Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters










Publication year range
1.
Blood Adv ; 7(13): 3036-3048, 2023 07 11.
Article in English | MEDLINE | ID: mdl-36735416

ABSTRACT

Activated protein C (APC) is a pleiotropic coagulation protease with anticoagulant, anti-inflammatory, and cytoprotective activities. Selective modulation of these APC activities contributes to our understanding of the regulation of these physiological mechanisms and permits the development of therapeutics for the pathologies associated with these pathways. An antibody library targeting the nonactive site of APC was generated using llama antibodies (nanobodies). Twenty-one nanobodies were identified that selectively recognize APC compared with the protein C zymogen. Overall, 3 clusters of nanobodies were identified based on the competition for APC in biolayer interferometry studies. APC functional assays for anticoagulant activity, histone H3 cleavage, and protease-activated receptor 1 (PAR1) cleavage were used to understand their diversity. These functional assays revealed 13 novel nanobody-induced APC activity profiles via the selective modulation of APC pleiotropic activities, with the potential to regulate specific mechanisms for therapeutic purposes. Within these, 3 nanobodies (LP2, LP8, and LP17) inhibited all 3 APC functions. Four nanobodies (LP1, LP5, LP16, and LP20) inhibited only 2 of the 3 functions. Monofunction inhibition specific to APC anticoagulation activity was observed only by 2 nanobodies (LP9 and LP11). LP11 was also found to shift the ratio of APC cleavage of PAR1 at R46 relative to R41, which results in APC-mediated biased PAR1 signaling and APC cytoprotective effects. Thus, LP11 has an activity profile that could potentially promote hemostasis and cytoprotection in bleedings associated with hemophilia or coagulopathy by selectively modulating APC anticoagulation and PAR1 cleavage profile.


Subject(s)
Protein C , Single-Domain Antibodies , Protein C/metabolism , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/metabolism , Endothelial Cells/metabolism , Anticoagulants/pharmacology , Antibodies/pharmacology
2.
Sci Rep ; 11(1): 16170, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373558

ABSTRACT

Proteinase-activated receptor-1 (PAR1), triggered by thrombin and other serine proteinases such as tissue kallikrein-4 (KLK4), is a key driver of inflammation, tumor invasiveness and tumor metastasis. The PAR1 transmembrane G-protein-coupled receptor therefore represents an attractive target for therapeutic inhibitors. We thus used a computational design to develop a new PAR1 antagonist, namely, a catalytically inactive human KLK4 that acts as a proteinase substrate-capture reagent, preventing receptor cleavage (and hence activation) by binding to and occluding the extracellular R41-S42 canonical PAR1 proteolytic activation site. On the basis of in silico site-saturation mutagenesis, we then generated KLK4S207A,L185D, a first-of-a-kind 'decoy' PAR1 inhibitor, by mutating the S207A and L185D residues in wild-type KLK4, which strongly binds to PAR1. KLK4S207A,L185D markedly inhibited PAR1 cleavage, and PAR1-mediated MAPK/ERK activation as well as the migration and invasiveness of melanoma cells. This 'substrate-capturing' KLK4 variant, engineered to bind to PAR1, illustrates proof of principle for the utility of a KLK4 'proteinase substrate capture' approach to regulate proteinase-mediated PAR1 signaling.


Subject(s)
Kallikreins/metabolism , Receptor, PAR-1/antagonists & inhibitors , Amino Acid Substitution , Binding Sites , Cell Line, Tumor , Computer Simulation , Drug Design , Humans , Kallikreins/chemistry , Kallikreins/genetics , Kinetics , MCF-7 Cells , Mutagenesis, Site-Directed , Neoplasm Invasiveness/prevention & control , Protein Engineering , Protein Interaction Domains and Motifs , Proteolysis , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Substrate Specificity , Thrombin/metabolism
3.
Eur Rev Med Pharmacol Sci ; 24(9): 5111-5117, 2020 05.
Article in English | MEDLINE | ID: mdl-32432775

ABSTRACT

OBJECTIVE: Parstatin, the N-terminal 41-amino-acid peptide cleaved by thrombin from protease-activated-receptor 1, was shown to be highly effective in preventing ocular angiogenesis and as such it has potential therapeutic applications in ocular neovascular diseases. In the frame of a safety program in preclinical development, we investigated whether parstatin exerts any cytotoxic effect in critical ocular cell populations. MATERIALS AND METHODS: Human retinal pigment epithelium cell-19 line (ARPE-19) and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay were used to investigate parstatin's effect in cell cultures. Parstatin 24-41 was used as a control peptide which lacks the hydrophobic domain to demonstrate the specificity and the structure-dependent effect of parstatin. Both peptides were used at concentrations ranging from 0.1-30 µM for 24, 48 and 72 hours. RESULTS: In the presence of parstatin the rate of ARPE-19 cell growth and viability were significantly decreased in a concentration-dependent and time-dependent manner, with an IC50 of approximately 10 µM. When ARPE-19 cells were treated with parstatin 24-41 no inhibitory effect was observed at any concentration or exposure time used. CONCLUSIONS: Parstatin has a clear detrimental effect on the viability of ARPE-19 cells and raises concerns about its use in the eye because of its possible toxic effects.


Subject(s)
Peptide Fragments/adverse effects , Peptide Fragments/toxicity , Retinal Pigment Epithelium/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Peptide Fragments/chemistry , Receptor, PAR-1/chemistry , Retinal Pigment Epithelium/metabolism
4.
Structure ; 27(10): 1517-1526.e3, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31422910

ABSTRACT

G protein-coupled receptors (GPCRs) show complex relationships between functional states and conformational plasticity that can be qualitatively and quantitatively described by contouring their free energy landscape. However, how ligands modulate the free energy landscape to direct conformation and function of GPCRs is not entirely understood. Here, we employ single-molecule force spectroscopy to parametrize the free energy landscape of the human protease-activated receptor 1 (PAR1), and delineate the mechanical, kinetic, and energetic properties of PAR1 being set into different functional states. Whereas in the inactive unliganded state PAR1 adopts mechanically rigid and stiff conformations, upon agonist or antagonist binding the receptor mechanically softens, while increasing its conformational flexibility, and kinetic and energetic stability. By mapping the free energy landscape to the PAR1 structure, we observe key structural regions putting this conformational plasticity into effect. Our insight, complemented with previously acquired knowledge on other GPCRs, outlines a more general framework to understand how GPCRs stabilize certain functional states.


Subject(s)
Guanidines/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Binding Sites , Guanidines/chemistry , Humans , Ligands , Models, Molecular , Oligopeptides/chemistry , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Secondary , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Single Molecule Imaging
5.
Biochemistry ; 58(8): 1048-1060, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30672691

ABSTRACT

Thrombin, derived from zymogen prothrombin (ProT), is a serine protease involved in procoagulation, anticoagulation, and platelet activation. Thrombin's actions are regulated through anion-binding exosites I and II (ABE I and ABE II) that undergo maturation during activation. Mature ABEs can utilize exosite-based communication to fulfill thrombin functions. However, the conformational basis behind such long-range communication and the resultant ligand binding affinities are not well understood. Protease activated receptors (PARs), involved in platelet activation and aggregation, are known to target thrombin ABE I. Unexpectedly, PAR3 (44-56) can already bind to pro-ABE I of ProT. Nuclear magnetic resonance (NMR) ligand-enzyme titrations were used to characterize how individual PAR1 (49-62) residues interact with pro-ABE I and mature ABE I. 1D proton line broadening studies demonstrated that binding affinities for native PAR1P (49-62, P54) and for the weak binding variant PAR1G (49-62, P54G) increased as ProT was converted to mature thrombin. 1H,15N-HSQC titrations revealed that PAR1G residues K51, E53, F55, D58, and E60 exhibited less affinity to pro-ABE I than comparable residues in PAR3G (44-56, P51G). Individual PAR1G residues then displayed tighter binding upon exosite maturation. Long-range communication between thrombin exosites was examined by saturating ABE II with phosphorylated GpIbα (269-282, 3Yp) and monitoring the binding of PAR1 and PAR3 peptides to ABE I. Individual PAR residues exhibited increased affinities in this dual-ligand environment supporting the presence of interexosite allostery. Exosite maturation and beneficial long-range allostery are proposed to help stabilize an ABE I conformation that can effectively bind PAR ligands.


Subject(s)
Anions/chemistry , Peptide Fragments/metabolism , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Thrombin/metabolism , Binding Sites , Humans , Ligands , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Thrombin/chemistry
6.
Bioconjug Chem ; 29(11): 3783-3792, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30362700

ABSTRACT

Nature has evolved mechanisms to increase the specificity of enzymes and enhance their activity to support particular biological functions. Mimicking these mechanisms with artificial systems, such as nanoparticles, would greatly benefit bioanalysis. Here, we have taken inspiration from platelet cells to create a fluorescent nanoparticle probe with enhanced sensitivity toward its protease target. Platelets use protease-activated receptor 1 (PAR1) to enhance thrombin activity that initiates their aggregation in the early stages of blood clotting. We therefore coconjugated multiple copies each of a peptide substrate and a fragment of PAR1 to a semiconductor quantum dot (QD) to mimic this behavior. Thrombin activity toward conjugates with and without the PAR1 fragment were tracked via Förster resonance energy transfer (FRET). The co-conjugated PAR1 increased thrombin-catalyzed hydrolysis of the substrate by several-fold up to multiple orders of magnitude, albeit dependent on the surface chemistry of the QD. The enhancement effect arose from a combination of selective binding between the PAR1 fragment and thrombin, and from colocalization of the substrate and the PAR1 fragment at the QD interface. Substrate-receptor co-conjugation is thus a promising strategy for the rational design of nanoparticle bioconjugates with optimized sensitivity and specificity for biosensing and imaging.


Subject(s)
Fluorescent Dyes/chemistry , Peptides/chemistry , Quantum Dots/chemistry , Receptor, PAR-1/chemistry , Thrombin/chemistry , Amino Acid Sequence , Biomimetic Materials/chemistry , Blood Platelets/chemistry , Humans , Models, Molecular , Proteolysis , Quantum Dots/ultrastructure
7.
Biochemistry ; 57(39): 5748-5758, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30102523

ABSTRACT

The pathways that G protein-coupled receptor (GPCR) ligands follow as they bind to or dissociate from their receptors are largely unknown. Protease-activated receptor-1 (PAR1) is a GPCR activated by intramolecular binding of a tethered agonist peptide that is exposed by thrombin cleavage. By contrast, the PAR1 antagonist vorapaxar is a lipophilic drug that binds in a pocket almost entirely occluded from the extracellular solvent. The binding and dissociation pathway of vorapaxar is unknown. Starting with the crystal structure of vorapaxar bound to PAR1, we performed temperature-accelerated molecular dynamics simulations of ligand dissociation. In the majority of simulations, vorapaxar exited the receptor laterally into the lipid bilayer through openings in the transmembrane helix (TM) bundle. Prior to full dissociation, vorapaxar paused in metastable intermediates stabilized by interactions with the receptor and lipid headgroups. Derivatives of vorapaxar with alkyl chains predicted to extend between TM6 and TM7 into the lipid bilayer inhibited PAR1 with apparent on rates similar to that of the parent compound in cell signaling assays. These data are consistent with vorapaxar binding to PAR1 via a pathway that passes between TM6 and TM7 from the lipid bilayer, in agreement with the most consistent pathway observed by molecular dynamics. While there is some evidence of entry of the ligand into rhodopsin and lipid-activated GPCRs from the cell membrane, our study provides the first such evidence for a peptide-activated GPCR and suggests that metastable intermediates along drug binding and dissociation pathways can be stabilized by specific interactions between lipids and the ligand.


Subject(s)
Lactones/metabolism , Lipid Bilayers/metabolism , Pyridines/metabolism , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Animals , Binding Sites , Fibroblasts , Humans , Lactones/chemistry , Ligands , Molecular Dynamics Simulation , Molecular Structure , Phosphatidylcholines/metabolism , Protein Binding , Protein Conformation , Pyridines/chemistry , Rats , Receptor, PAR-1/chemistry
8.
Blood ; 132(2): 159-169, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29866816

ABSTRACT

Protein C is a plasma serine protease zymogen whose active form, activated protein C (APC), exerts potent anticoagulant activity. In addition to its antithrombotic role as a plasma protease, pharmacologic APC is a pleiotropic protease that activates diverse homeostatic cell signaling pathways via multiple receptors on many cells. Engineering of APC by site-directed mutagenesis provided a signaling selective APC mutant with 3 Lys residues replaced by 3 Ala residues, 3K3A-APC, that lacks >90% anticoagulant activity but retains normal cell signaling activities. This 3K3A-APC mutant exerts multiple potent neuroprotective activities, which require the G-protein-coupled receptor, protease activated receptor 1. Potent neuroprotection in murine ischemic stroke models is linked to 3K3A-APC-induced signaling that arises due to APC's cleavage in protease activated receptor 1 at a noncanonical Arg46 site. This cleavage causes biased signaling that provides a major explanation for APC's in vivo mechanism of action for neuroprotective activities. 3K3A-APC appeared to be safe in ischemic stroke patients and reduced bleeding in the brain after tissue plasminogen activator therapy in a recent phase 2 clinical trial. Hence, it merits further clinical testing for its efficacy in ischemic stroke patients. Recent studies using human fetal neural stem and progenitor cells show that 3K3A-APC promotes neurogenesis in vitro as well as in vivo in the murine middle cerebral artery occlusion stroke model. These recent advances should encourage translational research centered on signaling selective APC's for both single-agent therapies and multiagent combination therapies for ischemic stroke and other neuropathologies.


Subject(s)
Neurons/metabolism , Protein C/metabolism , Receptor, PAR-1/metabolism , Animals , Biomarkers , Blood-Brain Barrier/metabolism , Combined Modality Therapy , Disease Susceptibility , Enzyme Activation , Gene Expression Regulation , Humans , Protein C/chemistry , Protein C/genetics , Receptor, PAR-1/chemistry , Receptor, PAR-1/genetics , Regeneration , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Stroke/etiology , Stroke/metabolism , Stroke/therapy
9.
Structure ; 26(6): 829-838.e4, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29731231

ABSTRACT

The protease-activated receptor 1 (PAR1), a G protein-coupled receptor (GPCR) involved in hemostasis, thrombosis, and inflammation, is activated by thrombin or other coagulation proteases. This activation is inhibited by the irreversible antagonist vorapaxar used for anti-platelet therapy. Despite detailed structural and functional information, how vorapaxar binding alters the structural properties of PAR1 to prevent activation is hardly known. Here we apply dynamic single-molecule force spectroscopy to characterize how vorapaxar binding changes the mechanical, kinetic, and energetic properties of human PAR1 under physiologically relevant conditions. We detect structural segments stabilizing PAR1 and quantify their properties in the unliganded and the vorapaxar-bound state. In the presence of vorapaxar, most structural segments increase conformational variability, lifetime, and free energy, and reduce mechanical rigidity. These changes highlight a general trend in how GPCRs are affected by strong antagonists.


Subject(s)
Lactones/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation , Single Molecule Imaging
10.
Biochem Biophys Res Commun ; 495(1): 1300-1304, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29196264

ABSTRACT

Activated Protein C (APC) is a serine-protease that displays antithrombotic and anti-inflammatory properties. In addition, cleavage of protease-activated receptor 1 (PAR1) by APC exerts endothelial cytoprotective actions. The effects of APC on endothelial cells may be reproduced by TR47, a PAR1-based peptide that mimics the novel N-terminus of PAR1 generated upon cleavage at Arg-46 by APC. In this study we demonstrate that wild-type APC and its signaling-proficient mutant, APC-2Cys (which has dramatically reduced anticoagulant activity), display similar inhibitory effects towards the transendothelial migration of A375 human melanoma cells. Consistent with this observation, APC and APC-2Cys significantly reduced the in vivo metastatic potential of the B16F10 murine melanoma cells. TR47 recapitulated the in vitro and in vivo protective profiles of APC and APC-2Cys. Treatment of EA.hy926 endothelial cells with TR47 (20 µM) significantly decreased the A375 cell migration. In addition, treatment of C57/BL6 mice with a single TR47 dose (125 µg/animal) strongly reduced the metastatic burden of B16F10 cells. Together, our results suggest that protection of the endothelial barrier by APC/TR47-mediated signaling pathways might be a valuable therapeutic approach to prevent metastasis.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic/drug effects , Melanoma/metabolism , Melanoma/secondary , Peptides/administration & dosage , Receptor, PAR-1/chemistry , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology , Peptides/chemistry
11.
J Mol Graph Model ; 77: 205-217, 2017 10.
Article in English | MEDLINE | ID: mdl-28881236

ABSTRACT

Currently, Vorapaxar is the only recently FDA-approved antiplatelet drug targeting Protease-activated receptor 1 (PAR1). However, a novel antagonist, F16357, has been shown to prevent painful bladder syndrome, also known as interstitial cystitis (IC). Unfortunately, there is no high resolution structure of the F16357-receptor complex, hindering its optimization as a therapeutic agent. In this study, we used docking and molecular dynamic (MD) simulations to investigate the detailed interactions between F16357 and PAR1 at a molecular level. The recently solved crystal structure of human PAR1 complexed with Vorapaxar was used in our docking of F16357 into the binding pocket of the receptor. To enhance binding pose selection, F16357 was docked first without constraints and then with a positional constraint to invert its orientation to become similar to that of Vorapaxar. The three systems, with crystal Vorapaxar, F16357 and an inverted F16357, were subjected to 3.0µs MD simulations. The MM-GBSA binding energy analysis showed that F16357 binds more strongly in a pose obtained from an unrestrained docking than in the inverted pose from a restrained docking; and Vorapaxar binds more strongly than F17357. This ordering is consistent with the experimental pIC50 values. Our structural data showed subtle changes in the binding pose between Vorapaxar and F16357. Transmembrane helices 1, 2, 5, and 7 were most significantly affected; most notably a large kink at F2795.47 in TM helix 5 of the Vorapaxar complex was completely absent in the F16357 complex. The results of this study facilitate the future development of other therapeutic PAR1 antagonists.


Subject(s)
Lactones/chemistry , Piperazines/chemistry , Protein Conformation/drug effects , Pyridines/chemistry , Receptor, PAR-1/chemistry , Binding Sites , Humans , Lactones/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Piperazines/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors
12.
J Thromb Haemost ; 15(11): 2198-2207, 2017 11.
Article in English | MEDLINE | ID: mdl-28834159

ABSTRACT

Essentials The basis of cytoprotective protease-activated receptor 1 (PAR1) signaling is not fully understood. Activated protein C chimera (APCFVII-82 ) was used to identify requirements for PAR1 signaling. APCFVII-82 did not initiate PAR1 signaling, but conferred monocyte anti-inflammatory activity. APC-specific light chain residues are required for cytoprotective PAR1 signaling. SUMMARY: Background Activated protein C (APC) cell signaling is largely reliant upon its ability to mediate protease-activated receptor (PAR) 1 proteolysis when bound to the endothelial cell (EC) protein C (PC) receptor (EPCR). Furthermore, EPCR-bound PC modulates PAR1 signaling by thrombin to induce APC-like EC cytoprotection. Objective The molecular determinants of EPCR-dependent cytoprotective PAR1 signaling remain poorly defined. To address this, a PC-factor VII chimera (PCFVII-82 ) possessing FVII N-terminal domains and conserved EPCR binding was characterized. Methods Activated PC-FVII chimera (APCFVII-82 ) anticoagulant activity was measured with calibrated automated thrombography and activated FV degradation assays. APCFVII-82 signaling activity was characterized by the use of reporter assays of PAR1 proteolysis and EC barrier integrity. APCFVII-82 anti-inflammatory activity was assessed according to its inhibition of nuclear factor-κB (NF-κB) activation and cytokine secretion from monocytes. Results PCFVII-82 was activated normally by thrombin on ECs, but was unable to inhibit plasma thrombin generation. Surprisingly, APCFVII-82 did not mediate EPCR-dependent PAR1 proteolysis, confer PAR1-dependent protection of thrombin-induced EC barrier disruption, or limit PAR1-dependent attenuation of interleukin-6 release from lipopolysaccharide (LPS)-stimulated macrophages. Interestingly, EPCR occupation by active site-blocked APCFVII-82 was, like FVII, unable to mimic EC barrier stabilization induced by PC upon PAR1 proteolysis by thrombin. APCFVII-82 did, however, diminish LPS-induced NF-κB activation and tumor necrosis factor-α release from monocytes in an apolipoprotein E receptor 2-dependent manner, with similar efficacy as wild-type APC. Conclusions These findings identify a novel role for APC light chain amino acid residues outside the EPCR-binding site in enabling cytoprotective PAR1 signaling.


Subject(s)
Endothelial Cells/metabolism , Factor VII/metabolism , Inflammation/prevention & control , Macrophages/metabolism , Monocytes/metabolism , Protein C/metabolism , Receptor, PAR-1/metabolism , Animals , Binding Sites , Blood Coagulation , Capillary Permeability , Endothelial Protein C Receptor/metabolism , Factor VII/chemistry , Factor VII/genetics , HEK293 Cells , Humans , Inflammation/metabolism , Interleukin-6/metabolism , LDL-Receptor Related Proteins/metabolism , Mice , NF-kappa B/metabolism , Protein Binding , Protein C/chemistry , Protein C/genetics , Protein Interaction Domains and Motifs , RAW 264.7 Cells , Receptor, PAR-1/chemistry , Recombinant Fusion Proteins/chemistry , Signal Transduction , Structure-Activity Relationship , Thrombin/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
J Biol Chem ; 292(33): 13867-13878, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28652403

ABSTRACT

Vascular inflammation and thrombosis require the concerted actions of several different agonists, many of which act on G protein-coupled receptors (GPCRs). GPCR dimerization is a well-established phenomenon that can alter protomer function. In platelets and other cell types, protease-activated receptor-4 (PAR4) has been shown to dimerize with the purinergic receptor P2Y12 to coordinate ß-arrestin-mediated Akt signaling, an important mediator of integrin activation. However, the mechanism by which the PAR4-P2Y12 dimer controls ß-arrestin-dependent Akt signaling is not known. We now report that PAR4 and P2Y12 heterodimer internalization is required for ß-arrestin recruitment to endosomes and Akt signaling. Using bioluminescence resonance energy transfer, immunofluorescence microscopy, and co-immunoprecipitation in cells expressing receptors exogenously and endogenously, we demonstrate that PAR4 and P2Y12 specifically interact and form dimers expressed at the cell surface. We also found that activation of PAR4 but not of P2Y12 drives internalization of the PAR4-P2Y12 heterodimer. Remarkably, activated PAR4 internalization was required for recruitment of ß-arrestin to endocytic vesicles, which was dependent on co-expression of P2Y12. Interestingly, stimulation of the PAR4-P2Y12 heterodimer promotes ß-arrestin and Akt co-localization to intracellular vesicles. Moreover, activated PAR4-P2Y12 internalization is required for sustained Akt activation. Thus, internalization of the PAR4-P2Y12 heterodimer is necessary for ß-arrestin recruitment to endosomes and Akt signaling and lays the foundation for examining whether blockade of PAR4 internalization reduces integrin and platelet activation.


Subject(s)
Endocytosis , Proto-Oncogene Proteins c-akt/agonists , Receptors, Purinergic P2Y12/metabolism , Receptors, Thrombin/agonists , Signal Transduction , beta-Arrestin 2/metabolism , Amino Acid Substitution , Animals , Bioluminescence Resonance Energy Transfer Techniques , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endosomes/metabolism , Humans , Immunoprecipitation , Microscopy, Fluorescence , Protein Multimerization , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptor, PAR-1/agonists , Receptor, PAR-1/chemistry , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/genetics , Receptors, Thrombin/chemistry , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , beta-Arrestin 2/chemistry
14.
Oncotarget ; 8(10): 16829-16842, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28187433

ABSTRACT

Doxycycline have been reported to exert anti-cancer activity and have been assessed as anti-cancer agents in clinical trials. However, the direct targets of doxycycline in cancer cells remain unclear. In this study, we used a chemical proteomics approach to identify the Protease-activated receptor 1 (PAR1) as a specific target of inhibition of doxycycline. Binding assays and single-molecule imaging assays were performed to confirm the inhibition of doxycycline to PAR1. The effect of doxycycline on multi-omics and cell functions were assessed based on a PAR1/thrombin model. Molecular docking and molecular dynamic simulations revealed that doxycycline interacts with key amino acids in PAR1. Mutation of PAR1 further confirmed the computation-based results. Moreover, doxycycline provides highly selective inhibition of PAR1 signaling in tumors in vitro and in vivo. Using pathological clinical samples co-stained for doxycycline and PAR1, it was found that doxycycline fluorescence intensity and PAR1 expression shown a clear positive correlation. Thus, doxycycline may be a useful targeted anti-cancer drug that should be further investigated in clinical trials.


Subject(s)
Doxycycline/pharmacology , Receptor, PAR-1/antagonists & inhibitors , A549 Cells , Animals , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Progression , Doxycycline/chemistry , Drug Screening Assays, Antitumor , Female , HCT116 Cells , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Dynamics Simulation , Molecular Targeted Therapy , Random Allocation , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Nat Commun ; 6: 8857, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26561004

ABSTRACT

A current challenge in life sciences is to image cell membrane receptors while characterizing their specific interactions with various ligands. Addressing this issue has been hampered by the lack of suitable nanoscopic methods. Here we address this challenge and introduce multifunctional high-resolution atomic force microscopy (AFM) to image human protease-activated receptors (PAR1) in the functionally important lipid membrane and to simultaneously localize and quantify their binding to two different ligands. Therefore, we introduce the surface chemistry to bifunctionalize AFM tips with the native receptor-activating peptide and a tris-N-nitrilotriacetic acid (tris-NTA) group binding to a His10-tag engineered to PAR1. We further introduce ways to discern between the binding of both ligands to different receptor sites while imaging native PAR1s. Surface chemistry and nanoscopic method are applicable to a range of biological systems in vitro and in vivo and to concurrently detect and localize multiple ligand-binding sites at single receptor resolution.


Subject(s)
Microscopy, Atomic Force/methods , Receptor, PAR-1/chemistry , Binding Sites , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation
16.
SAR QSAR Environ Res ; 26(10): 795-807, 2015.
Article in English | MEDLINE | ID: mdl-26501801

ABSTRACT

Protease-activated receptor 1 (PAR1) has been established as a promising target in many diseases, including various cancers. Strong evidence also suggests its role in metastasis. It is proved experimentally that PAR1 can induce numerous cell phenotypes, i.e. proliferation and differentiation. A strong link between PAR1 gene overexpression and high levels of ß-catenin was suggested by a study of the PAR1-Gα(13)-DVL axis in ß-catenin stabilization in cancers. An in vitro study was carried out to analyze PAR1 expression by flow cytometry on CD38+138+ plasma cells obtained from patients either at diagnosis (n: 46) (newly diagnosed multiple myeloma (NDMM)) or at relapse (n: 45) (relapsed/refractory multiple myeloma (RRMM)) and compared with the controls. Our previously synthesized benzoxazole (XT2B) and benzamide (XT5) derivatives were tested with in vitro 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, which revealed significant inhibitory activity on PAR1. We provide docking studies using Autodock Vina of these newly tested compounds to compare with the known PAR1 inhibitors in order to examine the binding mechanisms. In addition, the docking results are validated using HYDE binding assessment and a neural network (NN) scoring function.


Subject(s)
Antineoplastic Agents/chemistry , Benzamides/chemistry , Benzoxazoles/chemistry , Molecular Docking Simulation , Multiple Myeloma/drug therapy , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/chemistry , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzoxazoles/pharmacology , Humans , In Vitro Techniques , Multiple Myeloma/enzymology , Neural Networks, Computer , Plasma Cells/drug effects , Plasma Cells/enzymology , Protein Binding , Quantitative Structure-Activity Relationship , Receptor, PAR-1/metabolism , Recurrence
17.
Nat Methods ; 12(9): 845-851, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26167642

ABSTRACT

Imaging native membrane receptors and testing how they interact with ligands is of fundamental interest in the life sciences but has proven remarkably difficult to accomplish. Here, we introduce an approach that uses force-distance curve-based atomic force microscopy to simultaneously image single native G protein-coupled receptors in membranes and quantify their dynamic binding strength to native and synthetic ligands. We measured kinetic and thermodynamic parameters for individual protease-activated receptor-1 (PAR1) molecules in the absence and presence of antagonists, and these measurements enabled us to describe PAR1's ligand-binding free-energy landscape with high accuracy. Our nanoscopic method opens an avenue to directly image and characterize ligand binding of native membrane receptors.


Subject(s)
Lipid Bilayers/chemistry , Microscopy, Atomic Force/methods , Models, Chemical , Molecular Imaging/methods , Receptor, PAR-1/chemistry , Binding Sites , Computer Simulation , Energy Transfer , Humans , Kinetics , Ligands , Protein Binding , Stress, Mechanical
18.
J Biol Chem ; 290(25): 15785-15798, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25934391

ABSTRACT

G protein-coupled receptors (GPCRs) are remarkably versatile signaling systems that are activated by a large number of different agonists on the outside of the cell. However, the inside surface of the receptors that couple to G proteins has not yet been effectively modulated for activity or treatment of diseases. Pepducins are cell-penetrating lipopeptides that have enabled chemical and physical access to the intracellular face of GPCRs. The structure of a third intracellular (i3) loop agonist, pepducin, based on protease-activated receptor-1 (PAR1) was solved by NMR and found to closely resemble the i3 loop structure predicted for the intact receptor in the on-state. Mechanistic studies revealed that the pepducin directly interacts with the intracellular H8 helix region of PAR1 and allosterically activates the receptor through the adjacent (D/N)PXXYYY motif through a dimer-like mechanism. The i3 pepducin enhances PAR1/Gα subunit interactions and induces a conformational change in fluorescently labeled PAR1 in a very similar manner to that induced by thrombin. As pepducins can potentially be made to target any GPCR, these data provide insight into the identification of allosteric modulators to this major drug target class.


Subject(s)
Biomimetic Materials/chemistry , Cell-Penetrating Peptides/chemistry , GTP-Binding Protein alpha Subunits/chemistry , Receptor, PAR-1/chemistry , Allosteric Regulation/genetics , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Nuclear Magnetic Resonance, Biomolecular , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism
19.
Curr Top Med Chem ; 15(20): 2080-114, 2015.
Article in English | MEDLINE | ID: mdl-25986685

ABSTRACT

PAR1, member of the family of protease-activated receptors, is a GPCR whose activation requires a proteolytic cleavage at its extracellular N-terminus to unveil a tethered activating ligand. Although thrombin is the main activator of this receptor, diverse other proteases can also activate and disarm PAR1. Besides, tethered activating ligand-based peptides (PAR-APs) can also activate the receptor. PAR1 mainly signals via G proteins but, it can also signal using ß-arrestin pathways and by transactivation of other receptors. This complex PAR1 interactome is completed with the receptor desensitization, trafficking, and degradation. PAR1 has shown species-, cellular-, and physiological or pathological state-dependent specificity. This review try to give an overview on the complex PAR1 interactome, its therapeutic impact upon the cardiovascular, immune and nervous systems, inflammation and cancer, as well as, on its modulation with agonists and antagonists.


Subject(s)
Arrestins/chemistry , GTP-Binding Proteins/chemistry , Receptor, PAR-1/chemistry , Thrombin/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Ligands , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Organ Specificity , Protein Interaction Mapping , Protein Transport , Proteolysis , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/genetics , Signal Transduction , Species Specificity , beta-Arrestins
20.
Mediators Inflamm ; 2015: 437695, 2015.
Article in English | MEDLINE | ID: mdl-25878399

ABSTRACT

Aside from their role in hemostasis, coagulant and fibrinolytic proteases are important mediators of inflammation in diseases such as asthma, atherosclerosis, rheumatoid arthritis, and cancer. The blood circulating zymogens of these proteases enter damaged tissue as a consequence of vascular leak or rupture to become activated and contribute to extravascular coagulation or fibrinolysis. The coagulants, factor Xa (FXa), factor VIIa (FVIIa), tissue factor, and thrombin, also evoke cell-mediated actions on structural cells (e.g., fibroblasts and smooth muscle cells) or inflammatory cells (e.g., macrophages) via the proteolytic activation of protease-activated receptors (PARs). Plasmin, the principle enzymatic mediator of fibrinolysis, also forms toll-like receptor-4 (TLR-4) activating fibrin degradation products (FDPs) and can release latent-matrix bound growth factors such as transforming growth factor-ß (TGF-ß). Furthermore, the proteases that convert plasminogen into plasmin (e.g., urokinase plasminogen activator) evoke plasmin-independent proinflammatory actions involving coreceptor activation. Selectively targeting the receptor-mediated actions of hemostatic proteases is a strategy that may be used to treat inflammatory disease without the bleeding complications of conventional anticoagulant therapies. The mechanisms by which proteases of the coagulant and fibrinolytic systems contribute to extravascular inflammation in disease will be considered in this review.


Subject(s)
Blood Coagulation , Fibrinolysis , Inflammation/etiology , Peptide Hydrolases/physiology , Factor X/physiology , Fibrin Fibrinogen Degradation Products/physiology , Fibrinolysin/physiology , Humans , Protein Multimerization , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/chemistry , Receptor, PAR-1/physiology , Receptors, Growth Factor/genetics , Tissue Plasminogen Activator/physiology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...