem of the state of the tetramerization domain. Structural studies on the isolated tetramerization domain show it is in a folded tetrameric conformation, but there are conflicting models from electron microscopy of the full-length protein, one of which proposes that the domain is not tetramerically folded and the tetrameric protein is stabilized by interactions between the N and C termini. Here, we present methyl-transverse relaxation optimized NMR spectroscopy (methyl-TROSY) investigations on the full-length and separate domains of the protein with its methionine residues enriched with (13)C to probe its quaternary structure. We obtained high-quality spectra of both the full-length tetrameric p53 and its DNA complex, observing the environment at 11 specific methyl sites. The tetramerization domain was as tetramerically folded in the full-length constructs as in the isolated domain. The N and C termini were intrinsically disordered in both the full-length protein and its complex with a 20-residue specific DNA sequence. Additionally, we detected in the interface of the core (DNA-binding) and N-terminal parts of the protein a slow conformational exchange process that was modulated by specific recognition of DNA, indicating allosteric processes.


Asunto(s)
ADN/química , Pliegue de Proteína , Proteína p53 Supresora de Tumor/química , Regulación Alostérica , ADN/metabolismo , Humanos , Resonancia Magnética Nuclear Biomolecular , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/metabolismo
16.
Proc Natl Acad Sci U S A ; 109(41): 16552-7, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012405

RESUMEN

Proper timing of gene expression requires that transcription factors (TFs) efficiently locate and bind their target sites within a genome. Theoretical studies have long proposed that one-dimensional sliding along DNA while simultaneously reading its sequence can accelerate TF's location of target sites. Sliding by prokaryotic and eukaryotic TFs were subsequently observed. More recent theoretical investigations have argued that simultaneous reading and sliding is not possible for TFs without their possessing at least two DNA-binding modes. The tumor suppressor p53 has been shown to slide on DNA, and recent experiments have offered structural and single molecule support for a two-mode model for the protein. If the model is applicable to p53, then the requirement that TFs be able to read while sliding implies that noncognate sites will affect p53's mobility on DNA, which will thus be generally sequence-dependent. Here, we confirm this prediction with single-molecule microscopy measurements of p53's local diffusivity on noncognate DNA. We show how a two-mode model accurately predicts the variation in local diffusivity, while a single-mode model does not. We further determine that the best model of sequence-specific binding energy includes terms for "hemi-specific" binding, with one dimer of tetrameric p53 binding specifically to a half-site and the other binding nonspecifically to noncognate DNA. Our work provides evidence that the recognition by p53 of its targets and the timing thereof can depend on its noncognate binding properties and its ability to change between multiple modes of binding, in addition to the much better-studied effects of cognate-site binding.


Asunto(s)
ADN/genética , ADN/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Algoritmos , Animales , Secuencia de Bases , Sitios de Unión/genética , ADN/química , Regulación de la Expresión Génica , Humanos , Cinética , Modelos Genéticos , Conformación de Ácido Nucleico , Motivos de Nucleótidos/genética , Unión Proteica , Multimerización de Proteína , Factores de Transcripción/química , Proteína p53 Supresora de Tumor/química
17.
Proc Natl Acad Sci U S A ; 109(42): 16906-10, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23035244

RESUMEN

The proteins MDM2 and MDM4 are key negative regulators of the tumor suppressor protein p53, which are frequently upregulated in cancer cells. They inhibit the transactivation activity of p53 by binding separately or in concert to its transactivation domain. MDM2 is also a ubiquitin ligase that leads to the degradation of p53. Accordingly, MDM2 and MDM4 are important targets for drugs to inhibit their binding to p53. We found from in silico screening and confirmed by experiment that lithocholic acid (LCA) binds to the p53 binding sites of both MDM2 and MDM4 with a fivefold preference for MDM4. LCA is an endogenous steroidal bile acid, variously reported to have both carcinogenic and apoptotic activities. The comparison of LCA effects on apoptosis in HCT116 p53(+/+) vs. p53(-/-) cells shows a predominantly p53-mediated induction of caspase-3/7. The dissociation constants are in the µM region, but only modest inhibition of binding of MDM2 and MDM4 is required to negate their upregulation because they have to compete with transcriptional coactivator p300 for binding to p53. Binding was weakened by structural changes in LCA, and so it may be a natural ligand of MDM2 and MDM4, raising the possibility that MDM proteins may be sensors for specific steroids.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Ácido Litocólico/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cromatografía de Afinidad , Escherichia coli , Polarización de Fluorescencia , Humanos , Espectroscopía de Resonancia Magnética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ultracentrifugación
18.
Proc Natl Acad Sci U S A ; 108(2): 569-73, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187427

RESUMEN

Current questions in protein folding mechanisms include how fast can a protein fold and are there energy barriers for the folding and unfolding of ultrafast folding proteins? The small 3-helical engrailed homeodomain protein folds in 1.7 µs to form a well-characterized intermediate, which rearranges in 17 µs to native structure. We found that the homologous pituitary-specific transcription factor homeodomain (Pit1) folded in a similar manner, but in two better separated kinetic phases of 2.3 and 46 µs. The greater separation and better fluorescence changes facilitated a detailed kinetic analysis for the ultrafast phase for formation of the intermediate. Its folding rate constant changed little with denaturant concentration or mutation but unfolding was very sensitive to denaturant and energy changes on mutation. The folding rate constant of 3 × 10(5) s(-1) in water decreased with increasing viscosity, and was extrapolated to 4.4 × 10(5) s(-1) at zero viscosity. Thus, the formation of the intermediate was partly rate limited by chain diffusion and partly by an energy barrier to give a very diffuse transition state, which was followed by the formation of structure. Conversely, the unfolding reaction required the near complete disruption of the tertiary structure of the intermediate in a highly cooperative manner, being exquisitely sensitive to individual mutations. The folding is approaching, but has not reached, the downhill-folding scenario of energy landscape theory. Under folding conditions, there is a small energy barrier between the denatured and transition states but a larger barrier between native and transition states.


Asunto(s)
Factor de Transcripción Pit-1/química , Secuencia de Aminoácidos , Biofisica/métodos , Concentración de Iones de Hidrógeno , Cinética , Conformación Molecular , Datos de Secuencia Molecular , Mutación , Pliegue de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Programas Informáticos , Temperatura , Termodinámica , Viscosidad
19.
Proc Natl Acad Sci U S A ; 108(14): 5596-601, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21422286

RESUMEN

Members of the homeodomain superfamily are three-helix bundle proteins whose second and third helices form a helix-turn-helix motif (HTH). Their folding mechanism slides from the ultrafast, three-state framework mechanism for the engrailed homeodomain (EnHD), in which the HTH motif is independently stable, to an apparent two-state nucleation-condensation model for family members with an unstable HTH motif. The folding intermediate of EnHD has nearly native HTH structure, but it is not docked with helix1. The determinant of whether two- or three-state folding was hypothesized to be the stability of the HTH substructure. Here, we describe a detailed Φ-value analysis of the folding of the Pit1 homeodomain, which has similar ultrafast kinetics to that of EnHD. Formation of helix1 was strongly coupled with formation of HTH, which was initially surprising because they are uncoupled in the EnHD folding intermediate. However, we found a key difference between Pit1 and EnHD: The isolated peptide corresponding to the HTH motif in Pit1 was not folded in the absence of H1. Independent molecular dynamics simulations of Pit1 unfolding found an intermediate with H1 misfolded onto the HTH motif. The Pit1 folding pathway is the connection between that of EnHD and the slower folding homeodomains and provides a link in the transition of mechanisms from two- to three-state folding in this superfamily. The malleability of folding intermediates can lead to unstable substructures being stabilized by a variety of nonnative interactions, adding to the continuum of folding mechanisms.


Asunto(s)
Secuencias Hélice-Giro-Hélice/genética , Proteínas de Homeodominio/genética , Modelos Moleculares , Pliegue de Proteína , Factor de Transcripción Pit-1/metabolismo , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Proteínas de Homeodominio/fisiología , Cinética , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Datos de Secuencia Molecular
20.
Proc Natl Acad Sci U S A ; 108(2): 563-8, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21178072

RESUMEN

The tumor suppressor p53 slides along DNA while searching for its cognate site. Central to this process is the basic C-terminal domain, whose regulatory role and its coordination with the core DNA-binding domain is highly debated. Here we use single-molecule techniques to characterize the search process and disentangle the roles played by these two DNA-binding domains in the search process. We demonstrate that the C-terminal domain is capable of rapid translocation, while the core domain is unable to slide and instead hops along DNA. These findings are integrated into a model, in which the C-terminal domain mediates fast sliding of p53, while the core domain samples DNA by frequent dissociation and reassociation, allowing for rapid scanning of long DNA regions. The model further proposes how modifications of the C-terminal domain can activate "latent" p53 and reconciles seemingly contradictory data on the action of different domains and their coordination.


Asunto(s)
Proteína p53 Supresora de Tumor/química , ADN/química , Difusión , Dimerización , Escherichia coli/metabolismo , Genes p53 , Humanos , Cinética , Mutación , Fosfatos/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Elementos de Respuesta , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA