Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 152(3): 557-69, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374350

RESUMEN

Dimerization-driven activation of the intracellular kinase domains of the epidermal growth factor receptor (EGFR) upon extracellular ligand binding is crucial to cellular pathways regulating proliferation, migration, and differentiation. Inactive EGFR can exist as both monomers and dimers, suggesting that the mechanism regulating EGFR activity may be subtle. The membrane itself may play a role but creates substantial difficulties for structural studies. Our molecular dynamics simulations of membrane-embedded EGFR suggest that, in ligand-bound dimers, the extracellular domains assume conformations favoring dimerization of the transmembrane helices near their N termini, dimerization of the juxtamembrane segments, and formation of asymmetric (active) kinase dimers. In ligand-free dimers, by holding apart the N termini of the transmembrane helices, the extracellular domains instead favor C-terminal dimerization of the transmembrane helices, juxtamembrane segment dissociation and membrane burial, and formation of symmetric (inactive) kinase dimers. Electrostatic interactions of EGFR's intracellular module with the membrane are critical in maintaining this coupling.


Asunto(s)
Membrana Celular/metabolismo , Receptores ErbB/química , Membrana Celular/química , Dimerización , Receptores ErbB/metabolismo , Humanos , Lípidos de la Membrana/metabolismo , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Estructura Terciaria de Proteína , Electricidad Estática
2.
Cell ; 152(3): 543-56, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374349

RESUMEN

How the epidermal growth factor receptor (EGFR) activates is incompletely understood. The intracellular portion of the receptor is intrinsically active in solution, and to study its regulation, we measured autophosphorylation as a function of EGFR surface density in cells. Without EGF, intact EGFR escapes inhibition only at high surface densities. Although the transmembrane helix and the intracellular module together suffice for constitutive activity even at low densities, the intracellular module is inactivated when tethered on its own to the plasma membrane, and fluorescence cross-correlation shows that it fails to dimerize. NMR and functional data indicate that activation requires an N-terminal interaction between the transmembrane helices, which promotes an antiparallel interaction between juxtamembrane segments and release of inhibition by the membrane. We conclude that EGF binding removes steric constraints in the extracellular module, promoting activation through N-terminal association of the transmembrane helices.


Asunto(s)
Membrana Celular/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/química , Transducción de Señal , Animales , Células COS , Membrana Celular/química , Chlorocebus aethiops , Dimerización , Receptores ErbB/metabolismo , Humanos , Modelos Moleculares
3.
Cell ; 149(4): 860-70, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22579287

RESUMEN

The mutation and overexpression of the epidermal growth factor receptor (EGFR) are associated with the development of a variety of cancers, making this prototypical dimerization-activated receptor tyrosine kinase a prominent target of cancer drugs. Using long-timescale molecular dynamics simulations, we find that the N lobe dimerization interface of the wild-type EGFR kinase domain is intrinsically disordered and that it becomes ordered only upon dimerization. Our simulations suggest, moreover, that some cancer-linked mutations distal to the dimerization interface, particularly the widespread L834R mutation (also referred to as L858R), facilitate EGFR dimerization by suppressing this local disorder. Corroborating these findings, our biophysical experiments and kinase enzymatic assays indicate that the L834R mutation causes abnormally high activity primarily by promoting EGFR dimerization rather than by allowing activation without dimerization. We also find that phosphorylation of EGFR kinase domain at Tyr845 may suppress the intrinsic disorder, suggesting a molecular mechanism for autonomous EGFR signaling.


Asunto(s)
Receptores ErbB/química , Receptores ErbB/genética , Neoplasias/metabolismo , Mutación Puntual , Transducción de Señal , Secuencia de Aminoácidos , Cristalografía por Rayos X , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Gefitinib , Humanos , Lapatinib , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Pliegue de Proteína , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Estructura Terciaria de Proteína , Quinazolinas/farmacología , Alineación de Secuencia
4.
Blood ; 2024 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-38643492

RESUMEN

Secondary kinase domain mutations in BCR::ABL1 represent the most common cause of resistance to tyrosine kinase inhibitor (TKI) therapy in chronic myeloid leukemia patients. The first five approved BCR::ABL1 TKIs target the ATP-binding pocket. Mutations confer resistance to these ATP-competitive TKIs and those approved for other malignancies by decreasing TKI affinity and/or increasing ATP affinity. Asciminib, the first highly active allosteric TKI approved for any malignancy, targets an allosteric regulatory pocket in the BCR::ABL1 kinase C-lobe. As a non-ATP-competitive inhibitor, the activity of asciminib is predicted to be impervious to increases in ATP affinity. Here we report several known mutations that confer resistance to ATP-competitive TKIs in the BCR::ABL1 kinase N-lobe that are distant from the asciminib binding pocket yet unexpectedly confer in vitro resistance to asciminib. Among these is BCR::ABL1 M244V, which confers clinical resistance even to escalated asciminib doses. We demonstrate that BCR::ABL1 M244V does not impair asciminib binding, thereby invoking a novel mechanism of resistance. Molecular dynamics simulations of the M244V substitution implicate stabilization of an active kinase conformation through impact on the -C helix as a mechanism of resistance. These N-lobe mutations may compromise the clinical activity of ongoing combination studies of asciminib with ATP-competitive TKIs.

5.
PLoS Comput Biol ; 18(3): e1009817, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35239648

RESUMEN

Protein-protein interactions (PPIs) are ubiquitous biomolecular processes that are central to virtually all aspects of cellular function. Identifying small molecules that modulate specific disease-related PPIs is a strategy with enormous promise for drug discovery. The design of drugs to disrupt PPIs is challenging, however, because many potential drug-binding sites at PPI interfaces are "cryptic": When unoccupied by a ligand, cryptic sites are often flat and featureless, and thus not readily recognizable in crystal structures, with the geometric and chemical characteristics of typical small-molecule binding sites only emerging upon ligand binding. The rational design of small molecules to inhibit specific PPIs would benefit from a better understanding of how such molecules bind at PPI interfaces. To this end, we have conducted unbiased, all-atom MD simulations of the binding of four small-molecule inhibitors (SP4206 and three SP4206 analogs) to interleukin 2 (IL2)-which performs its function by forming a PPI with its receptor-without incorporating any prior structural information about the ligands' binding. In multiple binding events, a small molecule settled into a stable binding pose at the PPI interface of IL2, resulting in a protein-small-molecule binding site and pose virtually identical to that observed in an existing crystal structure of the IL2-SP4206 complex. Binding of the small molecule stabilized the IL2 binding groove, which when the small molecule was not bound emerged only transiently and incompletely. Moreover, free energy perturbation (FEP) calculations successfully distinguished between the native and non-native IL2-small-molecule binding poses found in the simulations, suggesting that binding simulations in combination with FEP may provide an effective tool for identifying cryptic binding sites and determining the binding poses of small molecules designed to disrupt PPI interfaces by binding to such sites.


Asunto(s)
Descubrimiento de Drogas , Interleucina-2 , Sitios de Unión , Interleucina-2/química , Interleucina-2/metabolismo , Ligandos , Unión Proteica
6.
Mol Cell ; 55(6): 891-903, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25201411

RESUMEN

Trans-autophosphorylation is among the most prevalent means of protein kinase activation, yet its molecular basis is poorly defined. In Toll-like receptor and interleukin-1 receptor signaling pathways, the kinase IRAK4 is recruited to the membrane-proximal adaptor MyD88 through death domain (DD) interactions, forming the oligomeric Myddosome and mediating NF-κB activation. Here we show that unphosphorylated IRAK4 dimerizes in solution with a KD of 2.5 µM and that Myddosome assembly greatly enhances IRAK4 kinase domain (KD) autophosphorylation at sub-KD concentrations. The crystal structure of the unphosphorylated IRAK4(KD) dimer captures a conformation that appears to represent the actual trans-autophosphorylation reaction, with the activation loop phosphosite of one IRAK4 monomer precisely positioned for phosphotransfer by its partner. We show that dimerization is crucial for IRAK4 autophosphorylation in vitro and ligand-dependent signaling in cells. These studies identify a mechanism for oligomerization-driven allosteric autoactivation of IRAK4 that may be general to other kinases activated by autophosphorylation.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/química , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/química , Factor 88 de Diferenciación Mieloide/metabolismo , Dominio Catalítico , Células Cultivadas , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Fosforilación , Conformación Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Dispersión de Radiación , Especificidad por Sustrato
7.
Proc Natl Acad Sci U S A ; 112(36): E4975-84, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26305940

RESUMEN

Calcium-dependent protein kinases (CDPKs) comprise the major group of Ca2+-regulated kinases in plants and protists. It has long been assumed that CDPKs are activated, like other Ca2+-regulated kinases, by derepression of the kinase domain (KD). However, we found that removal of the autoinhibitory domain from Toxoplasma gondii CDPK1 is not sufficient for kinase activation. From a library of heavy chain-only antibody fragments (VHHs), we isolated an antibody (1B7) that binds TgCDPK1 in a conformation-dependent manner and potently inhibits it. We uncovered the molecular basis for this inhibition by solving the crystal structure of the complex and simulating, through molecular dynamics, the effects of 1B7-kinase interactions. In contrast to other Ca2+-regulated kinases, the regulatory domain of TgCDPK1 plays a dual role, inhibiting or activating the kinase in response to changes in Ca2+ concentrations. We propose that the regulatory domain of TgCDPK1 acts as a molecular splint to stabilize the otherwise inactive KD. This dependence on allosteric stabilization reveals a novel susceptibility in this important class of parasite enzymes.


Asunto(s)
Proteínas Quinasas/química , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Toxoplasma/enzimología , Regulación Alostérica , Animales , Anticuerpos Antiprotozoarios/química , Anticuerpos Antiprotozoarios/metabolismo , Anticuerpos Antiprotozoarios/farmacología , Biocatálisis/efectos de los fármacos , Western Blotting , Calcio/metabolismo , Camélidos del Nuevo Mundo , Células Cultivadas , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Simulación de Dinámica Molecular , Mutación , Fosforilación , Unión Proteica , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Toxoplasma/genética
8.
Proc Natl Acad Sci U S A ; 110(18): 7270-5, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23576739

RESUMEN

The epidermal growth factor receptor (EGFR) is a key protein in cellular signaling, and its kinase domain (EGFR kinase) is an intensely pursued target of small-molecule drugs. Although both catalytically active and inactive conformations of EGFR kinase have been resolved crystallographically, experimental characterization of the transitions between these conformations remains difficult. Using unbiased, all-atom molecular dynamics simulations, we observed EGFR kinase spontaneously transition from the active to the so-called "Src-like inactive" conformation by way of two sets of intermediate conformations: One corresponds to a previously identified locally disordered state and the other to previously undescribed "extended" conformations, marked by the opening of the ATP-binding site between the two lobes of the kinase domain. We also simulated the protonation-dependent transition of EGFR kinase to another ["Asp-Phe-Gly-out" ("DFG-out")] inactive conformation and observed similar intermediate conformations. A key element observed in the simulated transitions is local unfolding, or "cracking," which supports a prediction of energy landscape theory. We used hydrogen-deuterium (H/D) exchange measurements to corroborate our simulations and found that the simulated intermediate conformations correlate better with the H/D exchange data than existing active or inactive EGFR kinase crystal structures. The intermediate conformations revealed by our simulations of the transition process differ significantly from the existing crystal structures and may provide unique possibilities for structure-based drug discovery.


Asunto(s)
Biocatálisis , Receptores ErbB/química , Secuencias de Aminoácidos , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Activación Enzimática , Receptores ErbB/metabolismo , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo
9.
PLoS Comput Biol ; 10(7): e1003742, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25058506

RESUMEN

The epidermal growth factor receptor (EGFR) plays a key role in regulating cell proliferation, migration, and differentiation, and aberrant EGFR signaling is implicated in a variety of cancers. EGFR signaling is triggered by extracellular ligand binding, which promotes EGFR dimerization and activation. Ligand-binding measurements are consistent with a negatively cooperative model in which the ligand-binding affinity at either binding site in an EGFR dimer is weaker when the other site is occupied by a ligand. This cooperativity is widely believed to be central to the effects of ligand concentration on EGFR-mediated intracellular signaling. Although the extracellular portion of the human EGFR dimer has been resolved crystallographically, the crystal structures do not reveal the structural origin of this negative cooperativity, which has remained unclear. Here we report the results of molecular dynamics simulations suggesting that asymmetrical interactions of the two binding sites with the membrane may be responsible (perhaps along with other factors) for this negative cooperativity. In particular, in our simulations the extracellular domains of an EGFR dimer spontaneously lay down on the membrane in an orientation in which favorable membrane contacts were made with one of the bound ligands, but could not be made with the other. Similar interactions were observed when EGFR was glycosylated, as it is in vivo.


Asunto(s)
Membrana Celular/química , Receptores ErbB/química , Receptores ErbB/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Animales , Sitios de Unión , Pollos , Biología Computacional , Ligandos , Mamíferos , Simulación de Dinámica Molecular , Conformación Proteica , Pez Cebra
10.
Proc Natl Acad Sci U S A ; 109(27): 10843-8, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22711838

RESUMEN

Members of the RAS small GTPase family regulate cellular responses to extracellular stimuli by mediating the flux through downstream signal transduction cascades. RAS activity is strongly dependent on its subcellular localization and its nucleotide-binding status, both of which are modulated by posttranslational modification. We have determined that RAS is posttranslationally acetylated on lysine 104. Molecular dynamics simulations suggested that this modification affects the conformational stability of the Switch II domain, which is critical for the ability of RAS to interact with guanine nucleotide exchange factors. Consistent with this model, an acetylation-mimetic mutation in K-RAS4B suppressed guanine nucleotide exchange factor-induced nucleotide exchange and inhibited in vitro transforming activity. These data suggest that lysine acetylation is a negative regulatory modification on RAS. Because mutations in RAS family members are extremely common in cancer, modulation of RAS acetylation may constitute a therapeutic approach.


Asunto(s)
Genes ras/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteínas ras/química , Proteínas ras/metabolismo , Acetilación , Animales , Células COS , Chlorocebus aethiops , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisina/metabolismo , Mutagénesis Sitio-Dirigida , Prenilación/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína/fisiología , Relación Estructura-Actividad , Proteínas ras/genética
11.
Proc Natl Acad Sci U S A ; 108(32): 13118-23, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21778406

RESUMEN

How drugs bind to their receptors--from initial association, through drug entry into the binding pocket, to adoption of the final bound conformation, or "pose"--has remained unknown, even for G-protein-coupled receptor modulators, which constitute one-third of all marketed drugs. We captured this pharmaceutically critical process in atomic detail using the first unbiased molecular dynamics simulations in which drug molecules spontaneously associate with G-protein-coupled receptors to achieve final poses matching those determined crystallographically. We found that several beta blockers and a beta agonist all traverse the same well-defined, dominant pathway as they bind to the ß(1)- and ß(2)-adrenergic receptors, initially making contact with a vestibule on each receptor's extracellular surface. Surprisingly, association with this vestibule, at a distance of 15 Å from the binding pocket, often presents the largest energetic barrier to binding, despite the fact that subsequent entry into the binding pocket requires the receptor to deform and the drug to squeeze through a narrow passage. The early barrier appears to reflect the substantial dehydration that takes place as the drug associates with the vestibule. Our atomic-level description of the binding process suggests opportunities for allosteric modulation and provides a structural foundation for future optimization of drug-receptor binding and unbinding rates.


Asunto(s)
Preparaciones Farmacéuticas , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Alprenolol/química , Alprenolol/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Desecación , Espacio Extracelular/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 2/química , Termodinámica
12.
Nat Commun ; 14(1): 1885, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37019905

RESUMEN

Proteins often undergo large conformational changes when binding small molecules, but atomic-level descriptions of such events have been elusive. Here, we report unguided molecular dynamics simulations of Abl kinase binding to the cancer drug imatinib. In the simulations, imatinib first selectively engages Abl kinase in its autoinhibitory conformation. Consistent with inferences drawn from previous experimental studies, imatinib then induces a large conformational change of the protein to reach a bound complex that closely resembles published crystal structures. Moreover, the simulations reveal a surprising local structural instability in the C-terminal lobe of Abl kinase during binding. The unstable region includes a number of residues that, when mutated, confer imatinib resistance by an unknown mechanism. Based on the simulations, NMR spectra, hydrogen-deuterium exchange measurements, and thermostability measurements and estimates, we suggest that these mutations confer imatinib resistance by exacerbating structural instability in the C-terminal lobe, rendering the imatinib-bound state energetically unfavorable.


Asunto(s)
Antineoplásicos , Piperazinas , Mesilato de Imatinib , Piperazinas/farmacología , Pirimidinas/farmacología , Benzamidas , Antineoplásicos/farmacología , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/farmacología , Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl
13.
Proc Natl Acad Sci U S A ; 106(51): 21608-13, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-20007378

RESUMEN

The kinase domain of human epidermal growth factor receptor (HER) 3/ErbB3, a member of the EGF receptor (EGFR) family, lacks several residues that are critical for catalysis. Because catalytic activity in EGFR family members is switched on by an allosteric interaction between kinase domains in an asymmetric kinase domain dimer, HER3 might be specialized to serve as an activator of other EGFR family members. We have determined the crystal structure of the HER3 kinase domain and show that it appears to be locked into an inactive conformation that resembles that of EGFR and HER4. Although the crystal structure shows that the HER3 kinase domain binds ATP, we confirm that it is catalytically inactive but can serve as an activator of the EGFR kinase domain. The HER3 kinase domain forms a dimer in the crystal, mediated by hydrophobic contacts between the N-terminal lobes of the kinase domains. This N-lobe dimer closely resembles a dimer formed by inactive HER4 kinase domains in crystal structures determined previously, and molecular dynamics simulations suggest that the HER3 and HER4 N-lobe dimers are stable. The kinase domains of HER3 and HER4 form similar chains in their respective crystal lattices, in which N-lobe dimers are linked together by reciprocal exchange of C-terminal tails. The conservation of this tiling pattern in HER3 and HER4, which is the closest evolutionary homolog of HER3, might represent a general mechanism by which this branch of the HER receptors restricts ligand-independent formation of active heterodimers with other members of the EGFR family.


Asunto(s)
Receptores ErbB/química , Regulación Alostérica , Secuencia de Aminoácidos , Biocatálisis , Cristalografía por Rayos X , Dimerización , Receptores ErbB/metabolismo , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido
14.
Proc Natl Acad Sci U S A ; 106(1): 139-44, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19109437

RESUMEN

In many protein kinases, a characteristic conformational change (the "DFG flip") connects catalytically active and inactive conformations. Many kinase inhibitors--including the cancer drug imatinib--selectively target a specific DFG conformation, but the function and mechanism of the flip remain unclear. Using long molecular dynamics simulations of the Abl kinase, we visualized the DFG flip in atomic-level detail and formulated an energetic model predicting that protonation of the DFG aspartate controls the flip. Consistent with our model's predictions, we demonstrated experimentally that the kinetics of imatinib binding to Abl kinase have a pH dependence that disappears when the DFG aspartate is mutated. Our model suggests a possible explanation for the high degree of conservation of the DFG motif: that the flip, modulated by electrostatic changes inherent to the catalytic cycle, allows the kinase to access flexible conformations facilitating nucleotide binding and release.


Asunto(s)
Simulación por Computador , Preparaciones Farmacéuticas/química , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/metabolismo , Secuencias de Aminoácidos , Ácido Aspártico , Catálisis , Concentración de Iones de Hidrógeno , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Electricidad Estática
15.
J Mol Biol ; 434(17): 167628, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35595169

RESUMEN

Allostery plays a primary role in regulating protein activity, making it an important mechanism in human disease and drug discovery. Identifying allosteric regulatory sites to explore their biological significance and therapeutic potential is invaluable to drug discovery; however, identification remains a challenge. Allosteric sites are often "cryptic" without clear geometric or chemical features. Since allosteric regulatory sites are often less conserved in protein kinases than the orthosteric ATP binding site, allosteric ligands are commonly more specific than ATP competitive inhibitors. We present a generalizable computational protocol to predict allosteric ligand binding sites based on unbiased ligand binding simulation trajectories. We demonstrate the feasibility of this protocol by revisiting our previously published ligand binding simulations using the first identified viral proto-oncogene, Src kinase, as a model system. The binding paths for kinase inhibitor PP1 uncovered three metastable intermediate states before binding the high-affinity ATP-binding pocket, revealing two previously known allosteric sites and one novel site. Herein, we validate the novel site using a combination of virtual screening and experimental assays to identify a V-type allosteric small-molecule inhibitor that targets this novel site with specificity for Src over closely related kinases. This study provides a proof-of-concept for employing unbiased ligand binding simulations to identify cryptic allosteric binding sites and is widely applicable to other protein-ligand systems.


Asunto(s)
Adenosina Trifosfato , Simulación por Computador , Inhibidores de Proteínas Quinasas , Familia-src Quinasas , Adenosina Trifosfato/química , Regulación Alostérica , Sitio Alostérico , Sitios de Unión , Humanos , Ligandos , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/química
16.
Cell Rep ; 38(5): 110291, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108525

RESUMEN

Effective inactivation of the HER2-HER3 tumor driver has remained elusive because of the challenging attributes of the pseudokinase HER3. We report a structure-function study of constitutive HER2-HER3 signaling to identify opportunities for targeting. The allosteric activation of the HER2 kinase domain (KD) by the HER3 KD is required for tumorigenic signaling and can potentially be targeted by allosteric inhibitors. ATP binding within the catalytically inactive HER3 KD provides structural rigidity that is important for signaling, but this is mimicked, not opposed, by small molecule ATP analogs, reported here in a bosutinib-bound crystal structure. Mutational disruption of ATP binding and molecular dynamics simulation of the apo KD of HER3 identify a conformational coupling of the ATP pocket with a hydrophobic AP-2 pocket, analogous to EGFR, that is critical for tumorigenic signaling and feasible for targeting. The value of these potential target sites is confirmed in tumor growth assays using gene replacement techniques.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Compuestos de Anilina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Nitrilos/farmacología , Quinolinas/farmacología , Receptor ErbB-2/efectos de los fármacos , Transducción de Señal/fisiología
17.
J Am Chem Soc ; 133(24): 9181-3, 2011 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-21545110

RESUMEN

Although the thermodynamic principles that control the binding of drug molecules to their protein targets are well understood, detailed experimental characterization of the process by which such binding occurs has proven challenging. We conducted relatively long, unguided molecular dynamics simulations in which a ligand (the cancer drug dasatinib or the kinase inhibitor PP1) was initially placed at a random location within a box that also contained a protein (Src kinase) to which that ligand was known to bind. In several of these simulations, the ligand correctly identified its target binding site, forming a complex virtually identical to the crystallographically determined bound structure. The simulated trajectories provide a continuous, atomic-level view of the entire binding process, revealing persistent and noteworthy intermediate conformations and shedding light on the role of water molecules. The technique we employed, which does not assume any prior knowledge of the binding site's location, may prove particularly useful in the development of allosteric inhibitors that target previously undiscovered binding sites.


Asunto(s)
Simulación de Dinámica Molecular , Pirazoles/metabolismo , Pirimidinas/metabolismo , Tiazoles/metabolismo , Sitios de Unión , Dasatinib , Unión Proteica , Conformación Proteica , Pirazoles/farmacología , Pirimidinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo
18.
Elife ; 102021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34114565

RESUMEN

Plexins are semaphorin receptors that play essential roles in mammalian neuronal axon guidance and in many other important mammalian biological processes. Plexin signaling depends on a semaphorin-induced dimerization mechanism and is modulated by small GTPases of the Rho family, of which RND1 serves as a plexin activator yet its close homolog RhoD an inhibitor. Using molecular dynamics (MD) simulations, we showed that RND1 reinforces the plexin dimerization interface, whereas RhoD destabilizes it due to their differential interaction with the cell membrane. Upon binding plexin at the Rho-GTPase-binding domain (RBD), RND1 and RhoD interact differently with the inner leaflet of the cell membrane and exert opposite effects on the dimerization interface via an allosteric network involving the RBD, RBD linkers, and a buttress segment adjacent to the dimerization interface. The differential membrane interaction is attributed to the fact that, unlike RND1, RhoD features a short C-terminal tail and a positively charged membrane interface.


Asunto(s)
Semaforinas/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Axones/metabolismo , Humanos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Neuronas/metabolismo , Unión Proteica , Multimerización de Proteína , Transducción de Señal , Proteínas de Unión al GTP rho/química
19.
Nat Commun ; 12(1): 4950, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34400635

RESUMEN

Upon ligand binding, bone morphogenetic protein (BMP) receptors form active tetrameric complexes, comprised of two type I and two type II receptors, which then transmit signals to SMAD proteins. The link between receptor tetramerization and the mechanism of kinase activation, however, has not been elucidated. Here, using hydrogen deuterium exchange mass spectrometry (HDX-MS), small angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations, combined with analysis of SMAD signaling, we show that the kinase domain of the type I receptor ALK2 and type II receptor BMPR2 form a heterodimeric complex via their C-terminal lobes. Formation of this dimer is essential for ligand-induced receptor signaling and is targeted by mutations in BMPR2 in patients with pulmonary arterial hypertension (PAH). We further show that the type I/type II kinase domain heterodimer serves as the scaffold for assembly of the active tetrameric receptor complexes to enable phosphorylation of the GS domain and activation of SMADs.


Asunto(s)
Receptores de Activinas Tipo I/química , Receptores de Activinas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/química , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Transducción de Señal/fisiología , Receptores de Activinas Tipo I/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proteínas Morfogenéticas Óseas/metabolismo , Hipertensión Pulmonar Primaria Familiar/metabolismo , Humanos , Ligandos , Modelos Moleculares , Mutación , Fosforilación , Unión Proteica , Dominios Proteicos , Hipertensión Arterial Pulmonar , Dispersión del Ángulo Pequeño , Transducción de Señal/genética , Proteínas Smad/metabolismo , Difracción de Rayos X
20.
Nat Struct Mol Biol ; 28(10): 847-857, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34625747

RESUMEN

The protein K-Ras functions as a molecular switch in signaling pathways regulating cell growth. In the human mitogen-activated protein kinase (MAPK) pathway, which is implicated in many cancers, multiple K-Ras proteins are thought to assemble at the cell membrane with Ras effector proteins from the Raf family. Here we propose an atomistic structural model for such an assembly. Our starting point was an asymmetric guanosine triphosphate-mediated K-Ras dimer model, which we generated using unbiased molecular dynamics simulations and verified with mutagenesis experiments. Adding further K-Ras monomers in a head-to-tail fashion led to a compact helical assembly, a model we validated using electron microscopy and cell-based experiments. This assembly stabilizes K-Ras in its active state and presents composite interfaces to facilitate Raf binding. Guided by existing experimental data, we then positioned C-Raf, the downstream kinase MEK1 and accessory proteins (Galectin-3 and 14-3-3σ) on and around the helical assembly. The resulting Ras-Raf signalosome model offers an explanation for a large body of data on MAPK signaling.


Asunto(s)
Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/metabolismo , Galectinas/química , Galectinas/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , MAP Quinasa Quinasa 1/metabolismo , Microscopía Electrónica , Microscopía Electrónica de Transmisión , Simulación de Dinámica Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Mutagénesis , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Reproducibilidad de los Resultados , Transducción de Señal , Factores de Transcripción/química , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA