Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 386
Filter
1.
Protein J ; 43(3): 603-612, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734856

ABSTRACT

Disintegrins, a family of snake venom protein, which are capable of modulating the activity of integrins that play a fundamental role in the regulation of many physiological and pathological processes. The main purpose of this study is to obtain the recombinant disintegrin (r-DI) and evaluate its biological activity. In this study, we explored a high-level expression prokaryotic system and purification strategy for r-DI. Then, r-DI was treated to assay effects on cell growth, migration, and invasion. The affinity for the interactions of r-DI with integrin was determined using Surface plasmon resonance (SPR) analyses. The r-DI can be expressed in Escherichia coli and purified by one-step chromatography. The r-DI can inhibit B16F10 cells proliferation, migration, and invasion. Also, we found that r-DI could interact with the integrin αIIbß3 (GPIIb/IIIa). The r-DI can be expressed, purified, characterized through functional assays, and can also maintain strong biological activities. Thus, this study showed potential therapeutic effects of r-DI for further functional and structural studies.


Subject(s)
Disintegrins , Escherichia coli , Recombinant Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Animals , Disintegrins/chemistry , Disintegrins/genetics , Disintegrins/isolation & purification , Disintegrins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Mice , Viperidae/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Cell Line, Tumor , Gene Expression , Cell Movement/drug effects , Cell Proliferation/drug effects , Crotalid Venoms/chemistry , Crotalid Venoms/genetics , Crotalinae , Venomous Snakes
2.
Biophys J ; 122(3): 533-543, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36566352

ABSTRACT

The platelet integrin αIIbß3 undergoes long-range conformational transitions associated with its functional conversion from inactive (low-affinity) to active (high-affinity) during hemostasis. Although new conformations that are intermediate between the well-characterized bent and extended states have been identified, their molecular dynamic properties and functions in the assembly of adhesions remain largely unexplored. In this study, we evaluated the properties of intermediate conformations of integrin αIIbß3 and characterized their effects on the assembly of adhesions by combining all-atom simulations, principal component analysis, and mesoscale modeling. Our results show that in the low-affinity, bent conformation, the integrin ectodomain tends to pivot around the legs; in intermediate conformations, the headpiece becomes partially extended, away from the lower legs. In the fully open, active state, αIIbß3 is flexible, and the motions between headpiece and lower legs are accompanied by fluctuations of the transmembrane helices. At the mesoscale, bent integrins form only unstable adhesions, but intermediate or open conformations stabilize the adhesions. These studies reveal a mechanism by which small variations in ligand binding affinity and enhancement of the ligand-bound lifetime in the presence of actin retrograde flow stabilize αIIbß3 integrin adhesions.


Subject(s)
Molecular Dynamics Simulation , Platelet Glycoprotein GPIIb-IIIa Complex , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Ligands , Blood Platelets/metabolism , Protein Structure, Secondary , Protein Conformation
3.
J Am Soc Nephrol ; 33(10): 1841-1856, 2022 10.
Article in English | MEDLINE | ID: mdl-36038265

ABSTRACT

BACKGROUND: Bleeding diatheses, common among patients with ESKD, can lead to serious complications, particularly during invasive procedures. Chronic urea overload significantly increases cyanate concentrations in patients with ESKD, leading to carbamylation, an irreversible modification of proteins and peptides. METHODS: To investigate carbamylation as a potential mechanistic link between uremia and platelet dysfunction in ESKD, we used liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) to quantify total homocitrulline, and biotin-conjugated phenylglyoxal labeling and Western blot to detect carbamylated integrin α IIb ß 3 (a receptor required for platelet aggregation). Flow cytometry was used to study activation of isolated platelets and platelet-rich plasma. In a transient transfection system, we tested activity and fibrinogen binding of different mutated forms of the receptor. We assessed platelet adhesion and aggregation in microplate assays. RESULTS: Carbamylation inhibited platelet activation, adhesion, and aggregation. Patients on hemodialysis exhibited significantly reduced activation of α IIb ß 3 compared with healthy controls. We found significant carbamylation of both subunits of α IIb ß 3 on platelets from patients receiving hemodialysis versus only minor modification in controls. In the transient transfection system, modification of lysine 185 in the ß 3 subunit was associated with loss of receptor activity and fibrinogen binding. Supplementation of free amino acids, which was shown to protect plasma proteins from carbamylation-induced damage in patients on hemodialysis, prevented loss of α IIb ß 3 activity in vitro. CONCLUSIONS: Carbamylation of α IIb ß 3-specifically modification of the K185 residue-might represent a mechanistic link between uremia and dysfunctional primary hemostasis in patients on hemodialysis. The observation that free amino acids prevented the carbamylation-induced loss of α IIb ß 3 activity suggests amino acid administration during dialysis may help to normalize platelet function.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex , Uremia , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Carbamylation , Tandem Mass Spectrometry , Blood Platelets , Uremia/complications , Uremia/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Amino Acids
4.
Adv Sci (Weinh) ; 9(7): e2103228, 2022 03.
Article in English | MEDLINE | ID: mdl-35023301

ABSTRACT

Conventional antiplatelet agents indiscriminately inhibit both thrombosis and hemostasis, and the increased bleeding risk thus hampers their use at more aggressive dosages to achieve adequate effect. Blocking integrin αIIbß3 outside-in signaling by separating the ß3/Src interaction, yet to be proven in vivo, may nonetheless resolve this dilemma. Identification of a specific druggable target for this strategy remains a fundamental challenge as Src SH3 is known to be responsible for binding to not only integrin ß3 but also the proteins containing the PXXP motif. In vitro and in vivo mutational analyses show that the residues, especially E97, in the RT loop of Src SH3 are critical for interacting with ß3. DCDBS84, a small molecule resulting from structure-based virtual screening, is structurally validated to be directed toward the projected target. It specifically disrupts ß3/Src interaction without affecting canonical PXXP binding and thus inhibits the outside-in signaling-regulated platelet functions. Treatment of mice with DCDBS84 causes a profound inhibition of thrombosis, equivalent to that induced by extremely high doses of αIIbß3 antagonist, but does not compromise primary hemostasis. Specific targets are revealed for a preferential inhibition of thrombosis that may lead to new classes of potent antithrombotics without hemorrhagic side effects.


Subject(s)
Blood Platelets , Thrombosis , Animals , Blood Platelets/metabolism , Hemostasis/physiology , Integrin beta3/chemistry , Integrin beta3/metabolism , Mice , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/metabolism , Thrombosis/prevention & control
5.
J Biomol Struct Dyn ; 40(20): 9701-9712, 2022.
Article in English | MEDLINE | ID: mdl-34060983

ABSTRACT

The tripeptide Arg-Gly-Asp acid (RGD) is a protein sequence in the binding of proteins to cell surfaces, and is involved in various biological processes such as cell adhesion to the extracellular matrix, platelet activation, hemostasis, etc. The C2 domain of the Von Willebrand Factor (VWF), containing the RGD motif, plays an important role in the initial homeostasis process. It binds to the αIIbß3 integrin and stimulates platelet aggregation. We have investigated, using the molecular Dynamic (MD) simulation method, the effect of the RGD-peptide length, and temperature variation, on the binding to the αIIbß3 integrin receptor. We examined 10 different structural modes of the αIIbß3 at three different temperatures; 237 K, 310 K and 318 K. Our findings show that the amino acids that form a binding pocket include Asp224, Tyr234, Ser226, Tyr190, Tyr189, Trp260, Trp262, Asp259, Lys253, Arg214, Asp217, Ser161 and Ala218 and that the ligand-receptor interaction was increased at higher temperatures. It was also found that the increase in the number of ligands' amino acids and their types (% glycine) plays an important role in the stability, conformation, and ligand-receptor interaction.Communicated by Ramaswamy H. Sarma.


Subject(s)
Molecular Dynamics Simulation , Platelet Glycoprotein GPIIb-IIIa Complex , Temperature , Ligands , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Oligopeptides/chemistry , Amino Acids
6.
Hematol Oncol Stem Cell Ther ; 15(1): 21-26, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-33600779

ABSTRACT

Glanzmann's thrombasthenia (GT) is an autosomal recessive congenital bleeding disorder of platelet aggregation. Mutations in ITGA2B and ITGB3 genes result in quantitative and/or qualitative abnormalities of the glycoprotein receptor complex IIb/IIIa (integrin αIIbß3), which in turn impairs platelet aggregation and lead to GT. In this study, whole genome single nucleotide polymorphism (SNP) genotyping as well as whole exome sequencing was performed in a large family segregating GT. Analysis of the genotypes localized the disease region to chromosome 17q21.2-q21.3. Filtration of whole exome data and candidate variants prioritization identified a pathogenic variant in the ITGB3 gene. The single nucleotide deletion variant (c.2113delC) in exon 13 of the ITGB3 gene is predicted to cause a frameshift and absence of vital C-terminal domains including the transmembrane helix and the cytoplasmic domain. Clinical variability of the bleeding phenotype in affected individuals with the same mutation suggests that other genetic and nongenetic factors are responsible for determining GT features.


Subject(s)
Integrin beta3 , Thrombasthenia , Humans , Exons , Frameshift Mutation , Integrin beta3/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Saudi Arabia , Thrombasthenia/genetics
7.
J Thromb Haemost ; 20(2): 285-292, 2022 02.
Article in English | MEDLINE | ID: mdl-34674369

ABSTRACT

The disulfide bond is a covalent bond formed between the sulfur atoms of two cysteine residues in proteins. Our understanding of the role of these ubiquitous bonds in protein function has changed dramatically over the past decade. Initially thought to be fully formed and inert in the native protein, we know now that both these assumptions are incorrect for many proteins. Here, we review recent evidence for production and function of multiple partially disulfide-bonded forms of plasma fibrinogen and platelet αIIbß3 integrin. The disulfide bonds are not cleaved in these mature proteins but rather a significant fraction of the bonds never form during maturation of the protein. The resulting different covalent states influence the functioning of the protein. These findings change our concept of the native, functional protein.


Subject(s)
Fibrinogen , Platelet Glycoprotein GPIIb-IIIa Complex , Thrombosis , Blood Platelets/metabolism , Cysteine/chemistry , Disulfides/chemistry , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
8.
J Biol Chem ; 297(5): 101318, 2021 11.
Article in English | MEDLINE | ID: mdl-34678312

ABSTRACT

Studying the tight activity regulation of platelet-specific integrin αIIbß3 is foundational and paramount to our understanding of integrin structure and activation. αIIbß3 is essential for the aggregation and adhesion function of platelets in hemostasis and thrombosis. Structural and mutagenesis studies have previously revealed the critical role of αIIbß3 transmembrane (TM) association in maintaining the inactive state. Gain-of-function TM mutations were identified and shown to destabilize the TM association leading to integrin activation. Studies using isolated TM peptides have suggested an altered membrane embedding of the ß3 TM α-helix coupled with αIIbß3 activation. However, controversies remain as to whether and how the TM α-helices change their topologies in the context of full-length integrin in native cell membrane. In this study, we utilized proline scanning mutagenesis and cysteine scanning accessibility assays to analyze the structure and function correlation of the αIIbß3 TM domain. Our identification of loss-of-function proline mutations in the TM domain suggests the requirement of a continuous TM α-helical structure in transmitting activation signals bidirectionally across the cell membrane, characterized by the inside-out activation for ligand binding and the outside-in signaling for cell spreading. Similar results were found for αLß2 and α5ß1 TM domains, suggesting a generalizable mechanism. We also detected a topology change of ß3 TM α-helix within the cell membrane, but only under conditions of cell adhesion and the absence of αIIb association. Our data demonstrate the importance of studying the structure and function of the integrin TM domain in the native cell membrane.


Subject(s)
Cell Membrane , Platelet Glycoprotein GPIIb-IIIa Complex , Signal Transduction , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , HEK293 Cells , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Conformation, alpha-Helical , Protein Domains
9.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504018

ABSTRACT

During activation the platelet cytoskeleton is reorganized, inducing adhesion to the extracellular matrix and cell spreading. These processes are critical for wound healing and clot formation. Initially, this task relies on the formation of strong cellular-extracellular matrix interactions, exposed in subendothelial lesions. Despite the medical relevance of these processes, there is a lack of high-resolution structural information on the platelet cytoskeleton controlling cell spreading and adhesion. Here, we present in situ structural analysis of membrane receptors and the underlying cytoskeleton in platelet protrusions by applying cryoelectron tomography to intact platelets. We utilized three-dimensional averaging procedures to study receptors at the plasma membrane. Analysis of substrate interaction-free receptors yielded one main structural class resolved to 26 Å, resembling the αIIbß3 integrin folded conformation. Furthermore, structural analysis of the actin network in pseudopodia indicates a nonuniform polarity of filaments. This organization would allow generation of the contractile forces required for integrin-mediated cell adhesion.


Subject(s)
Actin Cytoskeleton , Actins/chemistry , Blood Platelets/physiology , Cell Membrane/metabolism , Cell Surface Extensions/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Actins/metabolism , Cell Adhesion , Humans , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
10.
Blood ; 138(15): 1359-1372, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34375384

ABSTRACT

The αIIbß3 integrin receptor coordinates platelet adhesion, activation, and mechanosensing in thrombosis and hemostasis. Using differential cysteine alkylation and mass spectrometry, we have identified a disulfide bond in the αIIb subunit linking cysteines 490 and 545 that is missing in ∼1 in 3 integrin molecules on the resting and activated human platelet surface. This alternate covalent form of αIIbß3 is predetermined as it is also produced by human megakaryoblasts and baby hamster kidney fibroblasts transfected with recombinant integrin. From coimmunoprecipitation experiments, the alternate form selectively partitions into focal adhesions on the activated platelet surface. Its function was evaluated in baby hamster kidney fibroblast cells expressing a mutant integrin with an ablated C490-C545 disulfide bond. The disulfide mutant integrin has functional outside-in signaling but extended residency time in focal adhesions due to a reduced rate of clathrin-mediated integrin internalization and recycling, which is associated with enhanced affinity of the αIIb subunit for clathrin adaptor protein 2. Molecular dynamics simulations indicate that the alternate covalent form of αIIb requires higher forces to transition from bent to open conformational states that is in accordance with reduced affinity for fibrinogen and activation by manganese ions. These findings indicate that the αIIbß3 integrin receptor is produced in various covalent forms that have different cell surface distribution and function. The C490, C545 cysteine pair is conserved across all 18 integrin α subunits, and the disulfide bond in the αV and α2 subunits in cultured cells is similarly missing, suggesting that the alternate integrin form and function are also conserved.


Subject(s)
Focal Adhesions/metabolism , Integrin beta3/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Animals , Cell Line , Cricetinae , Disulfides/analysis , Focal Adhesions/genetics , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta3/chemistry , Integrin beta3/genetics , Molecular Dynamics Simulation , Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Membrane Glycoprotein IIb/chemistry , Platelet Membrane Glycoprotein IIb/genetics
12.
Thromb Haemost ; 120(10): 1432-1441, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32717755

ABSTRACT

Bleeding and thrombocytopenia to readministration are the most serious side effects of clinical integrin αIIbß3 antagonists such as RGD-containing peptides. Here we show that a non-RGD peptide ZDPI, identified from skin secretions of Amolops loloensis, inhibited platelet aggregation induced by agonists, such as adenosine diphosphate, collagen, arachidonic acid, PAR1AP, and integrin αIIbß3 allosteric activator, and reduces soluble fibrinogen binding to activated platelets without perturbing adhesion numbers on immobilized fibrinogen. Further study showed that ZDPI preferred to bind to the active conformation of integrin αIIbß3, and thus inhibited c-Src-mediated integrin signaling transduction. In contrast to currently used clinical blockers of integrin αIIbß3, which are all conformation-unspecific blockers, ZDPI conformation specifically binds to activated integrin αIIbß3, subsequently suppressing platelet spreading. In vivo study revealed that ZDPI inhibited carotid arterial thrombosis with limited bleeding and thrombocytopenia. A non-RGD peptide which targets the active conformation of integrin αIIbß3, such as ZDPI, might be an excellent candidate or template to develop antithrombotics without significant bleeding and thrombocytopenia side effects.


Subject(s)
Blood Platelets/drug effects , Peptides/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Fibrinogen/metabolism , Hemorrhage/chemically induced , Humans , Male , Mice, Inbred C57BL , Peptides/adverse effects , Peptides/chemistry , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Conformation/drug effects , Thrombocytopenia/chemically induced
14.
Curr Biol ; 30(9): 1614-1625.e5, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32169208

ABSTRACT

Cells can sense and respond to various mechanical stimuli from their surrounding environment. One of the explanations for mechanosensitivity, a lipid-bilayer model, suggests that a stretch of the membrane induced by mechanical force alters the physical state of the lipid bilayer, driving mechanosensors to assume conformations better matched to the altered membrane. However, mechanosensors of this class are restricted to ion channels. Here, we reveal that integrin αIIbß3, a prototypic adhesion receptor, can be activated by various mechanical stimuli including stretch, shear stress, and osmotic pressure. The force-induced integrin activation was not dependent on its known intracellular activation signaling events and was even observed in reconstituted cell-free liposomes. Instead, these mechanical stimuli were found to alter the lipid embedding of the integrin ß3 transmembrane domain (TMD) and subsequently weaken the αIIb-ß3 TMD interaction, which results in activation of the receptor. Moreover, artificial modulation of the membrane curvature near integrin αIIbß3 can induce its activation in cells as well as in lipid nanodiscs, suggesting that physical deformation of the lipid bilayer, either by mechanical force or curvature, can induce integrin activation. Thus, our results establish the adhesion receptor as a bona fide mechanosensor that directly senses and responds to the force-modulated lipid environment. Furthermore, this study expands the lipid-bilayer model by suggesting that the force-induced topological change of TMDs and subsequent alteration in the TMD interactome is a molecular basis of sensing mechanical force transmitted via the lipid bilayer.


Subject(s)
Cell Membrane/physiology , Lipid Bilayers , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Stress, Mechanical , Animals , Biomechanical Phenomena , Blood Platelets , CHO Cells , Cells, Immobilized , Cricetinae , Cricetulus , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Mice , Protein Binding , Protein Conformation , Protein Domains , Signal Transduction
15.
Macromol Biosci ; 20(3): e1900338, 2020 03.
Article in English | MEDLINE | ID: mdl-32053289

ABSTRACT

In this study, activated platelet-derived vesicles (Act-VEs) are developed as a novel hemostatic biomaterial. Spherical Act-VEs (114.40 ± 11.69 nm in size) with surface charges of -24.73 ± 1.32 mV are successfully prepared from thrombin-activated murine platelets with high surface expression of active glycoprotein IIb/IIIa (GP IIb/IIIa, also known as αIIbß3) and P-selectin. Although nanosized vesicles from resting platelets (VEs) and Act-VEs showed similar sizes and surface charges, Act-VEs formed much larger aggregates in the presence of thrombin and CaCl2 , compared to VEs. After incubation with fibrinogen, Act-VEs formed much denser fibrin networks compared to platelets or VEs, probably due to active αIIbß3 on the surfaces of the Act-VEs. After intravenous injection of the Act-VEs, tail bleeding time and the blood loss are greatly reduced by Act-VEs in vivo. In addition, Act-VEs showed approximately sevenfold lower release of pro-inflammatory interleukin-1ß (IL-1ß) during incubation for 4 days, compared to platelets. Taken together, the formulated Act-VEs can serve as a promising hemostatic biomaterial for the efficient formation of fibrin clots without releasing pro-inflammatory cytokine.


Subject(s)
Blood Platelets/chemistry , Extracellular Vesicles/chemistry , Hemorrhage/drug therapy , Hemostatics , Nanoparticles , Platelet Activation , Animals , Bleeding Time , Cell Line , Hemorrhage/blood , Hemostatics/chemistry , Hemostatics/pharmacology , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/pharmacology
16.
Biochim Biophys Acta Biomembr ; 1862(5): 183198, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31958436

ABSTRACT

Binding of integrin alphaIIbbeta3 (αiibß3) to its ligands is a highly restricted and regulated mechanism. Any modification of the protein structure yields a dysfunctional role, especially in a redox environment. Here, we examine the effect of nitrosative stress on the αiibß3 reconstituted into nanodiscs. Using single molecule force spectroscopy, we measured the interaction between αiibß3 and its ligand RGD and found that in the presence of exogenous nitric oxide (NO) two force regimes are generated: a low force regime of ~100pN indicating the presence of integrin in a normal status, and a broad spectrum of high force regime (~210-450pN) suggesting the protein modification/aggregation. By high resolution atomic force microscopy imaging, we demonstrate that both NO and nitrite (a stable product formed from NO) are involved in destabilizing the transmembrane protein complex leading to release of αiibß3 from the lipid bilayer and protein aggregation. Our experimental setup opens new ways for testing in a membrane environment the effect of radical species on integrins under clinically relevant conditions.


Subject(s)
Nitrosative Stress/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Integrins/chemistry , Integrins/metabolism , Ligands , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microscopy, Atomic Force/methods , Nitrates/metabolism , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Oligopeptides , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein Binding , Reactive Nitrogen Species/chemistry , Reactive Nitrogen Species/metabolism
17.
Biochemistry ; 58(30): 3251-3259, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31264850

ABSTRACT

Integrin αIIbß3, a transmembrane heterodimer, mediates platelet aggregation when it switches from an inactive to an active ligand-binding conformation following platelet stimulation. Central to regulating αIIbß3 activity is the interaction between the αIIb and ß3 extracellular stalks, which form a tight heterodimer in the inactive state and dissociate in the active state. Here, we demonstrate that alanine replacements of sensitive positions in the heterodimer stalk interface destabilize the inactive conformation sufficiently to cause constitutive αIIbß3 activation. To determine the structural basis for this effect, we performed a structural bioinformatics analysis and found that perturbing intersubunit contacts with favorable interaction geometry through substitutions to alanine quantitatively accounted for the degree of constitutive αIIbß3 activation. This mutational study directly assesses the relationship between favorable interaction geometry at mutation-sensitive positions and the functional activity of those mutants, giving rise to a simple model that highlights the importance of interaction geometry in contributing to the stability between protein-protein interactions.


Subject(s)
Mutagenesis/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Allosteric Regulation/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
18.
PLoS One ; 14(4): e0214969, 2019.
Article in English | MEDLINE | ID: mdl-30978226

ABSTRACT

Integrins are transmembrane proteins involved in hemostasis, wound healing, immunity and cancer. In response to intracellular signals and ligand binding, integrins adopt different conformations: the bent (resting) form; the intermediate extended form; and the ligand-occupied active form. An integrin undergoing such conformational dynamics is the heterodimeric platelet receptor αIIbß3. Although the dramatic rearrangement of the overall structure of αIIbß3 during the activation process is potentially related to changes in the protein secondary structure, this has not been investigated so far in a membrane environment. Here we examine the Mn2+- and drug-induced activation of αIIbß3 and the impact on the structure of this protein reconstituted into liposomes. By quartz crystal microbalance with dissipation monitoring and activation assays we show that Mn2+ induces binding of the conformation-specific antibody PAC-1, which only recognizes the extended, active integrin. Circular dichroism spectroscopy reveals, however, that Mn2+-treatment does not induce major secondary structural changes of αIIbß3. Similarly, we found that treatment with clinically relevant drugs (e.g. quinine) led to the activation of αIIbß3 without significant changes in protein secondary structure. Molecular dynamics simulation studies revealed minor local changes in the beta-sheet probability of several extracellular domains of the integrin. Our experimental setup represents a new approach to study transmembrane proteins, especially integrins, in a membrane environment and opens a new way for testing drug binding to integrins under clinically relevant conditions.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Quinine/chemistry , Antibodies/chemistry , Circular Dichroism , Humans , Liposomes , Manganese/chemistry , Manganese/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Domains , Protein Structure, Quaternary , Protein Structure, Secondary , Quartz Crystal Microbalance Techniques
19.
Proc Natl Acad Sci U S A ; 115(39): E9105-E9114, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30209215

ABSTRACT

Integrin α/ß heterodimer adopts a compact bent conformation in the resting state, and upon activation undergoes a large-scale conformational rearrangement. During the inside-out activation, signals impinging on the cytoplasmic tail of ß subunit induce the α/ß separation at the transmembrane and cytoplasmic domains, leading to the extended conformation of the ectodomain with the separated leg and the opening headpiece that is required for the high-affinity ligand binding. It remains enigmatic which integrin subunit drives the bent-to-extended conformational rearrangement in the inside-out activation. The ß3 integrins, including αIIbß3 and αVß3, are the prototypes for understanding integrin structural regulation. The Leu33Pro polymorphism located at the ß3 PSI domain defines the human platelet-specific alloantigen (HPA) 1a/b, which provokes the alloimmune response leading to clinically important bleeding disorders. Some, but not all, anti-HPA-1a alloantibodies can distinguish the αIIbß3 from αVß3 and affect their functions with unknown mechanisms. Here we designed a single-chain ß3 subunit that mimics a separation of α/ß heterodimer on inside-out activation. Our crystallographic and functional studies show that the single-chain ß3 integrin folds into a bent conformation in solution but spontaneously extends on the cell surface. This demonstrates that the ß3 subunit autonomously drives the membrane-dependent conformational rearrangement during integrin activation. Using the single-chain ß3 integrin, we identified the conformation-dependent property of anti-HPA-1a alloantibodies, which enables them to differently recognize the ß3 in the bent state vs. the extended state and in the complex with αIIb vs. αV This study provides deeper understandings of integrin conformational activation on the cell surface.


Subject(s)
Glucuronidase/chemistry , Integrin beta3/chemistry , Isoantibodies/chemistry , Antibody Specificity , Crystallography, X-Ray , Glucuronidase/metabolism , HEK293 Cells , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Integrin beta3/metabolism , Isoantibodies/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Domains , Protein Folding
20.
Elife ; 72018 06 22.
Article in English | MEDLINE | ID: mdl-29932420

ABSTRACT

How proteins harness mechanical force to control function is a significant biological question. Here we describe a human cell surface receptor that couples ligand binding and force to trigger a chemical event which controls the adhesive properties of the receptor. Our studies of the secreted platelet oxidoreductase, ERp5, have revealed that it mediates release of fibrinogen from activated platelet αIIbß3 integrin. Protein chemical studies show that ligand binding to extended αIIbß3 integrin renders the ßI-domain Cys177-Cys184 disulfide bond cleavable by ERp5. Fluid shear and force spectroscopy assays indicate that disulfide cleavage is enhanced by mechanical force. Cell adhesion assays and molecular dynamics simulations demonstrate that cleavage of the disulfide induces long-range allosteric effects within the ßI-domain, mainly affecting the metal-binding sites, that results in release of fibrinogen. This coupling of ligand binding, force and redox events to control cell adhesion may be employed to regulate other protein-protein interactions.


Subject(s)
Blood Platelets/metabolism , Fibrinogen/chemistry , Mechanotransduction, Cellular , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Allosteric Regulation , Binding Sites , Blood Platelets/chemistry , Blood Platelets/cytology , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Molecular Dynamics Simulation , Oxidation-Reduction , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Interaction Domains and Motifs , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...