Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
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
2.
Methods Mol Biol ; 2217: 3-15, 2021.
Article in English | MEDLINE | ID: mdl-33215372

ABSTRACT

Soluble ligand and conformation-dependent antibody binding assay of integrins on the cell surface is an effective approach to evaluate the activation status of integrins in live cells. The ligands or antibodies are usually labeled with biotin or a fluorescent dye and incubated with integrin-expressing cells in suspension. The cell-bound ligands and antibodies are then detected by flow cytometry. Here we describe the detailed protocols of soluble ligand or antibody binding assay for αIIbß3, αVß3, α5ß1, and αLß2 integrins that are transiently or stably expressed in the model cell lines such as HEK293 or CHO-k1 cells.


Subject(s)
Biological Assay , Integrin alphaVbeta3/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry , Platelet Membrane Glycoprotein IIb/chemistry , Receptors, Vitronectin/chemistry , Staining and Labeling/methods , Animals , Antibodies/chemistry , Antibodies/metabolism , CHO Cells , Cell Adhesion , Cricetulus , Flow Cytometry , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Intercellular Adhesion Molecule-1 , Ligands , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Plasmids/chemistry , Plasmids/metabolism , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding , Receptors, Vitronectin/genetics , Receptors, Vitronectin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
3.
Biosci Rep ; 41(1)2021 01 29.
Article in English | MEDLINE | ID: mdl-33305306

ABSTRACT

ORF7a is an accessory protein common to SARS-CoV1 and the recently discovered SARS-CoV2, which is causing the COVID-19 pandemic. The ORF7a protein has a structural homology with ICAM-1 which binds to the T lymphocyte integrin receptor LFA-1. As COVID-19 has a strong immune component as part of the disease, we sought to determine whether SARS-CoV2 would have a similar structural interaction with LFA-1. Using molecular docking simulations, we found that SARS-CoV2 ORF7a has the key structural determinants required to bind LFA-1 but also the related leukocyte integrin Mac-1, which is also known to be expressed by macrophages. Our study shows that SARS-CoV2 ORF7a protein has a conserved Ig immunoglobulin-like fold containing an integrin binding site that provides a mechanistic hypothesis for SARS-CoV2's interaction with the human immune system. This suggests that experimental investigation of ORF7a-mediated effects on immune cells such as T lymphocytes and macrophages (leukocytes) could help understand the disease further and develop effective treatments.


Subject(s)
COVID-19/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Macrophage-1 Antigen/immunology , SARS-CoV-2/immunology , Viral Proteins/chemistry , Viral Proteins/immunology , Binding Sites , Humans , Lymphocyte Function-Associated Antigen-1/chemistry , Macrophage-1 Antigen/chemistry , Molecular Docking Simulation , Protein Conformation , Protein Interaction Domains and Motifs , SARS-CoV-2/chemistry
4.
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
5.
PLoS One ; 13(10): e0205871, 2018.
Article in English | MEDLINE | ID: mdl-30335797

ABSTRACT

The oral bacterium, Aggregatibacter actinomycetemcomitans, which is associated with localized aggressive periodontitis, as well as systemic infections including endocarditis, produces numerous virulence factors, including a repeats-in-toxin (RTX) protein called leukotoxin (LtxA), which kills human immune cells. The strains of A. actinomycetemcomitans most closely associated with disease have been shown to produce the most LtxA, suggesting that LtxA plays a significant role in the virulence of this organism. LtxA, like many of the RTX toxins, can be divided into four functional domains: an N-terminal hydrophobic domain, which contains a significant fraction of hydrophobic residues and has been proposed to play a role in the membrane interaction of the toxin; the central domain, which contains two lysine residues that are the sites of post-translational acylation; the repeat domain that is characteristic of the RTX toxins, and a C-terminal domain thought to be involved in secretion. In its initial interaction with the host cell, LtxA must bind to both cholesterol and an integrin receptor, lymphocyte function-associated antigen-1 (LFA-1). While both interactions are essential for toxicity, the domains of LtxA involved remain unknown. We therefore undertook a series of experiments, including tryptophan quenching and trypsin digestion, to characterize the structure of LtxA upon interaction with membranes of various lipid compositions. Our results demonstrate that LtxA adopts a U-shaped conformation in the membrane, with the N- and C-terminal domains residing outside of the membrane.


Subject(s)
Aggregatibacter actinomycetemcomitans/chemistry , Bacterial Proteins/chemistry , Cholesterol/chemistry , Hemolysin Proteins/chemistry , Lymphocyte Function-Associated Antigen-1/chemistry , Virulence Factors/chemistry , Aggregatibacter actinomycetemcomitans/growth & development , Aggregatibacter actinomycetemcomitans/pathogenicity , Amino Acid Sequence , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Binding Sites , Cholesterol/metabolism , Dimyristoylphosphatidylcholine/chemistry , Dimyristoylphosphatidylcholine/metabolism , Hemolysin Proteins/isolation & purification , Hemolysin Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Jurkat Cells , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Liposomes/chemistry , Liposomes/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proteolysis , Trypsin/chemistry , Virulence Factors/isolation & purification , Virulence Factors/metabolism
6.
ACS Infect Dis ; 4(7): 1073-1081, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29742342

ABSTRACT

The Gram-negative bacterium Aggregatibacter actinomycetemcomitans, commonly associated with localized aggressive periodontitis (LAP), secretes an RTX (repeats-in-toxin) protein leukotoxin (LtxA) that targets human white blood cells, an interaction that is driven by its recognition of the lymphocyte function-associated antigen-1 (LFA-1) integrin. In this study, we report on the inhibition of LtxA-LFA-1 binding as an antivirulence strategy to inhibit LtxA-mediated cytotoxicity. Specifically, we designed and synthesized peptides corresponding to the reported LtxA binding domain on LFA-1 and characterized their capability to inhibit LtxA binding to LFA-1 and subsequent cytotoxic activity in human immune cells. We found that several of these peptides, corresponding to sequential ß-strands in the LtxA-binding domain of LFA-1, inhibit LtxA activity, demonstrating the effectiveness of this approach. Further investigations into the mechanism by which these peptides inhibit LtxA binding to LFA-1 reveal a correlation between toxin-peptide affinity and LtxA-mediated cytotoxicity, leading to a diminished association between LtxA and LFA-1 on the cell membrane. Our results demonstrate the possibility of using target-based peptides to inhibit LtxA activity, and we expect that a similar approach could be used to hinder the activity of other RTX toxins.


Subject(s)
Anti-Bacterial Agents/pharmacology , Exotoxins/antagonists & inhibitors , Lymphocyte Function-Associated Antigen-1/chemistry , Peptides/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Exotoxins/chemistry , Exotoxins/toxicity , Humans , Lymphocyte Function-Associated Antigen-1/pharmacology , Models, Biological , Peptides/chemistry , Protein Binding , Structure-Activity Relationship , THP-1 Cells , Virulence Factors/antagonists & inhibitors , Virulence Factors/chemistry
7.
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
8.
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
9.
J Chem Theory Comput ; 13(12): 6343-6357, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29112408

ABSTRACT

Allostery describes the functional coupling between sites in biomolecules. Recently, the role of changes in protein dynamics for allosteric communication has been highlighted. A quantitative and predictive description of allostery is fundamental for understanding biological processes. Here, we integrate an ensemble-based perturbation approach with the analysis of biomolecular rigidity and flexibility to construct a model of dynamic allostery. Our model, by definition, excludes the possibility of conformational changes, evaluates static, not dynamic, properties of molecular systems, and describes allosteric effects due to ligand binding in terms of a novel free-energy measure. We validated our model on three distinct biomolecular systems: eglin c, protein tyrosine phosphatase 1B, and the lymphocyte function-associated antigen 1 domain. In all cases, it successfully identified key residues for signal transmission in very good agreement with the experiment. It correctly and quantitatively discriminated between positively or negatively cooperative effects for one of the systems. Our model should be a promising tool for the rational discovery of novel allosteric drugs.


Subject(s)
Lymphocyte Function-Associated Antigen-1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Proteins/chemistry , Allosteric Regulation , Lymphocyte Function-Associated Antigen-1/metabolism , Models, Molecular , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proteins/genetics , Proteins/metabolism , Thermodynamics
10.
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
11.
Cell Rep ; 18(4): 857-865, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28122237

ABSTRACT

T cells are the most abundant cell type found in afferent lymph, but their migration through lymphatic vessels (LVs) remains poorly understood. Performing intravital microscopy in the murine skin, we imaged T cell migration through afferent LVs in vivo. T cells entered into and actively migrated within lymphatic capillaries but were passively transported in contractile collecting vessels. Intralymphatic T cell number and motility were increased during contact-hypersensitivity-induced inflammation and dependent on ICAM-1/LFA-1 interactions. In vitro, blockade of endothelial cell-expressed ICAM-1 reduced T cell adhesion, crawling, and transmigration across lymphatic endothelium and decreased T cell advancement from capillaries into lymphatic collectors in skin explants. In vivo, T cell migration to draining lymph nodes was significantly reduced upon ICAM-1 or LFA-1 blockade. Our findings indicate that T cell migration through LVs occurs in distinct steps and reveal a key role for ICAM-1/LFA-1 interactions in this process.


Subject(s)
Inflammation/immunology , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Skin/metabolism , T-Lymphocytes/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Adhesion/drug effects , Cell Movement/drug effects , Flow Cytometry , Inflammation/chemically induced , Inflammation/pathology , Intercellular Adhesion Molecule-1/chemistry , Interferon-gamma/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Lymphatic Vessels/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Oxazolone/toxicity , Skin/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Time-Lapse Imaging , Tumor Necrosis Factor-alpha/pharmacology
12.
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
13.
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
14.
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
15.
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
16.
Structure ; 23(4): 745-53, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25773142

ABSTRACT

Lymphocyte function-associated antigen 1 (LFA-1) is an integrin that transmits information in two directions across the plasma membrane of leukocytes, in so-called outside-in and inside-out signaling mechanisms. To investigate the structural basis of these mechanisms, we studied the conformational space of the apo I-domain using replica-averaged metadynamics simulations in combination with nuclear magnetic resonance chemical shifts. We thus obtained a free energy landscape that reveals the existence of three conformational substates of this domain. The three substates include conformations similar to existing crystallographic structures of the low-affinity I-domain, the inactive I-domain with an allosteric antagonist inhibitor bound underneath α helix 7, and an intermediate affinity state of the I-domain. The multiple substates were validated with residual dipolar coupling measurements. These results suggest that the presence of three substates in the apo I-domain enables the precise regulation of the binding process that is essential for the physiological function of LFA-1.


Subject(s)
Lymphocyte Function-Associated Antigen-1/chemistry , Molecular Dynamics Simulation , Allosteric Regulation , Amino Acid Sequence , Molecular Sequence Data , Protein Structure, Tertiary , Signal Transduction
17.
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
18.
Mol Biol Cell ; 26(1): 43-54, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25378583

ABSTRACT

Lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18, αLß2-integrin) and its ligands are essential for adhesion between T-cells and antigen-presenting cells, formation of the immunological synapse, and other immune cell interactions. LFA-1 function is regulated through conformational changes that include the modulation of ligand binding affinity and molecular extension. However, the relationship between molecular conformation and function is unclear. Here fluorescence resonance energy transfer (FRET) with new LFA-1-specific fluorescent probes showed that triggering of the pathway used for T-cell activation induced rapid unquenching of the FRET signal consistent with extension of the molecule. Analysis of the FRET quenching at rest revealed an unexpected result that can be interpreted as a previously unknown LFA-1 conformation.


Subject(s)
Cell Adhesion , Fluorescence Resonance Energy Transfer , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/chemistry , Cell Line, Tumor , Fluorescent Dyes/chemistry , Humans , Ligands , Molecular Structure , Protein Conformation , T-Lymphocytes/cytology
19.
Protein Sci ; 23(11): 1596-606, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25147050

ABSTRACT

Lymphocyte function-associated antigen-1 (LFA-1) is an integrin protein that transmits information across the plasma membrane through the so-called inside-out and outside-in signaling mechanisms. To investigate these mechanisms, we carried out an NMR analysis of the dynamics of the LFA-1 I-domain, which has enabled us to characterize the motions of this domain on a broad range of timescales. We studied first the internal motions on the nanosecond timescale by spin relaxation measurements and model-free analysis. We then extended this analysis to the millisecond timescale motions by measuring (15) N-(1) H residual dipolar couplings of the backbone amide groups. We analyzed these results in the context of the three major conformational states of the I-domain using their corresponding X-ray crystallographic structures. Our results highlight the importance of the low-frequency motions of the LFA-1 I-domain in the inactive apo-state. We found in particular that α-helix 7 is in a position in the apo-closed state that cannot be fully described by any of the existing X-ray structures, as it appears to be in dynamic exchange between different conformations. This type of motion seems to represent an inherent property of the LFA-1 I-domain and might be relevant for controlling the access to the allosteric binding pocket, as well as for the downward displacement of α-helix 7 that is required for the activation of LFA-1.


Subject(s)
Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation
20.
J Leukoc Biol ; 96(3): 411-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24799603

ABSTRACT

The histamine H4 receptor regulates the inflammatory response. However, it is not known whether this receptor has a functional role in human neutrophils. We found that fMLP (1 µM), but not histamine (0.1-1 µM), induced Mac-1-dependent adhesion, polarization, and degranulation (release of lactoferrin). A pretreatment of neutrophils with histamine (0.001-1 µM) or JNJ 28610244 (0.1-10 µM), a specific H4 receptor agonist, led to inhibition of degranulation. Total inhibition of degranulation was obtained with 0.1 µM histamine and 10 µM JNJ 28610244. Furthermore, such inhibition by histamine of degranulation was reversed by JNJ 7777120 and JNJ 28307474, two selective H4 receptor antagonists. However, neither histamine nor the H4 receptor agonist JNJ 28610244 prevented fMLP-induced, Mac-1-dependent adhesion, indicating that the H4 receptor may block signals emanating from Mac-1-controlling degranulation. Likewise, engagement of the H4 receptor by the selective agonist JNJ 28610244 blocked Mac-1-dependent activation of p38 MAPK, the kinase that controls neutrophil degranulation. We also show expression of the H4 receptor at the mRNA level in ultrapure human neutrophils and myeloid leukemia PLB-985 cells. We concluded that engagement of this receptor by selective H4 receptor agonists may represent a good, therapeutic approach to accelerate resolution of inflammation.


Subject(s)
Cell Degranulation , Neutrophils/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Degranulation/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Cells, Cultured , Cytochalasin B/pharmacology , Fibrinogen , Histamine/pharmacology , Humans , Indoles/pharmacology , Leukemia, Promyelocytic, Acute/pathology , Lymphocyte Function-Associated Antigen-1/chemistry , MAP Kinase Signaling System/drug effects , Macrophage-1 Antigen/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Oximes/pharmacology , Piperazines/pharmacology , Piperidines/pharmacology , Protein Conformation/drug effects , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Histamine H4 , p38 Mitogen-Activated Protein Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL