Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 76
1.
Nat Commun ; 14(1): 2929, 2023 05 22.
Article En | MEDLINE | ID: mdl-37217466

Cytotoxic-T-lymphocyte (CTL) mediated control of HIV-1 is enhanced by targeting highly networked epitopes in complex with human-leukocyte-antigen-class-I (HLA-I). However, the extent to which the presenting HLA allele contributes to this process is unknown. Here we examine the CTL response to QW9, a highly networked epitope presented by the disease-protective HLA-B57 and disease-neutral HLA-B53. Despite robust targeting of QW9 in persons expressing either allele, T cell receptor (TCR) cross-recognition of the naturally occurring variant QW9_S3T is consistently reduced when presented by HLA-B53 but not by HLA-B57. Crystal structures show substantial conformational changes from QW9-HLA to QW9_S3T-HLA by both alleles. The TCR-QW9-B53 ternary complex structure manifests how the QW9-B53 can elicit effective CTLs and suggests sterically hindered cross-recognition by QW9_S3T-B53. We observe populations of cross-reactive TCRs for B57, but not B53 and also find greater peptide-HLA stability for B57 in comparison to B53. These data demonstrate differential impacts of HLAs on TCR cross-recognition and antigen presentation of a naturally arising variant, with important implications for vaccine design.


HIV Infections , Humans , HLA-B Antigens/genetics , T-Lymphocytes, Cytotoxic , Peptides , Epitopes, T-Lymphocyte , Receptors, Antigen, T-Cell
2.
Nat Commun ; 13(1): 6393, 2022 10 27.
Article En | MEDLINE | ID: mdl-36302771

Vaccines targeting HIV-1's gp160 spike protein are stymied by high viral mutation rates and structural chicanery. gp160's membrane-proximal external region (MPER) is the target of naturally arising broadly neutralizing antibodies (bnAbs), yet MPER-based vaccines fail to generate bnAbs. Here, nanodisc-embedded spike protein was investigated by cryo-electron microscopy and molecular-dynamics simulations, revealing spontaneous ectodomain tilting that creates vulnerability for HIV-1. While each MPER protomer radiates centrally towards the three-fold axis contributing to a membrane-associated tripod structure that is occluded in the upright spike, tilting provides access to the opposing MPER. Structures of spike proteins with bound 4E10 bnAb Fabs reveal that the antibody binds exposed MPER, thereby altering MPER dynamics, modifying average ectodomain tilt, and imposing strain on the viral membrane and the spike's transmembrane segments, resulting in the abrogation of membrane fusion and informing future vaccine development.


AIDS Vaccines , HIV-1 , HIV-1/genetics , HIV Envelope Protein gp41/metabolism , HIV Antibodies , Broadly Neutralizing Antibodies , Cryoelectron Microscopy , Spike Glycoprotein, Coronavirus , Antibodies, Neutralizing
4.
J Biol Chem ; 296: 100255, 2021.
Article En | MEDLINE | ID: mdl-33837736

T lymphocytes discriminate between healthy and infected or cancerous cells via T-cell receptor-mediated recognition of peptides bound and presented by cell-surface-expressed major histocompatibility complex molecules (MHCs). Pre-T-cell receptors (preTCRs) on thymocytes foster development of αßT lymphocytes through their ß chain interaction with MHC displaying self-peptides on thymic epithelia. The specific binding of a preTCR with a peptide-MHC complex (pMHC) has been identified previously as forming a weak affinity complex with a distinct interface from that of mature αßTCR. However, a lack of appropriate tools has limited prior efforts to investigate this unique interface. Here we designed a small-scale linkage screening protocol using bismaleimide linkers for determining residue-specific distance constraints between transiently interacting protein pairs in solution. Employing linkage distance restraint-guided molecular modeling, we report the oriented solution docking geometry of a preTCRß-pMHC interaction. The linkage model of preTCRß-pMHC complex was independently verified with paramagnetic pseudocontact chemical shift (PCS) NMR of the unlinked protein mixtures. Using linkage screens, we show that the preTCR binds with differing affinities to peptides presented by MHC in solution. Moreover, the C-terminal peptide segment is a key determinant in preTCR-pMHC recognition. We also describe the process for future large-scale production and purification of the linked constructs for NMR, X-ray crystallography, and single-molecule electron microscopy studies.


Antigens, Surface/ultrastructure , Protein Binding/genetics , Receptors, Antigen, T-Cell/ultrastructure , T-Lymphocytes/ultrastructure , Antigens, Surface/chemistry , Antigens, Surface/genetics , Humans , Major Histocompatibility Complex/genetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Peptides/genetics , Protein Interaction Domains and Motifs/genetics , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/ultrastructure , T-Lymphocytes/chemistry , T-Lymphocytes/immunology , Thymocytes/chemistry , Thymocytes/ultrastructure
5.
Nucleic Acids Res ; 49(7): 4144-4154, 2021 04 19.
Article En | MEDLINE | ID: mdl-33784403

The nucleoprotein (NP) of influenza virus is the core component of the ribonucleoprotein (RNP) and performs multiple structural and functional roles. Structures of the influenza A, B and D NP molecules have been solved previously, but structural information on how NP interacts with RNA remains elusive. Here we present the crystal structure of an obligate monomer of H5N1 NP in complex with RNA nucleotides to 2.3 Å, and a C-terminal truncation of this mutant, also in complex with RNA nucleotides, to 3 Å. In both structures, three nucleotides were identified near two positive grooves of NP suggested to be important for RNA binding. Structural evidence supports that conformational changes of flexible loops and the C-terminal tail both play important roles in the binding of RNA. Based on the structure, we propose a mechanism by which NP captures RNA by flexible loops and transfers it onto the positive binding grooves. Binding of RNA by NP is a crucial step for template re-encapsidation during transcription and replication and cRNP formation. Our structures thus provide insights into the molecular virology of the influenza virus.


Influenza A Virus, H5N1 Subtype , Nucleoproteins/chemistry , RNA, Viral/chemistry , Influenza A Virus, H5N1 Subtype/genetics , Models, Molecular , Protein Binding , Protein Conformation
6.
Science ; 371(6525): 181-185, 2021 01 08.
Article En | MEDLINE | ID: mdl-33335016

Self-discrimination, a critical but ill-defined molecular process programmed during thymocyte development, requires myriad pre-T cell receptors (preTCRs) and αßTCRs. Using x-ray crystallography, we show how a preTCR applies the concave ß-sheet surface of its single variable domain (Vß) to "horizontally" grab the protruding MHC α2-helix. By contrast, αßTCRs purpose all six complementarity-determining region (CDR) loops of their paired VαVß module to recognize peptides bound to major histocompatibility complex molecules (pMHCs) in "vertical" head-to-head binding. The preTCR topological fit ensures that CDR3ß reaches the peptide's featured C-terminal segment for pMHC sampling, establishing the subsequent αßTCR canonical docking mode. "Horizontal" docking precludes germline CDR1ß- and CDR2ß-MHC binding to broaden ß-chain repertoire diversification before αßTCR-mediated selection refinement. Thus, one subunit successively attunes the recognition logic of related multicomponent receptors.


Receptors, Antigen, T-Cell, alpha-beta/chemistry , Thymocytes/immunology , Animals , Crystallography, X-Ray , Humans , Ligands , Major Histocompatibility Complex , Mice , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand
7.
Prog Biophys Mol Biol ; 153: 23-27, 2020 07.
Article En | MEDLINE | ID: mdl-32006526

T-cell based immunity is mediated through specific T cell receptor (TCR) recognition of a small antigenic peptide in complex with a host immune molecule, major histocompatibility complex (pMHC). The interaction of a TCR and its pMHC ligand is generally quite weak, degenerate and biophysically unfavorable. Yet, the resulting immune response is extremely effective, being both sensitive and specific. Recent observations indicate that the TCR is an anisotropic mechanosensor. The force sensed by TCR's recognition module is transmitted to the non-covalently associated signal transduction module. Multiple biophysical methods reveal that the molecular mechanism for TCR-pMHC interaction under force required to induce T cell signaling is linked to "catch bond" formation between a TCR and its cognate ligand pMHC. This kind of dynamic non-covalent bond actually increases the bond lifetime by deforming the molecule to make the interaction lock tighter. The key observation is that the more stimulatory the antigenic peptide, the more pronounced the catch bond and immune response. By contrast, an unrelated, non-antigenic peptide presented by the same MHC molecule does not form a catch bond, instead manifesting a slip bond associated with rapid TCR-pMHC dissociation. In summary, a weak interaction between a TCR and agonist ligand will be dramatically amplified by a catch bond under physical load generated by cell movement during immune surveillance. These new biophysical concepts, TCR mechanosensor and dynamic catch bond formation, begin to reveal how bioforces tune T cell signaling and should be potentially enlightening for immunotherapy design against cancers.


Immunotherapy , Mechanical Phenomena , Receptors, Antigen, T-Cell/metabolism , Humans , Major Histocompatibility Complex , Signal Transduction
8.
Trends Biochem Sci ; 45(1): 6-12, 2020 01.
Article En | MEDLINE | ID: mdl-31704057

Netrin is a prototypical axon guidance cue. Structural studies have revealed how netrin interacts with the deleted in colorectal cancer (DCC) receptor, other receptors, and co-factors for signaling. Recently, genetic studies suggested that netrin is involved in neuronal haptotaxis, which requires a reversible adhesion process. Structural data indicate that netrin can also mediate trans-adhesion between apposing cells decorated with its receptors on the condition that the auxiliary guidance cue draxin is present. Here, we propose that netrin is involved in conditional adhesion, a reversible and localized process that can contribute to cell adhesion and migration. We suggest that netrin-mediated adhesion and signaling are linked, and that local environmental factors in the ventricular zone, the floor plate, or other tissues coordinate its function.


DCC Receptor/metabolism , Netrins/metabolism , Signal Transduction , Animals , Cell Adhesion , DCC Receptor/chemistry , Humans , Netrins/chemistry , Netrins/genetics
9.
Proc Natl Acad Sci U S A ; 116(45): 22556-22566, 2019 11 05.
Article En | MEDLINE | ID: mdl-31624123

The membrane proximal external region (MPER) of HIV-1 envelope glycoprotein (gp) 41 is an attractive vaccine target for elicitation of broadly neutralizing antibodies (bNAbs) by vaccination. However, current details regarding the quaternary structural organization of the MPER within the native prefusion trimer [(gp120/41)3] are elusive and even contradictory, hindering rational MPER immunogen design. To better understand the structural topology of the MPER on the lipid bilayer, the adjacent transmembrane domain (TMD) was appended (MPER-TMD) and studied. Membrane insertion of the MPER-TMD was sensitive both to the TMD sequence and cytoplasmic residues. Antigen binding of MPER-specific bNAbs, in particular 10E8 and DH511.2_K3, was significantly impacted by the presence of the TMD. Furthermore, MPER-TMD assembly into 10-nm diameter nanodiscs revealed a heterogeneous membrane array comprised largely of monomers and dimers, as enumerated by bNAb Fab binding using single-particle electron microscopy analysis, arguing against preferential trimeric association of native MPER and TMD protein segments. Moreover, introduction of isoleucine mutations in the C-terminal heptad repeat to induce an extended MPER α-helical bundle structure yielded an antigenicity profile of cell surface-arrayed Env variants inconsistent with that found in the native prefusion state. In line with these observations, electron paramagnetic resonance analysis suggested that 10E8 inhibits viral membrane fusion by lifting the MPER N-terminal region out of the viral membrane, mandating the exposure of residues that would be occluded by MPER trimerization. Collectively, our data suggest that the MPER is not a stable trimer, but rather a dynamic segment adapted for structural changes accompanying fusion.


Cell Membrane/virology , HIV Envelope Protein gp41/chemistry , HIV-1/immunology , Antibodies, Neutralizing/immunology , Cell Membrane/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/immunology , HIV Infections/immunology , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/immunology , Protein Domains
10.
J Biomol NMR ; 73(6-7): 319-332, 2019 Jul.
Article En | MEDLINE | ID: mdl-30815789

Early studies of T cell structural biology using X-ray crystallography, surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC) focused on a picture of the αßT cell receptor (αßTCR) component domains and their cognate ligands (peptides bound to MHC molecules, i.e. pMHCs) as static interaction partners. Moving forward requires integrating this corpus of data with dynamic technologies such as NMR, molecular dynamics (MD) simulations and real-time single molecule (SM) studies exemplified by optical tweezers (OT). NMR bridges relevant timescales and provides the potential for an all-atom dynamic description of αßTCR components prior to and during interactions with binding partners. SM techniques have opened up vistas in understanding the non-equilibrium nature of T cell signaling through the introduction of force-mediated binding measurements into the paradigm for T cell function. In this regard, bioforces consequent to T-lineage cell motility are now perceived as placing piconewton (pN)-level loads on single receptor-pMHC bonds to impact structural change and αßT-lineage biology, including peptide discrimination, cellular activation, and developmental progression. We discuss herein essential NMR technologies in illuminating the role of ligand binding in the preT cell receptor (preTCR), the αßTCR developmental precursor, and convergence of NMR, SM and MD data in advancing our comprehension of T cell development. More broadly we review the central hypothesis that the αßTCR is a mechanosensor, fostered by breakthrough NMR-based structural insights. Collectively, elucidating dynamic aspects through the integrative use of NMR, SM, and MD shall advance fundamental appreciation of the mechanism of T cell signaling as well as inform translational efforts in αßTCR and chimeric T cell (CAR-T) immunotherapies and T cell vaccinology.


Histocompatibility Antigens/chemistry , Nuclear Magnetic Resonance, Biomolecular , Receptors, Antigen, T-Cell/chemistry , Histocompatibility Antigens/metabolism , Humans , Ligands , Mechanotransduction, Cellular , Models, Molecular , Protein Conformation , Receptors, Antigen, T-Cell/metabolism , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Neuron ; 97(6): 1261-1267.e4, 2018 03 21.
Article En | MEDLINE | ID: mdl-29503192

Axon guidance involves the spatiotemporal interplay between guidance cues and membrane-bound cell-surface receptors, present on the growth cone of the axon. Netrin-1 is a prototypical guidance cue that binds to deleted in colorectal cancer (DCC), and it has been proposed that the guidance cue Draxin modulates this interaction. Here, we present structural snapshots of Draxin/DCC and Draxin/Netrin-1 complexes, revealing a triangular relationship that affects Netrin-mediated haptotaxis and fasciculation. Draxin interacts with DCC through the N-terminal four immunoglobulin domains, and Netrin-1 through the EGF-3 domain, in the same region where DCC binds. Netrin-1 and DCC bind to adjacent sites on Draxin, which appears to capture Netrin-1 and tether it to the DCC receptor. We propose the conformational flexibility of the single-pass membrane receptor DCC is used to promote fasciculation and regulate axon guidance through concerted Netrin-1/Draxin binding. VIDEO ABSTRACT.


Axon Guidance/physiology , DCC Receptor/metabolism , Nerve Tissue Proteins/metabolism , Netrin-1/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , DCC Receptor/chemistry , DCC Receptor/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Netrin-1/chemistry , Netrin-1/genetics , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary
12.
Cell Discov ; 4: 8, 2018.
Article En | MEDLINE | ID: mdl-29479476

Netrin-1 plays a key role in axon guidance through binding to its receptor, Deleted in Colorectal Cancer (DCC). The initial step of signaling inside the cell after netrin-1/DCC ligation is the binding of DCC cytoplasmic P3 motif to focal adhesion targeting (FAT) domain of focal adhesion kinase (FAK). Here we report the crystal structure of P3/FAT complex. The helical P3 peptide interacts with a helix-swapped FAT dimer in a 2:2 ratio. Dimeric FAT binding is P3-specific and stabilized by a calcium ion. Biochemical studies showed that DCC-P3 motif and calcium ion could facilitate FAT dimerization in solution. Axon guidance assays confirm that the DCC/FAK complex is essential for netrin-1-induced chemoattraction. We propose that netrin-1/DCC engagement creates a small cluster of P3/FAT for FAK recruitment close to the cell membrane, which exerts a concerted effect with PIP2 for FAK signaling. We also compare P3/FAT binding with paxillin/FAT binding and discuss their distinct recognition specificity on a common FAT domain for axon attraction versus integrin signaling, respectively.

15.
Proc Natl Acad Sci U S A ; 114(13): 3403-3408, 2017 03 28.
Article En | MEDLINE | ID: mdl-28292891

Recognition by the leukocyte integrins αXß2 and αMß2 of complement iC3b-opsonized targets is essential for effector functions including phagocytosis. The integrin-binding sites on iC3b remain incompletely characterized. Here, we describe negative-stain electron microscopy and biochemical studies of αXß2 and αMß2 in complex with iC3b. Despite high homology, the two integrins bind iC3b at multiple distinct sites. αXß2 uses the αX αI domain to bind iC3b on its C3c moiety at one of two sites: a major site at the interface between macroglobulin (MG) 3 and MG4 domains, and a less frequently used site near the C345C domain. In contrast, αMß2 uses its αI domain to bind iC3b at the thioester domain and simultaneously interacts through a region near the αM ß-propeller and ß2 ßI domain with a region of the C3c moiety near the C345C domain. Remarkably, there is no overlap between the primary binding site of αXß2 and the binding site of αMß2 on iC3b. Distinctive binding sites on iC3b by integrins αXß2 and αMß2 may be biologically beneficial for leukocytes to more efficiently capture opsonized pathogens and to avoid subversion by pathogen factors.


Complement C3b/metabolism , Integrin alphaXbeta2/metabolism , Macrophage-1 Antigen/metabolism , Binding Sites , Complement C3b/chemistry , Complement C3b/genetics , Humans , Integrin alphaXbeta2/chemistry , Integrin alphaXbeta2/genetics , Leukocytes/chemistry , Leukocytes/metabolism , Macrophage-1 Antigen/chemistry , Macrophage-1 Antigen/genetics , Protein Binding , Protein Domains , Protein Structure, Tertiary
18.
Sci Adv ; 2(5): e1501118, 2016 05.
Article En | MEDLINE | ID: mdl-27386517

The Drosophila neural receptor Dscam1 (Down syndrome cell adhesion molecule 1) plays an essential role in neuronal wiring and self-avoidance. Dscam1 potentially encodes 19,008 ectodomains through alternative RNA splicing and exhibits exquisite isoform-specific homophilic binding, which makes it an exceptional example for studying protein binding specificity. However, structural information on Dscam1 is limited, which hinders illumination of the mechanism of Dscam1 isoform-specific recognition. Whether different Dscam1 isoforms adopt the same dimerization mode remains a subject of debate. We present 12 Dscam1 crystal structures, provide direct evidence indicating that all isoforms adopt a conserved homodimer geometry in a modular fashion, identify two mechanisms for the Ig2 binding domain to dispel electrostatic repulsion during dimerization, decode Ig2 binding specificity by a central motif at its symmetry center, uncover the role of glycosylation in Dscam1 homodimerization, and find electrostatic potential complementarity to help define the binding region and the antiparallel binding mode. We then propose a concept that the context of a protein may set restrictions to regulate its binding specificity, which provides a better understanding of protein recognition.


Drosophila Proteins/chemistry , Neural Cell Adhesion Molecules/chemistry , Protein Multimerization , Alternative Splicing , Amino Acid Sequence , Cell Adhesion Molecules , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Glycosylation , Models, Molecular , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Protein Binding , Protein Conformation , Protein Isoforms , Static Electricity
19.
Front Immunol ; 6: 441, 2015.
Article En | MEDLINE | ID: mdl-26388869

The αßTCR was recently revealed to function as a mechanoreceptor. That is, it leverages mechanical energy generated during immune surveillance and at the immunological synapse to drive biochemical signaling following ligation by a specific foreign peptide-MHC complex (pMHC). Here, we review the structural features that optimize this transmembrane (TM) receptor for mechanotransduction. Specialized adaptations include (1) the CßFG loop region positioned between Vß and Cß domains that allosterically gates both dynamic T cell receptor (TCR)-pMHC bond formation and lifetime; (2) the rigid super ß-sheet amalgams of heterodimeric CD3εγ and CD3εδ ectodomain components of the αßTCR complex; (3) the αßTCR subunit connecting peptides linking the extracellular and TM segments, particularly the oxidized CxxC motif in each CD3 heterodimeric subunit that facilitates force transfer through the TM segments and surrounding lipid, impacting cytoplasmic tail conformation; and (4) quaternary changes in the αßTCR complex that accompany pMHC ligation under load. How bioforces foster specific αßTCR-based pMHC discrimination and why dynamic bond formation is a primary basis for kinetic proofreading are discussed. We suggest that the details of the molecular rearrangements of individual αßTCR subunit components can be analyzed utilizing a combination of structural biology, single-molecule FRET, optical tweezers, and nanobiology, guided by insightful atomistic molecular dynamic studies. Finally, we review very recent data showing that the pre-TCR complex employs a similar mechanobiology to that of the αßTCR to interact with self-pMHC ligands, impacting early thymic repertoire selection prior to the CD4(+)CD8(+) double positive thymocyte stage of development.

...