Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
mBio ; 12(5): e0262521, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34700375

ABSTRACT

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.


Subject(s)
Cytomegalovirus/metabolism , Membrane Glycoproteins/metabolism , Protein Multimerization/physiology , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Cryoelectron Microscopy/methods , Cytomegalovirus/chemistry , Cytomegalovirus/genetics , Fibroblasts/virology , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Protein Binding , Receptors, Platelet-Derived Growth Factor/genetics , Viral Envelope Proteins/classification , Viral Envelope Proteins/genetics , Virus Internalization
2.
Methods Mol Biol ; 1575: 215-222, 2017.
Article in English | MEDLINE | ID: mdl-28255883

ABSTRACT

There is a need for a general reporter system to study the interactions between a ligand and membrane protein in living cells. Here, we demonstrate a method that allows identification of undiscovered ligand partners of a membrane protein in its natural milieu. This assay will be of importance especially for multiple pass transmembrane proteins such as ion channels and GPCRs.


Subject(s)
Protein Interaction Mapping/methods , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , HEK293 Cells , Humans , Ligands , Protein Binding , Receptors, Platelet-Derived Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
3.
Mol Biosyst ; 12(12): 3651-3665, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27731453

ABSTRACT

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.


Subject(s)
Computational Biology/methods , Protein Kinases/chemistry , Protein Kinases/genetics , Sequence Analysis/methods , Software , Amino Acid Sequence , Mutation , Phosphorylation , Position-Specific Scoring Matrices , Protein Interaction Domains and Motifs , Protein Kinase C beta/genetics , Protein Kinases/metabolism , Protein Processing, Post-Translational , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/genetics
4.
J Med Chem ; 59(17): 7901-14, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27502700

ABSTRACT

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.


Subject(s)
Airway Remodeling/drug effects , Hypertension, Pulmonary/drug therapy , Niacinamide/analogs & derivatives , Pyrazoles/chemistry , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Vascular Remodeling/drug effects , Administration, Inhalation , Animals , Cell Line , Cell Proliferation , Hypertension, Pulmonary/pathology , Lung/blood supply , Membranes, Artificial , Molecular Docking Simulation , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Niacinamide/chemical synthesis , Niacinamide/chemistry , Niacinamide/pharmacology , Permeability , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Rats , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/chemistry , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Structure-Activity Relationship
5.
Tumour Biol ; 37(8): 10053-66, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27193823

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasm Proteins/analysis , Neoplasms/chemistry , Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/analysis , Signal Transduction , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Aptamers, Peptide/therapeutic use , Clinical Trials as Topic , Drug Monitoring , Humans , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Neoplasms/mortality , Neoplasms/therapy , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/physiology , Prognosis , Protein Isoforms/analysis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , RNA Interference , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/physiology , Treatment Outcome
6.
Nanomedicine (Lond) ; 11(6): 581-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27003178

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Brain/drug effects , Dacarbazine/analogs & derivatives , Delayed-Action Preparations/metabolism , Glioblastoma/drug therapy , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/therapeutic use , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/pharmacokinetics , Dacarbazine/therapeutic use , Delayed-Action Preparations/chemistry , Drug Delivery Systems , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice, Nude , Micelles , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Protons , Receptors, Platelet-Derived Growth Factor/chemistry , Temozolomide
7.
Cell Signal ; 28(2): 81-93, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26631574

ABSTRACT

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.


Subject(s)
Cell Movement , Cell Proliferation , Glioblastoma/enzymology , MAP Kinase Signaling System , Membrane Microdomains/enzymology , Receptors, Platelet-Derived Growth Factor/metabolism , Cell Line, Tumor , Glioblastoma/physiopathology , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Platelet-Derived Growth Factor/chemistry , Type C Phospholipases/metabolism
8.
Biophys J ; 109(4): 737-49, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287626

ABSTRACT

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.


Subject(s)
Receptors, Platelet-Derived Growth Factor/chemistry , Circular Dichroism , Escherichia coli , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers/chemistry , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Stability
9.
J Invest Dermatol ; 134(4): 921-929, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24304816

ABSTRACT

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.


Subject(s)
Fibronectins/chemistry , Peptides/chemistry , Pinocytosis , Proto-Oncogene Proteins c-sis/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Becaplermin , Cell Survival , Clathrin/chemistry , Disease Progression , Endocytosis , Fibroblasts/metabolism , Humans , Ligands , MCF-7 Cells , Models, Animal , Phosphorylation , Signal Transduction , Skin/cytology , Transfection
10.
Front Biosci (Landmark Ed) ; 18(1): 106-19, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23276912

ABSTRACT

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.


Subject(s)
Platelet-Derived Growth Factor/physiology , Signal Transduction/physiology , Animals , Becaplermin , Bone Remodeling/physiology , Cell Movement/physiology , Cell Proliferation , Fibroblasts/physiology , Humans , Integrins/metabolism , Ligands , Mesenchymal Stem Cells/physiology , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/therapeutic use , Protein Multimerization , Proto-Oncogene Proteins c-sis/metabolism , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/physiology , Wound Healing/physiology
11.
Biochim Biophys Acta ; 1834(10): 2176-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23137658

ABSTRACT

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.


Subject(s)
Platelet-Derived Growth Factor/chemistry , Protein Isoforms/chemistry , Protein Precursors/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Blood Platelets/chemistry , Blood Platelets/cytology , Blood Platelets/physiology , Connective Tissue/chemistry , Connective Tissue/physiology , Connective Tissue Cells/chemistry , Connective Tissue Cells/cytology , Connective Tissue Cells/physiology , Gene Expression Regulation , Humans , Models, Molecular , Platelet-Derived Growth Factor/classification , Platelet-Derived Growth Factor/physiology , Protein Binding , Protein Isoforms/classification , Protein Isoforms/physiology , Protein Precursors/metabolism , Receptors, Platelet-Derived Growth Factor/classification , Receptors, Platelet-Derived Growth Factor/physiology , Signal Transduction
12.
J Biol Chem ; 287(44): 37420-33, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955284

ABSTRACT

Nectins belong to a family of immunoglobulin (Ig)-like cell-adhesion molecules comprising four members, nectin-1 through nectin-4. Nectins are involved in formation of the mechanical adhesive puncta adherentia junctions of synapses. Nectins share the same overall structural topology with an extracellular region containing three Ig modules, a transmembrane region, and a cytoplasmic region. In nectin-1, the first and second Ig module in the extracellular region are necessary for the trans-interaction with nectin-3 and formation of cis-dimers, respectively. The function of the third Ig module of nectin-1 remains unknown. We here report the structure in solution of the third, membrane-proximal Ig module of mouse nectin-1 (nectin-1 Ig3) solved by means of nuclear magnetic resonance (NMR) spectroscopy. It belongs to the C1 set of the Ig superfamily. Nectin-1 Ig3 was produced as a recombinant protein and induced neurite outgrowth in primary cultures of hippocampal and cerebellar granule neurons, an effect abolished by treatment with the fibroblast growth factor receptor (FGFR) inhibitor SU5402, or by transfection with a dominant-negative FGFR1 construct. We showed by surface plasmon resonance (SPR) analysis that nectin-1 Ig3 directly interacted with various isoforms of FGFR. Nectin-1 Ig3 induced phosphorylation of FGFR1c in the same manner as the whole nectin-1 ectodomain, and promoted survival of cerebellar granule neurons induced to undergo apoptosis. Finally, we constructed a peptide, nectide, by employing in silico modeling of various FGFR ligand-binding sites. Nectide mimicked all the effects of nectin-1 Ig3. We suggest that FGFR is a downstream signaling partner of nectin-1.


Subject(s)
Cell Adhesion Molecules/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Survival , Crystallography, X-Ray , Fibroblast Growth Factor 2/physiology , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Molecular Sequence Data , Nectins , Neurites/metabolism , Neurites/physiology , Neurons/cytology , Neurons/physiology , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Signal Transduction , Surface Plasmon Resonance
13.
Mol Cell Proteomics ; 11(5): 15-30, 2012 May.
Article in English | MEDLINE | ID: mdl-22027198

ABSTRACT

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.


Subject(s)
Protein Processing, Post-Translational , Receptor, trkA/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Recombinant Fusion Proteins/metabolism , Serine/metabolism , Threonine/metabolism , Amino Acid Motifs , Animals , Becaplermin , Cloning, Molecular , Humans , Isotope Labeling , Nerve Growth Factor/physiology , Neurites/metabolism , PC12 Cells , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Protein Interaction Maps , Protein Structure, Tertiary , Proteome/chemistry , Proteome/metabolism , Proto-Oncogene Proteins c-sis/physiology , Rats , Receptor, trkA/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Signal Transduction , Up-Regulation
14.
Structure ; 19(8): 1127-37, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21827948

ABSTRACT

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.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/chemistry , Membrane Lipids/chemistry , Phosphatidylinositol 3-Kinases/chemistry , Amino Acid Substitution , Animals , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase/genetics , Deuterium Exchange Measurement , Humans , Liposomes/chemistry , Mice , Models, Molecular , Mutagenesis, Site-Directed , Peptide Fragments , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Receptors, Platelet-Derived Growth Factor/chemistry , Surface Properties
15.
Biochem Biophys Res Commun ; 410(1): 134-9, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21651892

ABSTRACT

Prestin, a multipass transmembrane protein whose N- and C-termini are localized to the cytoplasm, must be trafficked to the plasma membrane to fulfill its cellular function as a molecular motor. One challenge in studying prestin sequence-function relationships within living cells is separating the effects of amino acid substitutions on prestin trafficking, plasma membrane localization and function. To develop an approach for directly assessing prestin levels at the plasma membrane, we have investigated whether fusion of prestin to a single pass transmembrane protein results in a functional fusion protein with a surface-exposed N-terminal tag that can be detected in living cells. We find that fusion of the biotin-acceptor peptide (BAP) and transmembrane domain of the platelet-derived growth factor receptor (PDGFR) to the N-terminus of prestin-GFP yields a membrane protein that can be metabolically-labeled with biotin, trafficked to the plasma membrane, and selectively detected at the plasma membrane using fluorescently-tagged streptavidin. Furthermore, we show that the addition of a surface detectable tag and a single-pass transmembrane domain to prestin does not disrupt its voltage-sensitive activity.


Subject(s)
Anion Transport Proteins/analysis , Biotinylation , Cell Membrane/chemistry , Recombinant Fusion Proteins/analysis , Amino Acid Sequence , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Biotin/chemistry , Biotin/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Patch-Clamp Techniques , Protein Structure, Tertiary , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sulfate Transporters
16.
Exp Hematol ; 39(5): 591-600, 2011 May.
Article in English | MEDLINE | ID: mdl-21310211

ABSTRACT

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.


Subject(s)
Proteins/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins , Mice , NIH 3T3 Cells , Protein Binding , Proteins/chemistry , Receptors, Platelet-Derived Growth Factor/chemistry
17.
Exp Cell Res ; 316(14): 2237-50, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20580638

ABSTRACT

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.


Subject(s)
Endocytosis/physiology , ErbB Receptors/metabolism , Hydrophobic and Hydrophilic Interactions , Receptors, Platelet-Derived Growth Factor/metabolism , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , Cell Line , Dimerization , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/chemistry , ErbB Receptors/genetics , Flow Cytometry , Fluorescent Antibody Technique , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphotransferases/metabolism , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/genetics , Sequence Alignment
18.
Proc Natl Acad Sci U S A ; 107(5): 1906-11, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080685

ABSTRACT

Structural analyses of the extracellular region of stem cell factor (SCF) receptor (also designated KIT) in complex with SCF revealed a sequence motif in a loop in the fourth Ig-like domain (D4) that is responsible for forming homotypic receptor contacts and for ligand-induced KIT activation and cell signaling. An identical motif was identified in the most membrane-proximal seventh Ig-like domain (D7) of vascular endothelial growth factor receptor 1 (VEGFR1), VEGFR2, and VEGFR3. In this report we demonstrate that ligand-induced tyrosine autophosphorylation and cell signaling via VEGFR1 or VEGFR2 harboring mutations in critical residues (Arg726 or Asp731) in D7 are strongly impaired. We also describe the crystal structure of D7 of VEGFR2 to a resolution of 2.7 A. The structure shows that homotypic D7 contacts are mediated by salt bridges and van der Waals contacts formed between Arg726 of one protomer and Asp731 of the other protomer. The structure of D7 dimer is very similar to the structure of D4 dimers seen in the crystal structure of KIT extracellular region in complex with SCF. The high similarity between VEGFR D7 and KIT D4 in both structure and function provides further evidence for common ancestral origins of type III and type V RTKs. It also reveals a conserved mechanism for RTK activation and a novel target for pharmacological intervention of pathologically activated RTKs.


Subject(s)
Receptors, Vascular Endothelial Growth Factor/chemistry , Receptors, Vascular Endothelial Growth Factor/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Conserved Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Phylogeny , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Static Electricity , Tyrosine/chemistry , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
J Am Chem Soc ; 131(38): 13568-9, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19725495

ABSTRACT

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.


Subject(s)
Peptides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Amino Acid Sequence , Animals , CHO Cells , Chromones/pharmacology , Cricetinae , Cricetulus , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Ligands , Molecular Sequence Data , Morpholines/pharmacology , Peptides/chemistry , Phosphoinositide-3 Kinase Inhibitors , Protein Transport/drug effects , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/metabolism
20.
Anal Chem ; 81(18): 7795-800, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19691297

ABSTRACT

Tunable aptamer capillary electrophoresis (CE) techniques were developed to enable the separation and detection of platelet derived growth factor (PDGF) isomers and their receptors. Using an aptamer that formed a stable complex with the B chain but not with the A chain of PDGF, we were able to tweak the electrophoretic mobilities of the PDGF isomers for their separation. PDGF-AB bound to a single aptamer molecule was well resolved from PDGF-BB bound to two aptamer molecules. Simultaneous determination of 50 pM of two isomers was accomplished in a single analysis. Furthermore, PDGF-AB was used as a connector to bring receptor alpha and fluorescent aptamer into a single complex molecule. As a result, the formation of a (receptor alpha)-(PDGF-AB)-(fluorescent aptamer) ternary complex enabled the detection of the receptor alpha by tunable aptamer CE. A competitive assay was developed to determine receptor beta, making use of the competition between the receptor beta and fluorescent aptamer in binding to PDGF-BB. Detection limits were 0.5 nM for PDGF receptor alpha and 3 nM for receptor beta. Determination of PDGF isomers and their receptors in diluted serum samples showed no interference from the sample matrix.


Subject(s)
Aptamers, Nucleotide/metabolism , Electrophoresis, Capillary/methods , Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/analysis , Aptamers, Nucleotide/chemistry , Fluorescent Dyes/chemistry , Platelet-Derived Growth Factor/chemistry , Protein Isoforms/analysis , Protein Isoforms/chemistry , Receptors, Platelet-Derived Growth Factor/blood , Receptors, Platelet-Derived Growth Factor/chemistry , Recombinant Proteins/analysis , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...