Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
mBio ; 12(5): e0262521, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34700375

RESUMEN

Human cytomegalovirus (HCMV) is a herpesvirus that produces disease in transplant patients and newborn children. Entry of HCMV into cells relies on gH/gL trimer (gHgLgO) and pentamer (gHgLUL128-131) complexes that bind cellular receptors. Here, we studied the structure and interactions of the HCMV trimer, formed by AD169 strain gH and gL and TR strain gO proteins, with the human platelet-derived growth factor receptor alpha (PDGFRα). Three trimer surfaces make extensive contacts with three PDGFRα N-terminal domains, causing PDGFRα to wrap around gO in a structure similar to a human hand, explaining the high-affinity interaction. gO is among the least conserved HCMV proteins, with 8 distinct genotypes. We observed high conservation of residues mediating gO-gL interactions but more extensive gO variability in the PDGFRα interface. Comparisons between our trimer structure and a previously determined structure composed of different subunit genotypes indicate that gO variability is accommodated by adjustments in the gO-PDGFRα interface. We identified two loops within gO that were disordered and apparently glycosylated, which could be deleted without disrupting PDGFRα binding. We also identified four gO residues that contact PDGFRα, which when mutated produced markedly reduced receptor binding. These residues fall within conserved contact sites of gO with PDGFRα and may represent key targets for anti-trimer neutralizing antibodies and HCMV vaccines. Finally, we observe that gO mutations distant from the gL interaction site impact trimer expression, suggesting that the intrinsic folding or stability of gO can impact the efficiency of trimer assembly. IMPORTANCE HCMV is a herpesvirus that infects a large percentage of the adult population and causes significant levels of disease in immunocompromised individuals and birth defects in the developing fetus. The virus encodes a complex protein machinery that coordinates infection of different cell types in the body, including a trimer formed of gH, gL, and gO subunits. Here, we studied the interactions of the HCMV trimer with its receptor on cells, the platelet derived growth factor receptor α (PDGFRα), to better understand how HCMV coordinates virus entry into cells. Our results add to our understanding of HCMV strain-specific differences and identify sites on the trimer that represent potential targets for therapeutic antibodies or vaccine development.


Asunto(s)
Citomegalovirus/metabolismo , Glicoproteínas de Membrana/metabolismo , Multimerización de Proteína/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Microscopía por Crioelectrón/métodos , Citomegalovirus/química , Citomegalovirus/genética , Fibroblastos/virología , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Unión Proteica , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas del Envoltorio Viral/clasificación , Proteínas del Envoltorio Viral/genética , Internalización del Virus
2.
Mol Biosyst ; 12(12): 3651-3665, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27731453

RESUMEN

Multiple sequence alignments (MSAs) are a fundamental analysis tool used throughout biology to investigate relationships between protein sequence, structure, function, evolutionary history, and patterns of disease-associated variants. However, their widespread application in systems biology research is currently hindered by the lack of user-friendly tools to simultaneously visualize, manipulate and query the information conceptualized in large sequence alignments, and the challenges in integrating MSAs with multiple orthogonal data such as cancer variants and post-translational modifications, which are often stored in heterogeneous data sources and formats. Here, we present the Multiple Sequence Alignment Ontology (MSAOnt), which represents a profile or consensus alignment in an ontological format. Subsets of the alignment are easily selected through the SPARQL Protocol and RDF Query Language for downstream statistical analysis or visualization. We have also created the Kinome Viewer (KinView), an interactive integrative visualization that places eukaryotic protein kinase cancer variants in the context of natural sequence variation and experimentally determined post-translational modifications, which play central roles in the regulation of cellular signaling pathways. Using KinView, we identified differential phosphorylation patterns between tyrosine and serine/threonine kinases in the activation segment, a major kinase regulatory region that is often mutated in proliferative diseases. We discuss cancer variants that disrupt phosphorylation sites in the activation segment, and show how KinView can be used as a comparative tool to identify differences and similarities in natural variation, cancer variants and post-translational modifications between kinase groups, families and subfamilies. Based on KinView comparisons, we identify and experimentally characterize a regulatory tyrosine (Y177PLK4) in the PLK4 C-terminal activation segment region termed the P+1 loop. To further demonstrate the application of KinView in hypothesis generation and testing, we formulate and validate a hypothesis explaining a novel predicted loss-of-function variant (D523NPKCß) in the regulatory spine of PKCß, a recently identified tumor suppressor kinase. KinView provides a novel, extensible interface for performing comparative analyses between subsets of kinases and for integrating multiple types of residue specific annotations in user friendly formats.


Asunto(s)
Biología Computacional/métodos , Proteínas Quinasas/química , Proteínas Quinasas/genética , Análisis de Secuencia/métodos , Programas Informáticos , Secuencia de Aminoácidos , Mutación , Fosforilación , Posición Específica de Matrices de Puntuación , Dominios y Motivos de Interacción de Proteínas , Proteína Quinasa C beta/genética , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética
3.
J Med Chem ; 59(17): 7901-14, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27502700

RESUMEN

A series of potent PDGFR inhibitors has been identified. The series was optimized for duration of action in the lung. A novel kinase occupancy assay was used to directly measure target occupancy after i.t. dosing. Compound 25 shows 24 h occupancy of the PDGFR kinase domain, after a single i.t. dose and has efficacy at 0.03 mg/kg, in the rat moncrotaline model of pulmonary arterial hypertension. Examination of PK/PD data from the optimization effort has revealed in vitro:in vivo correlations which link duration of action in vivo with low permeability and high basicity and demonstrate that nonspecific binding to lung tissue increases with lipophilicity.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Hipertensión Pulmonar/tratamiento farmacológico , Niacinamida/análogos & derivados , Pirazoles/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Remodelación Vascular/efectos de los fármacos , Administración por Inhalación , Animales , Línea Celular , Proliferación Celular , Hipertensión Pulmonar/patología , Pulmón/irrigación sanguínea , Membranas Artificiales , Simulación del Acoplamiento Molecular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Niacinamida/síntesis química , Niacinamida/química , Niacinamida/farmacología , Permeabilidad , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/química , Pirazoles/síntesis química , Pirazoles/farmacología , Ratas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/química , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Relación Estructura-Actividad
4.
Tumour Biol ; 37(8): 10053-66, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27193823

RESUMEN

Progress in cancer biology has led to an increasing discovery of oncogenic alterations of the platelet-derived growth factor receptors (PDGFRs) in cancers. In addition, their overexpression in numerous cancers invariably makes PDGFRs and platelet-derived growth factors (PDGFs) prognostic and treatment markers in some cancers. The oncologic alterations of the PDGFR/PDGF system affect the extracellular, transmembrane and tyrosine kinase domains as well as the juxtamembrane segment of the receptor. The receptor is also involved in fusions with intracellular proteins and receptor tyrosine kinase. These discoveries undoubtedly make the system an attractive oncologic therapeutic target. This review covers elementary biology of PDGFR/PDGF system and its role as a prognostic and treatment marker in cancers. In addition, the multifarious therapeutic targets of PDGFR/PDGF system are discussed. Great potential exists in the role of PDGFR/PDGF system as a prognostic and treatment marker and for further exploration of its multifarious therapeutic targets in safe and efficacious management of cancer treatments.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Neoplasias/análisis , Neoplasias/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Transducción de Señal , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Aptámeros de Péptidos/uso terapéutico , Ensayos Clínicos como Asunto , Monitoreo de Drogas , Humanos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Neoplasias/mortalidad , Neoplasias/terapia , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/fisiología , Pronóstico , Isoformas de Proteínas/análisis , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Resultado del Tratamiento
5.
Nanomedicine (Lond) ; 11(6): 581-95, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27003178

RESUMEN

AIM: Glioblastoma multiforme is a devastating disease with no curative options due to the difficulty in achieving sufficient quantities of effective chemotherapies into the tumor past the blood-brain barrier. Micelles loaded with temozolomide (TMZ) were designed to increase the delivery of this drug into the brain. MATERIALS & METHODS: pH-responsive micelles composed of distearoyl phosphoethanolamine-PEG-2000-amine and N-palmitoyl homocysteine were surface-functionalized with PDGF peptide and Dylight 680 fluorophore. RESULTS & CONCLUSION: PDGF-micelles containing TMZ have specific uptake and increased killing in glial cells compared with untargeted micelles. In vivo studies demonstrated selective accumulation of PDGF-micelles containing TMZ in orthotopic gliomas implanted in mice. Targeted micelle-based drug carrier systems hold potential for delivery of a wide variety of hydrophobic drugs thereby reducing its systemic toxicity.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Dacarbazina/análogos & derivados , Preparaciones de Acción Retardada/metabolismo , Glioblastoma/tratamiento farmacológico , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/uso terapéutico , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Dacarbazina/administración & dosificación , Dacarbazina/farmacocinética , Dacarbazina/uso terapéutico , Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones Desnudos , Micelas , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/metabolismo , Protones , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Temozolomida
6.
Cell Signal ; 28(2): 81-93, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26631574

RESUMEN

Platelet derived growth factor receptors (PDGFR) play an important role in tumor pathogenesis and are frequently overexpressed in glioblastoma. Earlier we have shown that only confluent glioblastoma cell cultures exhibit a biphasic calcium transient upon PDGF stimulation. Here, we examined how the change in cell density leads to differential cellular responses to the same PDGF stimulus. PDGF beta receptors and their specific phosphotyrosine residues were fluorescently co-labeled on A172 and T98G glioblastoma cells. The distribution in cell membrane microdomains (lipid rafts) and the phosphorylation state of PDGFR was measured by confocal microscopy and quantitated by digital image processing. Corresponding bulk data were obtained by Western blotting. Activation of relevant downstream signaling pathways was assessed by immunofluorescence in confocal microscopy and by Western blot analysis. Functional outcomes were confirmed with bulk and single cell proliferation assays and motility measurements. In non-confluent (sparse) cultures PDGF-BB stimulation significantly increased phosphorylation of Tyr716 specific for the Ras/MAPK pathway and Tyr751 specific for the phosphoinositide 3-kinase/Akt pathway. As cell monolayers reached confluence, Tyr771 and Tyr1021 were the prominently phosphorylated residues. Tyr771 serves as adaptor for Ras-GAP, which inactivates the MAPK pathway, and Tyr1021 feeds into the phospholipase C-gamma/PKC pathway. Coherent with this, MAPK phosphorylation, Ki-67 positivity and proliferation dominated in dispersed cells, and could be abolished with inhibitors of the MAPK pathway. At the same time, RhoA activation, redistribution of cortactin to leading edges, and increased motility were the prominent output features in confluent cultures. Importantly, the stimulus-evoked confluence-specific changes in the phosphorylation of tyrosine residues occurred mainly in GM1-rich lipid microdomains (rafts). These observations suggest that the same stimulus is able to promote distinctly relevant signaling outputs through a confluence dependent, lipid raft-based regulatory mechanism. In particular, cell division and survival in sparse cultures and inhibition of proliferation and promotion of migration in confluent monolayers. In our model, the ability to switch the final output of the same stimulus as a function of cell density could be a key to the balance of proliferation and invasion in malignant glioblastoma.


Asunto(s)
Movimiento Celular , Proliferación Celular , Glioblastoma/enzimología , Sistema de Señalización de MAP Quinasas , Microdominios de Membrana/enzimología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Línea Celular Tumoral , Glioblastoma/fisiopatología , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Fosfolipasas de Tipo C/metabolismo
7.
Biophys J ; 109(4): 737-49, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287626

RESUMEN

The oncogenic E5 protein from bovine papillomavirus is a short (44 amino acids long) integral membrane protein that forms homodimers. It activates platelet-derived growth factor receptor (PDGFR) ß in a ligand-independent manner by transmembrane helix-helix interactions. The nature of this recognition event remains elusive, as numerous mutations are tolerated in the E5 transmembrane segment, with the exception of one hydrogen-bonding residue. Here, we examined the conformation, stability, and alignment of the E5 protein in fluid lipid membranes of substantially varying bilayer thickness, in both the absence and presence of the PDGFR transmembrane segment. Quantitative synchrotron radiation circular dichroism analysis revealed a very long transmembrane helix for E5 of ∼26 amino acids. Oriented circular dichroism and solid-state (15)N-NMR showed that the alignment and stability of this unusually long segment depend critically on the membrane thickness. When reconstituted alone in exceptionally thick DNPC lipid bilayers, the E5 helix was found to be inserted almost upright. In moderately thick bilayers (DErPC and DEiPC), it started to tilt and became slightly deformed, and finally it became aggregated in conventional DOPC, POPC, and DMPC membranes due to hydrophobic mismatch. On the other hand, when E5 was co-reconstituted with the transmembrane segment of PDGFR, it was able to tolerate even the most pronounced mismatch and was stabilized by binding to the receptor, which has the same hydrophobic length. As E5 is known to activate PDGFR within the thin membranes of the Golgi compartment, we suggest that the intrinsic hydrophobic mismatch of these two interaction partners drives them together. They seem to recognize each other by forming a closely packed bundle of mutually aligned transmembrane helices, which is further stabilized by a specific pair of hydrogen-bonding residues.


Asunto(s)
Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Dicroismo Circular , Escherichia coli , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/química , Espectroscopía de Resonancia Magnética , Conformación Proteica , Estabilidad Proteica
8.
J Invest Dermatol ; 134(4): 921-929, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24304816

RESUMEN

Growth factor-binding domains identified in various extracellular matrix proteins have been shown to regulate growth factor activity in many ways. Recently, we identified a fibronectin peptide (P12) that can bind platelet-derived growth factor BB (PDGF-BB) and promote adult human dermal fibroblast (AHDF) survival under stress. In vivo experiments in a porcine burn injury model showed that P12 limited burn injury progression, suggesting an active role in tissue survival. In this report, we explored the molecular mechanism of this peptide in ADHF under nutrient deprivation. Our results showed that P12 acted like some cell-penetrating peptides in that it redirected ligand-bound PDGF receptor (PDGFR) from the clathrin-dependent endocytic pathway to a slower, macropinocytosis-like pathway. P12 slowed internalization and degradation of PDGF-BB, augmented its survival signals, and promoted cell survival after nutrient removal. Our findings demonstrate a mechanism for a potential therapeutic peptide that increases cell and tissue survival by acting as a cofactor to PDGF-BB.


Asunto(s)
Fibronectinas/química , Péptidos/química , Pinocitosis , Proteínas Proto-Oncogénicas c-sis/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Becaplermina , Supervivencia Celular , Clatrina/química , Progresión de la Enfermedad , Endocitosis , Fibroblastos/metabolismo , Humanos , Ligandos , Células MCF-7 , Modelos Animales , Fosforilación , Transducción de Señal , Piel/citología , Transfección
9.
Front Biosci (Landmark Ed) ; 18(1): 106-19, 2013 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-23276912

RESUMEN

Platelet-derived growth factors (PDGFs) and their receptors are major mitogens for many cell types of mensenchymal origin, including fibroblasts and vascular smooth muscle cells (VSMCs). Their role in enhancing migratory and proliferative responses and extracellular matrix synthesis in these cells, make them key regulators of critical biological functions and in tissue diseases including tissue remodeling, scarring and fibrosis. The activities of the PDGFs have been extensively characterized at the molecular and cellular level and in in vivo model systems. This, in turn, has lead to an increasing number of PDGF-based therapies designed to accelerate or combat defects in tissue repair. This review aims to summarize recent developments in the role of PDGF in key mesenchymal cell functions. Many of the current developments in this field have primarily focused on advancements in understanding cell differentiation, migration, proliferation and the development of emerging PDGF-based therapies and hence will be primary focus of this review.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal/fisiología , Animales , Becaplermina , Remodelación Ósea/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Fibroblastos/fisiología , Humanos , Integrinas/metabolismo , Ligandos , Células Madre Mesenquimatosas/fisiología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Cicatrización de Heridas/fisiología
10.
Biochim Biophys Acta ; 1834(10): 2176-86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23137658

RESUMEN

The four types of platelet-derived growth factors (PDGFs) and the two types of PDGF receptors (PDGFRs, which belong to class III receptor tyrosine kinases) have important functions in the development of connective tissue cells. Recent structural studies have revealed novel mechanisms of PDGFs in propeptide loading and receptor recognition/activation. The detailed structural understanding of PDGF-PDGFR signaling has provided a template that can aid therapeutic intervention to counteract the aberrant signaling of this normally silent pathway, especially in proliferative diseases such as cancer. This review summarizes the advances in the PDGF system with a focus on relating the structural and functional understandings, and discusses the basic aspects of PDGFs and PDGFRs, the mechanisms of activation, and the insights into the therapeutic antagonism of PDGFRs. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.


Asunto(s)
Factor de Crecimiento Derivado de Plaquetas/química , Isoformas de Proteínas/química , Precursores de Proteínas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Plaquetas/química , Plaquetas/citología , Plaquetas/fisiología , Tejido Conectivo/química , Tejido Conectivo/fisiología , Células del Tejido Conectivo/química , Células del Tejido Conectivo/citología , Células del Tejido Conectivo/fisiología , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Factor de Crecimiento Derivado de Plaquetas/clasificación , Factor de Crecimiento Derivado de Plaquetas/fisiología , Unión Proteica , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/fisiología , Precursores de Proteínas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/clasificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal
11.
Mol Cell Proteomics ; 11(5): 15-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22027198

RESUMEN

Stably transfected PC12 cells expressing a chimeric receptor composed of the extracellular domain of the platelet-derived growth factor receptor BB and the transmembrane and intracellular domains of TrkA, the nerve growth factor receptor, were stimulated for 20 min with platelet-derived growth factor and the resulting phosphoproteome was determined from affinity purified tryptic peptides identified by tandem MS (MS/MS) analyses. The changes in the levels of individual phosphorylation sites in stimulated cells versus control were ascertained by the stable isotope labeling of amino acids in cell culture technique. A total of 2035 peptides (806 proteins) were indentified and quantified in both data sets. Of these, 424 phosphopeptides on 259 proteins were found to be up-regulated and 392 sites on 206 proteins were down-regulated (1.8-fold or more). Protein kinases and phosphatases, as well as sites in many proteins involved in G-protein signaling, were prominently represented in the up-regulated group and more than half of the kinase up-regulated phosphosites could be clustered into three sequence motifs; a similar distribution was also found for the down-regulated sites. A comparison of the up-regulated motif profile observed to that calculated from a previous study of the EGFR-induced phosphoproteome in human HeLa cells at the same time point showed a considerable amount of similarity, supporting the view that RTK signal transduction pathways and downstream modifications are likely to be extensively overlapping.


Asunto(s)
Procesamiento Proteico-Postraduccional , Receptor trkA/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Treonina/metabolismo , Secuencias de Aminoácidos , Animales , Becaplermina , Clonación Molecular , Humanos , Marcaje Isotópico , Factor de Crecimiento Nervioso/fisiología , Neuritas/metabolismo , Células PC12 , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilación , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteoma/química , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas , Receptor trkA/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusión/química , Transducción de Señal , Regulación hacia Arriba
12.
Structure ; 19(8): 1127-37, 2011 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-21827948

RESUMEN

Phosphoinositide 3-kinase δ is upregulated in lymphocytic leukemias. Because the p85-regulatory subunit binds to any class IA subunit, it was assumed there is a single universal p85-mediated regulatory mechanism; however, we find isozyme-specific inhibition by p85α. Using deuterium exchange mass spectrometry (DXMS), we mapped regulatory interactions of p110δ with p85α. Both nSH2 and cSH2 domains of p85α contribute to full inhibition of p110δ, the nSH2 by contacting the helical domain and the cSH2 via the C terminus of p110δ. The cSH2 inhibits p110ß and p110δ, but not p110α, implying that p110α is uniquely poised for oncogenic mutations. Binding RTK phosphopeptides disengages the SH2 domains, resulting in exposure of the catalytic subunit. We find that phosphopeptides greatly increase the affinity of the heterodimer for PIP2-containing membranes measured by FRET. DXMS identified regions decreasing exposure at membranes and also regions gaining exposure, indicating loosening of interactions within the heterodimer at membranes.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/química , Lípidos de la Membrana/química , Fosfatidilinositol 3-Quinasas/química , Sustitución de Aminoácidos , Animales , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Medición de Intercambio de Deuterio , Humanos , Liposomas/química , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Propiedades de Superficie
13.
Exp Hematol ; 39(5): 591-600, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21310211

RESUMEN

OBJECTIVE: Platelet-derived growth factor receptors α and ß (PDGFRA, PDGFRB) are frequently expressed on hematopoietic cells and regulate cellular responses such as proliferation, differentiation, survival, and transformation. Stimulation by autocrine loops or activation by chromosomal translocation makes them important factors in development of hematopoietic disorders. Interaction with the ligand PDGF results in activation of the tyrosine kinase domain and phosphorylation of tyrosine residues, thereby creating binding sites for molecules containing Src homology 2 domains. We hypothesized that one such protein may be Lnk, a negative regulator of cytokine receptors, including Mpl, EpoR, c-Kit, and c-Fms. MATERIALS AND METHODS: Interaction of Lnk with PDGFRA, PDGFRB, or leukemogenic FIP1L1-PDGFRA or TEL-PDGFRB was studied in cotransfected 293T cells. Effects of Lnk on PDGFR signaling were shown in 293T and NIH3T3 cells, whereas its influence on either PDGF-dependent or factor-independent growth was investigated using Ba/F3 or 32D cells expressing wild-type PDGFR, FIP1L1-PDGFRA, or TEL-PDGFRB. RESULTS: We show that Lnk binds to PDGFR after exposure of cells to PDGF. Furthermore, Lnk can bind the FIP1L1-PDGFRA fusion protein. Mutation or deletion of the Lnk Src homology 2 domain completely abolished binding of Lnk to FIP1L1-PDGFRA, but just partly prevented binding to PDGFRA or PDGFRB. Expression of Lnk inhibited proliferation of PDGF-dependent Ba/F3 cells and diminished phosphorylation of Erk in PDGF-treated NIH3T3. 32D cells transformed by either FIP1L1-PDGFRA or TEL-PDGFRB stopped growing when Lnk was expressed. CONCLUSIONS: Lnk is a negative regulator of PDGFR signaling. Development of Lnk mimetic drugs might provide a novel therapeutic strategy for myeloproliferative disorders.


Asunto(s)
Proteínas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Unión Proteica , Proteínas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química
14.
Exp Cell Res ; 316(14): 2237-50, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20580638

RESUMEN

Like many other receptor tyrosine kinases (RTKs), platelet-derived growth factor (PDGF) receptor beta (PDGFR-beta) is internalized and degraded in lysosomes in response to PDGF stimulation, which regulates many aspects of cell signalling. However, little is known about the regulation of PDGFR-beta endocytosis. Given that ligand binding is essential for the rapid internalization of RTKs, the events induced by the ligand binding likely contribute to the regulation of ligand-induced RTK internalization. These events include receptor dimerization, activation of intrinsic tyrosine kinase activity and autophosphorylation. In this communication, we examined the role of PDGFR-beta kinase activity, PDGFR-beta dimerization and PDGFR-beta C-terminal motifs in PDGF-induced PDGFR-beta internalization. We showed that inhibition of PDGFR-beta kinase activity by chemical inhibitor or mutation did not block PDGF-induced PDGFR-beta endocytosis, suggesting that the kinase activity is not essential. We further showed that dimerization of PDGFR-beta is essential and sufficient to drive PDGFR-beta internalization independent of PDGFR-beta kinase activation. Moreover, we showed that the previously reported 14 amino acid sequence 952-965 is required for PDGF-induced PDGFR-beta internalization. Most importantly, we showed that this PDGFR-beta internalization motif is exchangeable with the EGFR internalization motif (1005-1017) in mediating ligand-induced internalization of both PDGFR-beta and EGFR. This indicates a common mechanism for the internalization of both PDGFR-beta and EGFR.


Asunto(s)
Endocitosis/fisiología , Receptores ErbB/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Dimerización , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/química , Receptores ErbB/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Fosfotransferasas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Alineación de Secuencia
15.
J Am Chem Soc ; 131(38): 13568-9, 2009 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-19725495

RESUMEN

It is now well-recognized that the spatial localization of proteins at the subcellular level is essential in regulating cell signaling. On the other hand, research efforts aimed at developing synthetic modulators of intracellular signaling processes have usually focused on the affinity and specificity of ligands to their target proteins, neglecting the localization of the molecules in cells. Here we show that it is feasible to rapidly and efficiently activate an endogenous signaling pathway by placing a synthetic ligand at a specific location within a cell. The agonist activity of the ligand is very weak when it is freely diffusing in the cytosol. This study highlights the importance of controlling the subcellular locations of ligand molecules in the design of new synthetic modulators (not only activators but also inhibitors) of endogenous biological events.


Asunto(s)
Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células CHO , Cromonas/farmacología , Cricetinae , Cricetulus , Citosol/efectos de los fármacos , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ligandos , Datos de Secuencia Molecular , Morfolinas/farmacología , Péptidos/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transporte de Proteínas/efectos de los fármacos , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo
16.
Cancer Res ; 69(7): 3032-41, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19318574

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) inhibitors have shown activity in the treatment of acute myelogenous leukemia (AML). Secondary mutations in target kinases can cause clinical resistance to therapeutic kinase inhibition. We have previously shown that sensitivity toward tyrosine kinase inhibitors varies between different activating FLT3 mutations. We therefore intended to determine whether different FLT3 inhibitors would produce distinct profiles of secondary, FLT3 resistance mutations. Using a cell-based screening approach, we generated FLT3-internal tandem duplication (ITD)-expressing cell lines resistant to the FLT3 inhibitors SU5614, PKC412, and sorafenib. Interestingly, the profile of resistance mutations emerging with SU5614 was limited to exchanges in the second part of the kinase domain (TK2) with exchanges of D835 predominating. In contrast, PKC412 exclusively produced mutations within tyrosine kinase domain 1 (TK1) at position N676. A mutation at N676 recently has been reported in a case of PKC412-resistant AML. TK1 mutations exhibited a differential response to SU5614, sorafenib, and sunitinib but strongly impaired response to PKC412. TK2 exchanges identified with SU5614 were sensitive to PKC412, sunitinib, or sorafenib, with the exception of Y842D, which caused a strong resistance to sorafenib. Of note, sorafenib also produced a highly distinct profile of resistance mutations with no overlap to SU5614 or PKC412, including F691L in TK1 and exchanges at position Y842 of TK2. Thus, different FLT3 kinase inhibitors generate distinct, nonoverlapping resistance profiles. This is in contrast to Bcr-Abl kinase inhibitors such as imatinib, nilotinib, and dasatinib, which display overlapping resistance profiles. Therefore, combinations of FLT3 inhibitors may be useful to prevent FLT3 resistance mutations in the setting of FLT3-ITD-positive AML.


Asunto(s)
Mutación , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética , Animales , Bencenosulfonatos/farmacología , Línea Celular , Resistencia a Medicamentos , Indoles/farmacología , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Estructura Terciaria de Proteína , Piridinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Sorafenib , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Secuencias Repetidas en Tándem , Tirosina Quinasa 3 Similar a fms/química
17.
Nat Struct Mol Biol ; 15(10): 1109-18, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18794843

RESUMEN

Protein kinases targeted by small-molecule inhibitors develop resistance through mutation of the 'gatekeeper' threonine residue of the active site. Here we show that the gatekeeper mutation in the cellular forms of c-ABL, c-SRC, platelet-derived growth factor receptor-alpha and -beta, and epidermal growth factor receptor activates the kinase and promotes malignant transformation of BaF3 cells. Structural analysis reveals that a network of hydrophobic interactions-the hydrophobic spine-characteristic of the active kinase conformation is stabilized by the gatekeeper substitution. Substitution of glycine for the residues constituting the spine disrupts the hydrophobic connectivity and inactivates the kinase. Furthermore, a small-molecule inhibitor that maximizes complementarity with the dismantled spine (compound 14) inhibits the gatekeeper mutation of BCR-ABL-T315I. These results demonstrate that mutation of the gatekeeper threonine is a common mechanism of activation for tyrosine kinases and provide structural insights to guide the development of next-generation inhibitors.


Asunto(s)
Mutación/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Treonina/genética , Treonina/metabolismo , Sitios de Unión , Proteína Tirosina Quinasa CSK , Línea Celular , Cristalografía por Rayos X , Activación Enzimática/genética , Receptores ErbB/química , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Familia-src Quinasas
18.
Proc Natl Acad Sci U S A ; 105(29): 9959-64, 2008 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-18632564

RESUMEN

The production of reactive oxygen species (ROS) exerts an additional tier of control over tyrosine phosphorylation-dependent signal transduction by transiently inhibiting the catalytic activity of specific protein tyrosine phosphatases (PTPs). Hence, the ability to detect reversible oxidation of PTPs in vivo is critical to understanding the complex biological role of ROS in the control of cellular signaling. Here, we describe an assay for identifying those PTPs that are reversibly oxidized in vivo, which utilizes the unique chemistry of the invariant catalytic Cys residue in labeling the active site with biotinylated small molecules under mildly acidic conditions. We have applied this cysteinyl-labeling assay to the study of platelet-derived growth factor (PDGF) receptor signaling in an angiomyolipoma cell model. Doing so has allowed us to detect reversible oxidation of several proteins in response to sustained PDGF stimulation. As in other cell systems, we have observed the reversible oxidation of the classical PTP SHP2 and the tumor suppressor phosphatase PTEN in response to PDGF stimulation. Furthermore, we detected reversible oxidation of members of two other subclasses of PTPs, the receptor PTP LAR and the dual-specificity phosphatase MKP1. These data demonstrate the broad selectivity of the assay, allowing us to detect representatives of all of the major subgroups of the PTP superfamily. We anticipate that this cysteinyl-labeling enrichment strategy can be applied broadly to study reversible oxidation as a mechanism of harnessing PTP catalytic activity in a variety of signaling pathways.


Asunto(s)
Angiomiolipoma/enzimología , Neoplasias Renales/enzimología , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Angiomiolipoma/genética , Angiomiolipoma/metabolismo , Becaplermina , Dominio Catalítico , Línea Celular Tumoral , Cisteína/química , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Oxidación-Reducción , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Especies Reactivas de Oxígeno/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Transformación Genética
19.
Anal Chem ; 80(5): 1497-504, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18237154

RESUMEN

This study describes the first instance of the use of two differently sized Au nanoparticles (Au NPs), acting separately as donor and acceptor, in homogeneous photoluminescence quenching assays developed for the analysis of proteins. Introduction of a breast cancer marker protein, platelet-derived growth factor AA (PDGF AA), to a solution of 11-mercaptoundecanoic acid-protected, 2.0-nm photoluminescent Au nanodots (L(AuND)) led to the preparation of PDGF AA-L(AuND) as the donor. Thiol-derivative PDGF binding aptamers (Apt) and 13-nm spherical Au NPs were used to synthesize the Apt-Q(AuNP) acceptor. The photoluminescence of PDGF AA-L(AuND) at 520 nm decreased when photoluminescence quenching occurred between Apt-Q(AuNP) and PDGF AA-L(AuND). We used the PDGF AA-L(AuND)/Apt-Q(AuNP)-based molecular light switching system to analyze PDGFs and PDGF alpha-receptor in separate homogeneous solutions. In the presence of PDGFs, the interaction between Apt-Q(AuNP) and PDGF AA-L(AuND) decreased as a result of competitive reactions between the PDGFs and Apt-Q(AuNP). Similarly, the interaction between Apt-Q(AuNP) and PDGF AA-L(AuND) reduced as a result of competitive reactions between PDGF alpha-receptor and PDGF AA-L(AuND). The limits of detection (LODs) for PDGF AA and PDGF alpha-receptor were 80 pM and 0.25 nM, respectively, resulting from a low background photoluminescence signal. When using the Apt-Q(AuNP) as selectors for (a) the enrichment of PDGF AA and (b) the removal of matrixes possessing intense background fluorescence from cell media and urine samples, the LOD for PDGF AA decreased to 10 pM. Unlike quantum dots, the L(AuND) provide the advantages of biocompatibility, ease of bioconjugation, and minimal toxicity.


Asunto(s)
Oro/química , Luminiscencia , Nanoestructuras/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Trombina/análisis , Línea Celular Tumoral , Humanos , Óptica y Fotónica , Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Trombina/química
20.
Org Biomol Chem ; 4(12): 2376-86, 2006 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-16763682

RESUMEN

Platelet-derived growth factor (PDGF) and its receptor PDGFR are required for tumor growth and angiogenesis, so disruption of the PDGF-PDGFR interaction should lead to starvation of tumors and reduction of tumor growth. Potent PDGF antagonists have been discovered through the synthesis of a series of calix[4]arene-based compounds that are designed to bind to the three-loop region of PDGF. The effect of lower-rim alkylation, linker and number of interacting head groups on the calix[4]arene scaffold on PDGF affinity and cellular activity has been investigated.


Asunto(s)
Calixarenos/química , Calixarenos/farmacología , Fenoles/química , Fenoles/farmacología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Femenino , Humanos , Cinética , Ratones , Ratones Desnudos , Modelos Moleculares , Datos de Secuencia Molecular , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Unión Proteica , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA