Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Bioorg Chem ; 127: 105976, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35777233

RESUMEN

Bicyclization has proven to be an effective strategy for significantly restricting conformational flexibility in peptides and peptidomimetics such as peptoids. Such constrained bicyclic peptoids would have far higher conformational rigidity than monocyclic and linear ones, allowing them to have enhanced binding affinity and selectivity for their biological targets. Herein, we show that bicyclic peptoids have superior cellular uptake efficiency than their linear counterparts regardless of their side chains and ring sizes. As a representative example, an 8-mer bicyclic peptoid achieves a CP50 value of 1.2 µM, which is > 5-times superior to the corresponding linear peptoid. Additionally, we also demonstrate that bicyclic peptide-peptoid hybrids are much more cell-permeable than native peptides. Due to their favorable properties including improved cellular uptake, resistance to proteolytic degradation, relatively large sizes, and enormous structural diversity, constrained bicyclic peptoids and peptide-peptoid hybrids will play an important role as potential drug leads, especially in targeting intracellular protein-protein interactions, which are traditionally considered undruggable.


Asunto(s)
Peptidomiméticos , Peptoides , Péptidos/química , Peptidomiméticos/química , Peptidomiméticos/farmacología , Peptoides/química , Peptoides/metabolismo , Peptoides/farmacología , Permeabilidad
2.
Int J Mol Sci ; 22(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203850

RESUMEN

Steroid receptor coactivator-1 (SRC-1) is a transcription coactivator playing a pivotal role in mediating a wide range of signaling pathways by interacting with related transcription factors and nuclear receptors. Aberrantly elevated SRC-1 activity is associated with cancer metastasis and progression, and therefore, suppression of SRC-1 is emerging as a promising therapeutic strategy. In this study, we developed a novel SRC-1 degrader for targeted degradation of cellular SRC-1. This molecule consists of a selective ligand for SRC-1 and a bulky hydrophobic group. Since the hydrophobic moiety on the protein surface could mimic a partially denatured hydrophobic region of a protein, SRC-1 could be recognized as an unfolded protein and experience the chaperone-mediated degradation in the cells through the ubiquitin-proteasome system (UPS). Our results demonstrate that a hydrophobic-tagged chimeric molecule is shown to significantly reduce cellular levels of SRC-1 and suppress cancer cell migration and invasion. Together, these results highlight that our SRC-1 degrader represents a novel class of therapeutic candidates for targeting cancer metastasis. Moreover, we believe that the hydrophobic tagging strategy would be widely applicable to develop peptide-based protein degraders with enhanced cellular activity.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Coactivador 1 de Receptor Nuclear/metabolismo , Proteolisis , Transactivadores/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Movimiento Celular , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Chaperonas Moleculares/metabolismo , Invasividad Neoplásica , Péptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Angew Chem Int Ed Engl ; 59(40): 17548-17555, 2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-33026161

RESUMEN

Aberrantly elevated steroid receptor coactivator-1 (SRC-1) expression and activity are strongly correlated with cancer progression and metastasis. Here we report, for the first time, the development of a proteolysis targeting chimera (PROTAC) that is composed of a selective SRC-1 binder linked to a specific ligand for UBR box, a unique class of E3 ligases recognizing N-degrons. We showed that the bifunctional molecule efficiently and selectively induced the degradation of SRC-1 in cells through the N-degron pathway. Importantly, given the ubiquitous expression of the UBR protein in most cells, PROTACs targeting the UBR box could degrade a protein of interest regardless of cell types. We also showed that the SRC-1 degrader significantly suppressed cancer cell invasion and migration in vitro and in vivo. Together, these results demonstrate that the SRC-1 degrader can be an invaluable chemical tool in the studies of SRC-1 functions. Moreover, our findings suggest PROTACs based on the N-degron pathway as a widely useful strategy to degrade disease-relevant proteins.


Asunto(s)
Coactivador 1 de Receptor Nuclear/antagonistas & inhibidores , Péptidos/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biocatálisis , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones Endogámicos BALB C , Invasividad Neoplásica/prevención & control , Neoplasias/tratamiento farmacológico , Coactivador 1 de Receptor Nuclear/metabolismo , Péptidos/metabolismo , Péptidos/uso terapéutico , Unión Proteica , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
4.
Chem Commun (Camb) ; 55(88): 13311-13314, 2019 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-31631199

RESUMEN

Herein, we report a strategy for generating conformationally restricted α-helix mimetic small molecules by introducing covalent bridges that limit rotation about the central axis of α-helix mimetics. We demonstrate that the bridged α-helix mimetics have enhanced binding affinity and specificity to the target protein due to the restricted conformation as well as extra interaction of the bridge with the protein surface.


Asunto(s)
Compuestos Heterocíclicos de Anillo en Puente/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Bibliotecas de Moléculas Pequeñas/química , Compuestos Heterocíclicos de Anillo en Puente/farmacología , Humanos , Células Jurkat , Modelos Moleculares , Conformación Molecular , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología
5.
Bioorg Chem ; 91: 103141, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31377386

RESUMEN

Multivalent protein-protein interactions including bivalent and trivalent interactions play a critical role in mediating a wide range of biological processes. Hence, there is a significant interest in developing molecules that can modulate those signaling pathways mediated by multivalent interactions. For example, multimeric molecules capable of binding to a receptor protein through a multivalent interaction could serve as modulators of such interactions. However, it is challenging to efficiently generate such multimeric ligands. Here, we have developed a facile solid-phase method that allows for the rapid generation of (homo- and hetero-) dimeric and trimeric protein ligands. The feasibility of this strategy was demonstrated by efficiently synthesizing fluorescently-labeled dimeric peptide ligands, which led to dramatically increased binding affinities (~400-fold improvement) relative to a monomeric 14-3-3σ protein ligand.


Asunto(s)
Proteínas 14-3-3/metabolismo , Biomarcadores de Tumor/metabolismo , Exorribonucleasas/metabolismo , Péptidos/metabolismo , Triazinas/metabolismo , Proteínas 14-3-3/antagonistas & inhibidores , Proteínas 14-3-3/química , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/química , Línea Celular Tumoral , Exorribonucleasas/antagonistas & inhibidores , Exorribonucleasas/química , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Estructura Molecular , Péptidos/síntesis química , Péptidos/toxicidad , Unión Proteica , Triazinas/síntesis química , Triazinas/toxicidad
6.
Biopolymers ; 110(6): e23289, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31150108

RESUMEN

Peptoids, oligomers of N-substituted glycines, have been attracting increasing interest due to their advantageous properties as peptidomimetics. However, due to the lack of chiral centers and amide hydrogen atoms, peptoids, in general, do not form folding structures except that they have α-chiral side chains. We have recently developed "peptoids with backbone chirality" as a new class of peptoid foldamers called α-ABpeptoids and demonstrated that they could have folding conformations owing to the methyl groups on chiral α-carbons in the backbone structure. Here we report α-ABpeptoid/ß3 -peptide oligomers as a unique peptidomimetic structure with a heterogeneous backbone. This hybrid structure contains a mixed α-ABpeptoid and ß3 -peptide residues arranged in an alternate manner. These α-ABpeptoid/ß3 -peptide oligomers could form intramolecular hydrogen bonding and have better cell permeability relative to pure peptide sequences. These oligomers were shown to adopt ordered folding structures based on circular dichroism studies. Overall, α-ABpeptoid/ß3 -peptide oligomers may represent a novel class of peptidomimetic foldamers and will find a wide range of applications in biomedical and material sciences.


Asunto(s)
Péptidos/química , Peptoides/química , Dicroismo Circular , Enlace de Hidrógeno , Resonancia Magnética Nuclear Biomolecular , Péptidos/metabolismo , Peptidomiméticos , Peptoides/metabolismo , Permeabilidad , Polímeros/química , Pliegue de Proteína , Estereoisomerismo
7.
Chembiochem ; 19(13): 1359-1364, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29577543

RESUMEN

The epithelial-mesenchymal transition (EMT) has been suggested as a new target for therapeutic intervention of metastatic cancer. Forkhead box protein C2 (FOXC2) is known to be necessary for initiating and maintaining EMT, and therefore bestows on cancer cells metastatic and cancer stem cell (CSC)-like phenotypes, allowing cells to acquire higher motility, invasiveness, self-renewal, and therapy resistance. Here, we describe the first inhibitor of FOXC2, MC-1-F2. MC-1-F2 was able to induce cadherin switching and reverse EMT through the degradation of FOXC2 and blocking of its nuclear localization. In addition, MC-1-F2 was very effective in inhibiting cancer cell migration and invasion. As the first small-molecule inhibitor of FOXC2 and the first compound targeting EMT-associated transcription factor, MC-1-F2 will pave the way for a new anticancer therapeutic agent targeting metastatic cancer and help to elucidate the network of EMT signaling pathways.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factores de Transcripción Forkhead/antagonistas & inhibidores , Piperazinas/farmacología , Triazinas/farmacología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Regulación hacia Arriba
8.
J Am Chem Soc ; 139(45): 16056-16059, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29090910

RESUMEN

The complex formation between transcription factors (TFs) and coactivator proteins is required for transcriptional activity, and thus disruption of aberrantly activated TF/coactivator interactions could be an attractive therapeutic strategy. However, modulation of such protein-protein interactions (PPIs) has proven challenging. Here we report a cell-permeable, proteolytically stable, stapled helical peptide directly targeting nuclear receptor coactivator 1 (NCOA1), a coactivator required for the transcriptional activity of signal transducer and activator of transcription 6 (STAT6). We demonstrate that this stapled peptide disrupts the NCOA1/STAT6 complex, thereby repressing STAT6-mediated transcription. Furthermore, we solved the first crystal structure of a stapled peptide in complex with NCOA1. The stapled peptide therefore represents an invaluable chemical probe for understanding the precise role of the NCOA1/STAT6 interaction and an excellent starting point for the development of a novel class of therapeutic agents.


Asunto(s)
Coactivador 1 de Receptor Nuclear/metabolismo , Péptidos/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Factor de Transcripción STAT6/metabolismo , Células A549 , Secuencia de Aminoácidos , Animales , Diseño de Fármacos , Células HEK293 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Coactivador 1 de Receptor Nuclear/antagonistas & inhibidores , Péptidos/química , Factor de Transcripción STAT6/antagonistas & inhibidores
9.
Mol Biosyst ; 13(4): 638-647, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28265599

RESUMEN

Protein folding is crucial for most proteins to achieve their correct three-dimensional conformations and function properly. Defects in protein folding frequently caused by mutations lead to a range of protein misfolding diseases, including Alzheimer's disease, Parkinson's disease, cystic fibrosis, amyloidosis, Gaucher disease, etc. One approach to treat these devastating diseases would be to use pharmacological chaperones, which are small-molecules that bind to and stabilize misfolded proteins, thereby correcting their pathogenic misfolding and rescuing their functions. As such, pharmacological chaperone therapy holds great promise for the treatment of numerous protein misfolding diseases. In this review, we highlight recent strategies for identifying small-molecules that act as pharmacological chaperones and revert protein misfolding diseases, with a focus on reports within the last five years.


Asunto(s)
Descubrimiento de Drogas/métodos , Ligandos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Chaperonas Moleculares/genética , Mutación , Unión Proteica , Pliegue de Proteína , Deficiencias en la Proteostasis
10.
Curr Med Chem ; 23(22): 2363-79, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27160538

RESUMEN

Skp2 is frequently overexpressed in many human cancers and plays a key role in tumorigenesis. As a component of the SCFSkp2ubiquitin E3 ligase complex, Skp2 is responsible for recruiting substrate proteins for their ubiquitination and subsequent degradation by the 26S proteasome. Thus, Skp2 promotes the cell cycle by down-regulating cell cycle proteins such as the tumor suppressor p27. Alternatively, Skp2 suppresses p53-dependent apoptosis by outcompeting p53 for binding to p300, thereby perturbing p300-mediated p53 acetylation and stabilization. Taken together, inhibition of Skp2 functions (either proteolytic function or non-proteolytic function) is emerging as a promising and novel anti-cancer strategy. In the present review, we highlight the development of Skp2 inhibitors with different mechanisms of action.


Asunto(s)
Antineoplásicos/química , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Ácidos Carboxílicos/química , Ácidos Carboxílicos/farmacología , Ácidos Carboxílicos/uso terapéutico , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Humanos , Hidroquinonas/química , Hidroquinonas/farmacología , Hidroquinonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Angew Chem Int Ed Engl ; 55(2): 602-6, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26593157

RESUMEN

Skp2 is thought to have two critical roles in tumorigenesis. As part of the SCF(Skp2) ubiquitin ligase, Skp2 drives the cell cycle by mediating the degradation of cell cycle proteins. Besides the proteolytic activity, Skp2 also blocks p53-mediated apoptosis by outcompeting p53 for binding p300. Herein, we exploit the Skp2/p300 interaction as a new target for Skp2 inhibition. An affinity-based high-throughput screen of a combinatorial cyclic peptoid library identified an inhibitor that binds to Skp2 and interferes with the Skp2/p300 interaction. We show that antagonism of the Skp2/p300 interaction by the inhibitor leads to p300-mediated p53 acetylation, resulting in p53-mediated apoptosis in cancer cells, without affecting Skp2 proteolytic activity. Our results suggest that inhibition of the Skp2/p300 interaction has a great potential as a new anticancer strategy, and our Skp2 inhibitor can be developed as a chemical probe to delineate Skp2 non-proteolytic function in tumorigenesis.


Asunto(s)
Apoptosis/fisiología , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/fisiología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Unión Proteica , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Factores de Transcripción p300-CBP/metabolismo
12.
ACS Comb Sci ; 18(1): 36-42, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26651509

RESUMEN

Many biologically active α-helical peptides adopt amphiphilic helical structures that contain hydrophobic residues on one side and hydrophilic residues on the other side. Therefore, α-helix mimetics capable of mimicking such amphiphilic helical peptides should possess higher binding affinity and specificity to target proteins. Here we describe an efficient method for generating amphiphilic α-helix mimetics. One-face α-helix mimetics having hydrophobic side chains on one side was readily converted into amphiphilic α-helix mimetics by introducing appropriate charged residues on the opposite side. We also demonstrate that such two-face amphiphilic α-helix mimetics indeed show remarkably improved binding affinity to a target protein, compared to one-face hydrophobic α-helix mimetics. We believe that generating a large combinatorial library of these amphiphilic α-helix mimetics can be valuable for rapid discovery of highly potent and specific modulators of protein-protein interactions.


Asunto(s)
Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Peptidomiméticos/química , Peptidomiméticos/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Secuencia de Aminoácidos , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Estructura Secundaria de Proteína , Técnicas de Síntesis en Fase Sólida/métodos
13.
Org Lett ; 16(21): 5710-3, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25310875

RESUMEN

A facile method for sequence determination of cyclic peptides/peptoids is described. Macrocyclic peptides/peptoids of 3-10 residues were efficiently synthesized through thioether formation. One-pot reaction of thioether-embedded cyclic peptides/peptoids involving cyanogen bromide-mediated ring-opening and cleavage provides linearized molecules, which can be efficiently sequenced by tandem mass spectrometry.


Asunto(s)
Bromuro de Cianógeno/química , Péptidos Cíclicos/química , Péptidos Cíclicos/síntesis química , Péptidos/química , Péptidos/síntesis química , Sulfuros/química , Secuencia de Aminoácidos , Estructura Molecular , Espectrometría de Masas en Tándem
14.
ACS Comb Sci ; 16(12): 695-701, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25336412

RESUMEN

α-Helices play a critical role in mediating many protein-protein interactions (PPIs) as recognition motifs. Therefore, there is a considerable interest in developing small molecules that can mimic helical peptide segments to modulate α-helix-mediated PPIs. Due to the relatively low aqueous solubility and synthetic difficulty of most current α-helix mimetic small molecules, one important goal in this area is to develop small molecules with favorable physicochemical properties and ease of synthesis. Here we designed phenyl-piperazine-triazine-based α-helix mimetics that possess improved water solubility and excellent synthetic accessibility. We developed a facile solid-phase synthetic route that allows for rapid creation of a large, diverse combinatorial library of α-helix mimetics. Further, we identified a selective inhibitor of the Mcl-1/BH3 interaction by screening a focused library of phenyl-piperazine-triazines, demonstrating that the scaffold is able to serve as functional mimetics of α-helical peptides. We believe that our phenyl-piperazine-triazine-based α-helix mimetics, along with the facile and divergent solid-phase synthetic method, have great potential as powerful tools for discovering potent inhibitors of given α-helix-mediated PPIs.


Asunto(s)
Benzoatos/síntesis química , Biomimética , Piperazinas/síntesis química , Triazinas/síntesis química , Benzoatos/química , Polarización de Fluorescencia , Modelos Moleculares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Piperazinas/química , Estructura Secundaria de Proteína , Técnicas de Síntesis en Fase Sólida/métodos , Triazinas/química
15.
Proc Natl Acad Sci U S A ; 111(30): 11007-12, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024216

RESUMEN

Pharmacological chaperones are small molecules that bind to proteins and stabilize them against thermal denaturation or proteolytic degradation, as well as assist or prevent certain protein-protein assemblies. These activities are being exploited for the development of treatments for diseases caused by protein instability and/or aberrant protein-protein interactions, such as those found in certain forms of cancers and neurodegenerative diseases. However, designing or discovering pharmacological chaperones for specific targets is challenging because of the relatively featureless protein target surfaces, the lack of suitable chemical libraries, and the shortage of efficient high-throughput screening methods. In this study, we attempted to address all these challenges by synthesizing a diverse library of small molecules that mimic protein α-helical secondary structures commonly found in protein-protein interaction surfaces. This was accompanied by establishing a facile "on-bead" high-throughput screening method that allows for rapid and efficient discovery of potential pharmacological chaperones and for identifying novel chaperones/inhibitors against a cancer-associated protein, myeloid cell leukemia 1 (MCL-1), and a Parkinson disease-associated protein, α-synuclein. Our data suggest that the compounds and methods described here will be useful tools for the development of pharmaceuticals for complex-disease targets that are traditionally deemed "undruggable."


Asunto(s)
Descubrimiento de Drogas , Chaperonas Moleculares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias , Enfermedad de Parkinson , alfa-Sinucleína , Humanos , Células Jurkat , Chaperonas Moleculares/síntesis química , Chaperonas Moleculares/química , Chaperonas Moleculares/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/metabolismo
16.
J Biol Chem ; 288(21): 15181-93, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23564455

RESUMEN

The adaptor protein Amot130 scaffolds components of the Hippo pathway to promote the inhibition of cell growth. This study describes how Amot130 through binding and activating the ubiquitin ligase AIP4/Itch achieves these effects. AIP4 is found to bind and ubiquitinate Amot130 at residue Lys-481. This both stabilizes Amot130 and promotes its residence at the plasma membrane. Furthermore, Amot130 is shown to scaffold a complex containing overexpressed AIP4 and the transcriptional co-activator Yes-associated protein (YAP). Consequently, Amot130 promotes the ubiquitination of YAP by AIP4 and prevents AIP4 from binding to large tumor suppressor 1. Amot130 is found to reduce YAP stability. Importantly, Amot130 inhibition of YAP dependent transcription is reversed by AIP4 silencing, whereas Amot130 and AIP4 expression interdependently suppress cell growth. Thus, Amot130 repurposes AIP4 from its previously described role in degrading large tumor suppressor 1 to the inhibition of YAP and cell growth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Angiomotinas , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Proteolisis , Proteínas Represoras/genética , Factores de Transcripción , Transcripción Genética/fisiología , Ubiquitina-Proteína Ligasas/genética , Proteínas Señalizadoras YAP
17.
J Chem Inf Model ; 52(7): 1821-32, 2012 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-22731511

RESUMEN

Molecular docking is widely used to obtain binding modes and binding affinities of a molecule to a given target protein. Despite considerable efforts, however, prediction of both properties by docking remains challenging mainly due to protein's structural flexibility and inaccuracy of scoring functions. Here, an integrated approach has been developed to improve the accuracy of binding mode and affinity prediction and tested for small molecule MDM2 and MDMX antagonists. In this approach, initial candidate models selected from docking are subjected to equilibration MD simulations to further filter the models. Free energy perturbation molecular dynamics (FEP/MD) simulations are then applied to the filtered ligand models to enhance the ability in predicting the near-native ligand conformation. The calculated binding free energies for MDM2 complexes are overestimated compared to experimental measurements mainly due to the difficulties in sampling highly flexible apo-MDM2. Nonetheless, the FEP/MD binding free energy calculations are more promising for discriminating binders from nonbinders than docking scores. In particular, the comparison between the MDM2 and MDMX results suggests that apo-MDMX has lower flexibility than apo-MDM2. In addition, the FEP/MD calculations provide detailed information on the different energetic contributions to ligand binding, leading to a better understanding of the sensitivity and specificity of protein-ligand interactions.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas Nucleares/química , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas/química , Sitios de Unión , Proteínas de Ciclo Celular , Química Farmacéutica , Estabilidad de Medicamentos , Predicción , Humanos , Ligandos , Modelos Moleculares , Proteínas Nucleares/antagonistas & inhibidores , Unión Proteica , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Termodinámica
18.
J Am Chem Soc ; 133(4): 676-9, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21171592

RESUMEN

There is considerable interest in developing non-peptidic, small-molecule α-helix mimetics to disrupt α-helix-mediated protein−protein interactions. Herein, we report the design of a novel pyrrolopyrimidine-based scaffold for such α-helix mimetics with increased conformational rigidity. We also developed a facile solid-phase synthetic route that is amenable to divergent synthesis of a large library. Using a fluorescence polarization-based assay, we identified cell-permeable, dual MDMX/MDM2 inhibitors, demonstrating that the designed molecules can act as α-helix mimetics.


Asunto(s)
Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirimidinas/química , Pirroles/química , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Línea Celular Tumoral , Diseño de Fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Permeabilidad , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Proteína p53 Supresora de Tumor/química
19.
J Biol Chem ; 284(50): 34522-30, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19846554

RESUMEN

It has been shown previously that sub-complexes of the 26 S proteasome can regulate gene expression via non-proteolytic mechanisms. One such mechanism is the disruption of activator.promoter complexes in an ATP-dependent fashion, which was discovered in the context of the yeast Gal4 system. This activity strongly inhibits Gal4-driven gene expression unless the activator is mono-ubiquitylated, which protects it from the ATPases. To address whether this paradigm is also applicable to medically important mammalian transcriptional activators we report here a study of the interaction of the proteasomal ATPases with p53. It is shown that p53 binds directly to the ATPases via its C-terminal tetramerization and regulatory domain and that p53.promoter complexes are indeed vulnerable to ATPase-dependent disruption by the ATPase complex in vitro. Knockdown of one of the ATPases, Rpt6, in living cells results in increased occupancy of the p21(waf1) promoter by p53 and increased expression of the gene, consistent with the idea that the proteasomal ATPases negatively regulate p53 function in a non-proteolytic fashion.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Transcripción Genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Adenosina Trifosfatasas/genética , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA