Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Publication year range
1.
Blood ; 136(19): 2200-2205, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32730588

ABSTRACT

Neutrophil adhesion and extravasation into tissue at sites of injury or infection depend on binding of the integrin lymphocyte function-associated antigen 1 (LFA-1) to ICAM-1 expressed on activated endothelial cells. The activation-dependent conformational change of LFA-1 to the high-affinity conformation (H+) requires kindlin-3 binding to the ß2-integrin cytoplasmic domain. Here we show that genetic deletion of the known kindlin interactor integrin-linked kinase (ILK) impaired neutrophil adhesion and extravasation in the cremaster muscle and in a clinically relevant model of renal ischemia reperfusion injury. Using in vitro microfluidic adhesion chambers and conformation-specific antibodies, we show that knockdown of ILK in HL-60 cells reduced the conformational change of ß2-integrins to the H+ conformation. Mechanistically, we found that ILK was required for protein kinase C (PKC) membrane targeting and chemokine-induced upregulation of its kinase activity. Moreover, PKC-α deficiency also resulted in impaired leukocyte adhesion in bone marrow chimeric mice. Mass spectrometric and western blot analyses revealed stimulation- and ILK-dependent phosphorylation of kindlin-3 upon activation. In summary, our data indicate an important role of ILK in kindlin-3-dependent conformational activation of LFA-1.


Subject(s)
Acute Kidney Injury/metabolism , CD18 Antigens/metabolism , Chemokines/pharmacology , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Animals , CD18 Antigens/chemistry , Cell Adhesion , Disease Models, Animal , HL-60 Cells , Humans , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Mice , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Phosphorylation , Reperfusion Injury/complications , Signal Transduction
2.
J Biol Chem ; 293(17): 6565-6577, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29507098

ABSTRACT

In αI integrins, including leukocyte function-associated antigen 1 (LFA-1), ligand-binding function is delegated to the αI domain, requiring extra steps in the relay of signals that activate ligand binding and coordinate it with cytoplasmic signals. Crystal structures reveal great variation in orientation between the αI domain and the remainder of the integrin head. Here, we investigated the mechanisms involved in signal relay to the αI domain, including whether binding of the ligand intercellular adhesion molecule-1 (ICAM-1) to the αI domain is linked to headpiece opening and engenders a preferred αI domain orientation. Using small-angle X-ray scattering and negative-stain EM, we define structures of ICAM-1, LFA-1, and their complex, and the effect of activation by Mn2+ Headpiece opening was substantially stabilized by substitution of Mg2+ with Mn2+ and became complete upon ICAM-1 addition. These agents stabilized αI-headpiece orientation, resulting in a well-defined orientation of ICAM-1 such that its tandem Ig-like domains pointed in the opposite direction from the ß-subunit leg of LFA-1. Mutations in the integrin ßI domain α1/α1' helix stabilizing either the open or the closed ßI-domain conformation indicated that α1/α1' helix movements are linked to ICAM-1 binding by the αI domain and to the extended-open conformation of the ectodomain. The LFA-1-ICAM-1 orientation described here with ICAM-1 pointing anti-parallel to the LFA-1 ß-subunit leg is the same orientation that would be stabilized by tensile force transmitted between the ligand and the actin cytoskeleton and is consistent with the cytoskeletal force model of integrin activation.


Subject(s)
Intercellular Adhesion Molecule-1/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry , Magnesium/chemistry , Manganese/chemistry , HEK293 Cells , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Manganese/metabolism , Protein Domains , Protein Structure, Quaternary , X-Ray Diffraction
3.
Proc Natl Acad Sci U S A ; 113(11): 2940-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26936951

ABSTRACT

High-resolution crystal structures of the headpiece of lymphocyte function-associated antigen-1 (integrin αLß2) reveal how the αI domain interacts with its platform formed by the α-subunit ß-propeller and ß-subunit ßI domains. The αLß2 structures compared with αXß2 structures show that the αI domain, tethered through its N-linker and a disulfide to a stable ß-ribbon pillar near the center of the platform, can undergo remarkable pivoting and tilting motions that appear buffered by N-glycan decorations that differ between αL and αX subunits. Rerefined ß2 integrin structures reveal details including pyroglutamic acid at the ß2 N terminus and bending within the EGF1 domain. Allostery is relayed to the αI domain by an internal ligand that binds to a pocket at the interface between the ß-propeller and ßI domains. Marked differences between the αL and αX subunit ß-propeller domains concentrate near the binding pocket and αI domain interfaces. Remarkably, movement in allostery in the ßI domain of specificity determining loop 1 (SDL1) causes concerted movement of SDL2 and thereby tightens the binding pocket for the internal ligand.


Subject(s)
Lymphocyte Function-Associated Antigen-1/chemistry , Allosteric Regulation , Allosteric Site , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Consensus Sequence , Crystallography, X-Ray , Humans , Leukocytes/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Motion , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
4.
J Immunol ; 196(7): 3019-31, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26921307

ABSTRACT

Regulation of thymocyte trafficking plays an important role during thymic selection, but our understanding of the molecular mechanisms underlying these processes is limited. In this study, we demonstrated that class III semaphorin E (sema3e), a guidance molecule during neural and vascular development, directly inhibited Rap1 activation and LFA-1-dependent adhesion through the GTPase-activating protein activity of plexin D1. Sema3e inhibited Rap1 activation of thymocytes in response to chemokines and TCR stimulation, LFA-mediated adhesion, and T cell-APC interactions. Immunological synapse (IS) formation in mature thymocytes on supported lipid bilayers was also attenuated by sema3e. Impaired IS formation was associated with reduced Rap1 activation on the contact surface and cell periphery. Moreover, a significant increase of CD4(+) thymocytes was detected in the medulla of mice with T cell lineage-specific deletion of plexin D1. Two-photon live imaging of thymic explants and slices revealed enhanced Rap1 activation and migration of CD69(+) double-positive and single-positive cells with plexin D1 deficiency. Our results demonstrate that sema3e/plexin D1 modulates IS formation and Ag-scanning activities of thymocytes within thymic tissues.


Subject(s)
Glycoproteins/metabolism , Immunological Synapses/metabolism , Immunomodulation , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Thymocytes/immunology , Thymocytes/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Adhesion/genetics , Cell Communication , Cell Movement/genetics , Chemokines/metabolism , Cytoskeletal Proteins , Glycoproteins/genetics , Intracellular Signaling Peptides and Proteins , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs , Semaphorins , Signal Transduction , rac GTP-Binding Proteins/metabolism
5.
J Chem Phys ; 148(12): 123332, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29604893

ABSTRACT

Cell adhesion complexes (CACs), which are activated by ligand binding, play key roles in many cellular functions ranging from cell cycle regulation to mediation of cell extracellular matrix adhesion. Inspired by single molecule pulling experiments using atomic force spectroscopy on leukocyte function-associated antigen-1 (LFA-1), expressed in T-cells, bound to intercellular adhesion molecules (ICAM), we performed constant loading rate (rf) and constant force (F) simulations using the self-organized polymer model to describe the mechanism of ligand rupture from CACs. The simulations reproduce the major experimental finding on the kinetics of the rupture process, namely, the dependence of the most probable rupture forces (f*s) on ln rf (rf is the loading rate) exhibits two distinct linear regimes. The first, at low rf, has a shallow slope, whereas the slope at high rf is much larger, especially for a LFA-1/ICAM-1 complex with the transition between the two occurring over a narrow rf range. Locations of the two transition states (TSs) extracted from the simulations show an abrupt change from a high value at low rf or constant force, F, to a low value at high rf or F. This unusual behavior in which the CACs switch from one brittle (TS position is a constant over a range of forces) state to another brittle state is not found in forced-rupture in other protein complexes. We explain this novel behavior by constructing the free energy profiles, F(Λ)s, as a function of a collective reaction coordinate (Λ), involving many key charged residues and a critical metal ion (Mg2+). The TS positions in F(Λ), which quantitatively agree with the parameters extracted using the Bell-Evans model, change abruptly at a critical force, demonstrating that it, rather than the molecular extension, is a good reaction coordinate. Our combined analyses using simulations performed in both the pulling modes (constant rf and F) reveal a new mechanism for the two loading regimes observed in the rupture kinetics in CACs.


Subject(s)
Coordination Complexes/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry , Cell Adhesion , Ions , Kinetics , Ligands , Lymphocyte Function-Associated Antigen-1/physiology , Magnesium/chemistry , Microscopy, Atomic Force , Physical Phenomena
6.
Am J Transplant ; 17(5): 1193-1203, 2017 May.
Article in English | MEDLINE | ID: mdl-27888551

ABSTRACT

Costimulation blockade (CoB) via belatacept is a lower-morbidity alternative to calcineurin inhibitor (CNI)-based immunosuppression. However, it has higher rates of early acute rejection. These early rejections are mediated in part by memory T cells, which have reduced dependence on the pathway targeted by belatacept and increased adhesion molecule expression. One such molecule is leukocyte function antigen (LFA)-1. LFA-1 exists in two forms: a commonly expressed, low-affinity form and a transient, high-affinity form, expressed only during activation. We have shown that antibodies reactive with LFA-1 regardless of its configuration are effective in eliminating memory T cells but at the cost of impaired protective immunity. Here we test two novel agents, leukotoxin A and AL-579, each of which targets the high-affinity form of LFA-1, to determine whether this more precise targeting prevents belatacept-resistant rejection. Despite evidence of ex vivo and in vivo ligand-specific activity, neither agent when combined with belatacept proved superior to belatacept monotherapy. Leukotoxin A approached a ceiling of toxicity before efficacy, while AL-579 failed to significantly alter the peripheral immune response. These data, and prior studies, suggest that LFA-1 blockade may not be a suitable adjuvant agent for CoB-resistant rejection.


Subject(s)
Abatacept/pharmacology , Graft Rejection/drug therapy , Graft Survival/immunology , Immunologic Memory/immunology , Kidney Transplantation/adverse effects , Lymphocyte Function-Associated Antigen-1/chemistry , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Glomerular Filtration Rate , Graft Rejection/etiology , Graft Rejection/pathology , Graft Survival/drug effects , Immunologic Memory/drug effects , Immunosuppressive Agents/pharmacology , Kidney Function Tests , Lymphocyte Function-Associated Antigen-1/metabolism , Macaca mulatta , Postoperative Complications , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
7.
Biotechnol Lett ; 39(12): 1843-1851, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28918491

ABSTRACT

OBJECTIVES: To provide insight into the dynamics of the shape-shifting mechanistic events associated with the opening (activation) of Lymphocyte Function Associated Antigen-1 upon allosteric modulation by an activator, ICAM Binding Enhancer-667 (IBE-667), using molecular dynamics simulation. RESULTS: Various parameters were used to appropriately describe and understand the sequence of events that characterized its activation across the simulation period such as residual distances, TriCα angles; as well as the dihedral angle. Our findings revealed a significant residual fluctuation and stability difference between both systems. Also, there was a synergistic coordination of the active MIDAS site by the downward pull of the α7 helix upon ligand binding, which appeared to be directly proportional to each other. CONCLUSION: Allosteric binding of IBE-667, activated LFA-1 integrin as evidenced by residual motion at the MIDAS region which appears to be synergistically coordinated by the downward pull of the α7 helix.


Subject(s)
Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Signal Transduction/physiology , Azepines/chemistry , Azepines/metabolism , Computational Biology , Humans , Indazoles/chemistry , Indazoles/metabolism , Molecular Dynamics Simulation , Protein Binding
8.
Biochem Biophys Res Commun ; 458(2): 251-5, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25640842

ABSTRACT

We have shown that Mg/EGTA (5 mM Mg(2+) and 1.5 mM EGTA) could effectively promote the adhesion of integrin αLß2 to its ligand ICAM-1 but could not promote that of the αMß2 to denatured BSA. In order to determine the structural differences between αL and αM that specifically contribute to Mg/EGTA sensitivity, a series of αL/αM chimeras were constructed. Our results showed that αLß2 with αM calf-1 domain completely lost the response to Mg/EGTA activation. In the reverse experiment, αMß2 would require the presence of both the αL calf-1 and calf-2 domain to initiate the Mg/EGTA sensitivity.


Subject(s)
Cell Adhesion/physiology , Egtazic Acid/chemistry , Egtazic Acid/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/chemistry , Magnesium/metabolism , Binding Sites , HEK293 Cells , Humans , Protein Binding , Structure-Activity Relationship
9.
J Phys Chem A ; 119(43): 10735-42, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26461140

ABSTRACT

Enflurane is a fluorinated volatile anesthetic, whose bioactive conformation is not known. Actually, a few studies have reported on the conformations of enflurane in nonpolar solution and gas phase. The present computational and spectroscopic (infrared and NMR) work shows that three pairs of isoenergetic conformers take place in the gas phase, neat liquid, polar, and nonpolar solutions. According to docking studies, a single conformation is largely preferred over its isoenergetic isomers to complex with the active site of Integrin LFA-1 enzyme (PDB code: 3F78 ), where the widely used anesthetic isoflurane (a constitutional isomer of enflurane) is known to bind. Weak hydrogen bonding from an electrostatic interaction between the CHF2 hydrogen and the central CF2 fluorines was not found to rule the conformational isomerism of enflurane. Moreover, intramolecular interactions based on steric, electrostatic, and hyperconjugative effects usually invoked to describe the anomeric effect are not responsible for the possible bioactive conformation of enflurane, which is rather governed by the enzyme induced fit.


Subject(s)
Enflurane/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Catalytic Domain , Lymphocyte Function-Associated Antigen-1/chemistry , Molecular Conformation , Molecular Docking Simulation , Quantum Theory , Solutions , Thermodynamics
10.
Proc Natl Acad Sci U S A ; 109(13): 4869-74, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411821

ABSTRACT

Integrins are cell membrane adhesion receptors involved in morphogenesis, immunity, tissue healing, and metastasis. A central, yet unresolved question regarding the function of integrins is how these receptors regulate both their conformation and dynamic nanoscale organization on the membrane to generate adhesion-competent microclusters upon ligand binding. Here we exploit the high spatial (nanometer) accuracy and temporal resolution of single-dye tracking to dissect the relationship between conformational state, lateral mobility, and microclustering of the integrin receptor lymphocyte function-associated antigen 1 (LFA-1) expressed on immune cells. We recently showed that in quiescent monocytes, LFA-1 preorganizes in nanoclusters proximal to nanoscale raft components. We now show that these nanoclusters are primarily mobile on the cell surface with a small (ca. 5%) subset of conformational-active LFA-1 nanoclusters preanchored to the cytoskeleton. Lateral mobility resulted crucial for the formation of microclusters upon ligand binding and for stable adhesion under shear flow. Activation of high-affinity LFA-1 by extracellular Ca(2+) resulted in an eightfold increase on the percentage of immobile nanoclusters and cytoskeleton anchorage. Although having the ability to bind to their ligands, these active nanoclusters failed to support firm adhesion in static and low shear-flow conditions because mobility and clustering capacity were highly compromised. Altogether, our work demonstrates an intricate coupling between conformation and lateral diffusion of LFA-1 and further underscores the crucial role of mobility for the onset of LFA-1 mediated leukocyte adhesion.


Subject(s)
Lymphocyte Function-Associated Antigen-1/metabolism , Monocytes/cytology , Monocytes/metabolism , Nanoparticles/chemistry , Actin Cytoskeleton/metabolism , Calcium/metabolism , Cell Adhesion , Cell Line , Cell Movement , Cluster Analysis , Diffusion , Extracellular Space/metabolism , Humans , Lymphocyte Function-Associated Antigen-1/chemistry , Protein Transport , Rheology , Stress, Mechanical
11.
J Biol Chem ; 288(32): 23607-21, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23792963

ABSTRACT

Aggregatibacter actinomycetemcomitans produces a repeats-in-toxin (RTX) leukotoxin (LtxA) that selectively kills human immune cells. Binding of LtxA to its ß2 integrin receptor (lymphocyte function-associated antigen-1 (LFA-1)) results in the clustering of the toxin·receptor complex in lipid rafts. Clustering occurs only in the presence of LFA-1 and cholesterol, and LtxA is unable to kill cells lacking either LFA-1 or cholesterol. Here, the interaction of LtxA with cholesterol was measured using surface plasmon resonance and differential scanning calorimetry. The binding of LtxA to phospholipid bilayers increased by 4 orders of magnitude in the presence of 40% cholesterol relative to the absence of cholesterol. The affinity was specific to cholesterol and required an intact secondary structure. LtxA contains two cholesterol recognition/amino acid consensus (CRAC) sites; CRAC(336) ((333)LEEYSKR(339)) is highly conserved among RTX toxins, whereas CRAC(503) ((501)VDYLK(505)) is unique to LtxA. A peptide corresponding to CRAC(336) inhibited the ability of LtxA to kill Jurkat (Jn.9) cells. Although peptides corresponding to both CRAC(336) and CRAC(503) bind cholesterol, only CRAC(336) competitively inhibited LtxA binding to this sterol. A panel of full-length LtxA CRAC mutants demonstrated that an intact CRAC(336) site was essential for LtxA cytotoxicity. The conservation of CRAC(336) among RTX toxins suggests that this mechanism may be conserved among RTX toxins.


Subject(s)
Bacterial Toxins/chemistry , Cholesterol/chemistry , Exotoxins/chemistry , Membrane Microdomains/chemistry , Pasteurellaceae/chemistry , Amino Acid Motifs , Bacterial Toxins/metabolism , Cholesterol/metabolism , Exotoxins/metabolism , Humans , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Microdomains/metabolism , Pasteurellaceae/metabolism , Protein Binding , Surface Plasmon Resonance
12.
Am J Pathol ; 183(5): 1548-1558, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24055259

ABSTRACT

Cannabinoid receptor 2 (CB2) is highly expressed in immune cells and stimulation decreases inflammatory responses. We tested the idea that selective CB2 activation in human monocytes suppresses their ability to engage the brain endothelium and migrate across the blood-brain barrier (BBB), preventing consequent injury. Intravital videomicroscopy was used to quantify adhesion of leukocytes to cortical vessels in lipopolysaccharide-induced neuroinflammation, after injection of ex vivo CB2-activated leukocytes into mice; CB2 agonists markedly decreased adhesion of ex vivo labeled cells in vivo. In an in vitro BBB model, CB2 activation in monocytes largely attenuated adhesion to and migration across monolayers of primary human brain microvascular endothelial cells and diminished BBB damage. CB2 stimulation in monocytes down-regulated active forms of integrins, lymphocyte function-associated antigen 1 (LFA-1), and very late antigen 4 (VLA-4). Cells treated with CB2 agonists exhibited increased phosphorylation levels of inhibitory sites of the actin-binding proteins cofilin and VASP, which are upstream regulators of conformational integrin changes. Up-regulated by relevant stimuli, Rac1 and RhoA were suppressed by CB2 agonists in monocytes. CB2 stimulation decreased formation of lamellipodia, which play a key role in monocyte migration. These results indicate that selective CB2 activation in leukocytes decreases key steps in monocyte-BBB engagement, thus suppressing inflammatory leukocyte responses and preventing neuroinflammation.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Endothelium/metabolism , Leukocytes/metabolism , Receptor, Cannabinoid, CB2/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Line , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Encephalitis/metabolism , Encephalitis/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium/pathology , Humans , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/metabolism , Integrin beta1/metabolism , Lipopolysaccharides , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Microfilament Proteins/metabolism , Microvessels/pathology , Monocytes/metabolism , Monocytes/pathology , Phosphoproteins/metabolism , Phosphorylation , Pseudopodia/metabolism , Receptor, Cannabinoid, CB2/agonists , Transendothelial and Transepithelial Migration , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein
13.
Blood ; 119(18): 4275-82, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22431571

ABSTRACT

In inflammation, neutrophils and other leukocytes roll along the microvascular endothelium before arresting and transmigrating into inflamed tissues. Arrest requires conformational activation of the integrin lymphocyte function-associated antigen-1 (LFA-1). Mutations of the FERMT3 gene encoding kindlin-3 underlie the human immune deficiency known as leukocyte adhesion deficiency-III. Both kindlin-3 and talin-1, another FERM domain-containing cytoskeletal protein, are required for integrin activation, but their individual roles in the induction of specific integrin conformers are unclear. Here, we induce differential LFA-1 activation in neutrophils through engagement of the selectin ligand P-selectin glycoprotein ligand-1 or the chemokine receptor CXCR2. We find that talin-1 is required for inducing LFA-1 extension, which corresponds to intermediate affinity and induces neutrophil slow rolling, whereas both talin-1 and kindlin-3 are required for induction of the high-affinity conformation of LFA-1 with an open headpiece, which results in neutrophil arrest. In vivo, both slow rolling and arrest are defective in talin-1-deficient neutrophils, whereas only arrest is defective in kindlin-3-deficient neutrophils. We conclude that talin-1 and kindlin-3 serve distinct functions in LFA-1 activation.


Subject(s)
Chemotaxis, Leukocyte/physiology , Cytoskeletal Proteins/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Neutrophils/metabolism , Talin/physiology , Animals , Bone Marrow Transplantation , Cell Adhesion , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Green Fluorescent Proteins/analysis , HL-60 Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , K562 Cells , Lymphocyte Function-Associated Antigen-1/chemistry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/metabolism , Protein Binding , Protein Conformation , RNA Interference , RNA, Small Interfering/pharmacology , Radiation Chimera , Specific Pathogen-Free Organisms , Talin/antagonists & inhibitors , Talin/genetics
14.
Angew Chem Int Ed Engl ; 53(17): 4322-6, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24692345

ABSTRACT

Stabilization of protein-protein interactions by small molecules is a concept with few examples reported to date. Herein we describe the identification and X-ray co-crystal structure determination of IBE-667, an ICAM-1 binding enhancer for LFA-1. IBE-667 was designed based on the SAR information obtained from an on-bead screen of tagged one-bead one-compound combinatorial libraries by confocal nanoscanning and bead picking (CONA). Cellular assays demonstrate the activity of IBE-667 in promoting the binding of LFA-1 on activated immune cells to ICAM-1.


Subject(s)
Azepines/chemistry , Azepines/pharmacology , Indazoles/chemistry , Indazoles/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Binding/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Combinatorial Chemistry Techniques , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Intercellular Adhesion Molecule-1/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry
15.
Gene Ther ; 20(11): 1042-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23759703

ABSTRACT

Systemic and target-specific delivery of large genetic contents has been difficult to achieve. Although viruses effortlessly deliver kilobase-long genome into cells, its clinical use has been hindered by serious safety concerns and the mismatch between native tropisms and desired targets. Nonviral vectors, in contrast, are limited by low gene transfer efficiency and inherent cytotoxicity. Here we devised virus-mimetic polyplex particles (VMPs) based on electrostatic self-assembly among polyanionic peptide (PAP), cationic polymer polyethyleneimine (PEI) and nucleic acids. We fused PAP to the engineered ligand-binding domain of integrin αLß2 to target intercellular adhesion molecule-1 (ICAM-1), an inducible marker of inflammation. Fully assembled VMPs packaged large genetic contents, bound specifically to target molecules, elicited receptor-mediated endocytosis and escaped endosomal pathway, resembling intracellular delivery processes of viruses. Unlike conventional PEI-mediated transfection, molecular interaction-dependent gene delivery of VMPs was unaffected by the presence of serum and achieved higher efficiency without toxicity. By targeting overexpressed ICAM-1, VMPs delivered genes specifically to inflamed endothelial cells and macrophages both in vitro and in vivo. Simplicity and versatility of the platform and inflammation-specific delivery may open up opportunities for multifaceted gene therapy that can be translated into the clinic and treat a broad range of debilitating immune and inflammatory diseases.


Subject(s)
Biomimetic Materials , Gene Targeting/methods , Genetic Therapy/methods , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Vectors , HeLa Cells , Humans , Immune System Diseases/therapy , Inflammation/therapy , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/genetics , Mice , Mice, Inbred BALB C , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacokinetics , Virion/chemistry , Virion/genetics
16.
FASEB J ; 26(11): 4408-17, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22815384

ABSTRACT

We previously demonstrated that isoflurane targets lymphocyte function-associated antigen-1 (LFA-1), a critical adhesion molecule for leukocyte arrest. However, it remains to be determined how isoflurane interacts with the full ectodomain LFA-1 and modulates its conformation and function. Isoflurane binding sites on the full ectodomain LFA-1 were probed by photolabeling using photoactivatable isoflurane (azi-isoflurane). The adducted residues were determined by liquid chromatography/mass spectrometry analysis. Separately, docking simulations were performed to predict binding sites. Point mutations were introduced around isoflurane binding sites. The significance of isoflurane's effect was assessed in both intracellular adhesion molecule-1 (ICAM-1) binding assays and epitope mapping of activation-sensitive antibodies using flow cytometry. Two isoflurane binding sites were identified using photolabeling and were further validated by the docking simulation: one at the hydrophobic pocket in the ICAM-1 binding domain (the αI domain); the other at the ßI domain. Mutagenesis of the α'1 helix showed that isoflurane binding sites at the ßI domain were significantly important in modulating LFA-1 function and conformation. Epitope mapping using activation-sensitive antibodies suggested that isoflurane stabilized LFA-1 in the closed conformation. This study suggested that isoflurane binds to both the αI and ßI domains allosteric to the ICAM-1 binding site, and that isoflurane binding stabilizes LFA-1 in the closed conformation.


Subject(s)
Isoflurane/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Amino Acid Sequence , Antibodies, Monoclonal , Binding Sites , HEK293 Cells , Humans , Isoflurane/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Conformation , Protein Structure, Tertiary
17.
Anesth Analg ; 117(4): 803-811, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23960033

ABSTRACT

BACKGROUND: We previously demonstrated that propofol interacted with the leukocyte adhesion molecule leukocyte function-associated antigen-1 (LFA-1) and inhibited the production of interleukin-2 via LFA-1 in a dependent manner. However, the binding site(s) of propofol on LFA-1 remains unknown. METHODS: First, the inhibition of LFA-1's ligand binding by propofol was confirmed in an enzyme-linked immunosorbent assay (ELISA) ELISA-type assay. The binding site of propofol on LFA-1 was probed with a photolabeling experiment using a photoactivatable propofol analog called azi-propofol-m. The adducted residues of LFA-1 by this compound were determined using liquid chromatography-mass spectrometry. In addition, the binding of propofol to the ligand-binding domain of LFA-1 was examined using 1-aminoanthracene (1-AMA) displacement assay. Furthermore, the binding site(s) of 1-AMA and propofol on LFA-1 was studied using the docking program GLIDE. RESULTS: We demonstrated that propofol impaired the binding of LFA-1 to its ligand intercellular adhesion molecule-1. The photolabeling experiment demonstrated that the adducted residues were localized in the allosteric cavity of the ligand-binding domain of LFA-1 called "lovastatin site." The shift of fluorescence spectra was observed when 1-AMA was coincubated with the low-affinity conformer of LFA-1 ligand-binding domain (wild-type [WT] αL I domain), not with the high-affinity conformer, suggesting that 1-AMA bound only to WT αL I domain. In the 1-AMA displacement assay, propofol decreased 1-AMA fluorescence signal (at 520 nm), suggesting that propofol competed with 1-AMA and bound to the WT αL I domain. The docking simulation demonstrated that both 1-AMA and propofol bound to the lovastatin site, which agreed with the photolabeling experiment. CONCLUSIONS: We demonstrated that propofol bound to the lovastatin site in LFA-1. Previously we showed that the volatile anesthetics isoflurane and sevoflurane bound to this site. Taken together, the lovastatin site is an example of the common binding sites for anesthetics currently used clinically.


Subject(s)
Anesthetics, Inhalation/metabolism , Anesthetics, Intravenous/metabolism , Isoflurane/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Methyl Ethers/metabolism , Propofol/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Ligands , Lovastatin/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Protein Binding/physiology , Sevoflurane
18.
Biochem Pharmacol ; 211: 115504, 2023 05.
Article in English | MEDLINE | ID: mdl-36921634

ABSTRACT

Integrins are a family of cell surface receptors well-recognized for their therapeutic potential in a wide range of diseases. However, the development of integrin targeting medications has been impacted by unexpected downstream effects, reflecting originally unforeseen interference with the bidirectional signalling and cross-communication of integrins. We here selected one of the most severely affected target integrins, the integrin lymphocyte function-associated antigen-1 (LFA-1, αLß2, CD11a/CD18), as a prototypic integrin to systematically assess and overcome these known shortcomings. We employed a two-tiered ligand-based virtual screening approach to identify a novel class of allosteric small molecule inhibitors targeting this integrin's αI domain. The newly discovered chemical scaffold was derivatized, yielding potent bis-and tris-aryl-bicyclic-succinimides which inhibit LFA-1 in vitro at low nanomolar concentrations. The characterisation of these compounds in comparison to earlier LFA-1 targeting modalities established that the allosteric LFA-1 inhibitors (i) are devoid of partial agonism, (ii) selectively bind LFA-1 versus other integrins, (iii) do not trigger internalization of LFA-1 itself or other integrins and (iv) display oral availability. This profile differentiates the new generation of allosteric LFA-1 inhibitors from previous ligand mimetic-based LFA-1 inhibitors and anti-LFA-1 antibodies, and is projected to support novel immune regulatory regimens selectively targeting the integrin LFA-1. The rigorous computational and experimental assessment schedule described here is designed to be adaptable to the preclinical discovery and development of novel allosterically acting compounds targeting integrins other than LFA-1, providing an exemplary approach for the early characterisation of next generation integrin inhibitors.


Subject(s)
Lymphocyte Function-Associated Antigen-1 , Signal Transduction , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Ligands , Intercellular Adhesion Molecule-1/metabolism
19.
EMBO J ; 27(1): 62-75, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18079697

ABSTRACT

T lymphocytes use LFA-1 to migrate into lymph nodes and inflammatory sites. To investigate the mechanisms regulating this migration, we utilize mAbs selective for conformational epitopes as probes for active LFA-1. Expression of the KIM127 epitope, but not the 24 epitope, defines the extended conformation of LFA-1, which has intermediate affinity for ligand ICAM-1. A key finding is that KIM127-positive LFA-1 forms new adhesions at the T lymphocyte leading edge. This LFA-1 links to the cytoskeleton through alpha-actinin-1 and disruption at the level of integrin or actin results in loss of cell spreading and migratory speed due to a failure of attachment at the leading edge. The KIM127 pattern contrasts with high-affinity LFA-1 that expresses both 24 and KIM127 epitopes, is restricted to the mid-cell focal zone and controls ICAM-1 attachment. Identification of distinctive roles for intermediate- and high-affinity LFA-1 in T lymphocyte migration provides a biological function for two active conformations of this integrin for the first time.


Subject(s)
Actinin/metabolism , Cell Movement/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Humans , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/physiology , Molecular Sequence Data , Protein Binding/immunology , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Isoforms/metabolism , Protein Isoforms/physiology , T-Lymphocytes/immunology
20.
Blood ; 115(4): 804-14, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19965628

ABSTRACT

The small GTPase Rap1 and its effector RAPL regulate lymphocyte adhesion and motility. However, their precise regulatory roles in the adhesion cascade preceding entry into lymph nodes and during interstitial migration are unclear. Here, we show that Rap1 is indispensably required for the chemokine-triggered initial arrest step of rolling lymphocytes through LFA-1, whereas RAPL is not involved in rapid arrest. RAPL and talin play a critical role in stabilizing lymphocyte arrest to the endothelium of blood vessels under flow or to the high endothelial venules of peripheral lymph nodes in vivo. Further, mutagenesis and peptide studies suggest that release of a trans-acting restraint from the beta2 cytoplasmic region of LFA-1 is critical for Rap1-dependent initial arrest. Rap1 or RAPL deficiency severely impaired lymphocyte motility over lymph node stromal cells in vitro, and RAPL deficiency impaired high-velocity directional movement within lymph nodes. These findings reveal the several critical steps of Rap1, which are RAPL-dependent and -independent, in lymphocyte trafficking.


Subject(s)
Cell Adhesion/immunology , Cell Movement/immunology , Lymphocytes/cytology , rap1 GTP-Binding Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Line , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Leukocyte Rolling/physiology , Lymph Nodes/cytology , Lymph Nodes/physiology , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes/metabolism , Mice , Mutagenesis , Protein Structure, Tertiary , RNA Interference , Signal Transduction/immunology , Talin/genetics , Talin/metabolism , Transfection , rap1 GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL