Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nature ; 592(7855): 639-643, 2021 04.
Article in English | MEDLINE | ID: mdl-33790470

ABSTRACT

Some Plasmodium falciparum repetitive interspersed families of polypeptides (RIFINs)-variant surface antigens that are expressed on infected erythrocytes1-bind to the inhibitory receptor LAIR1, and insertion of DNA that encodes LAIR1 into immunoglobulin genes generates RIFIN-specific antibodies2,3. Here we address the general relevance of this finding by searching for antibodies that incorporate LILRB1, another inhibitory receptor that binds to ß2 microglobulin and RIFINs through their apical domains4,5. By screening plasma from a cohort of donors from Mali, we identified individuals with LILRB1-containing antibodies. B cell clones isolated from three donors showed large DNA insertions in the switch region that encodes non-apical LILRB1 extracellular domain 3 and 4 (D3D4) or D3 alone in the variable-constant (VH-CH1) elbow. Through mass spectrometry and binding assays, we identified a large set of RIFINs that bind to LILRB1 D3. Crystal and cryo-electron microscopy structures of a RIFIN in complex with either LILRB1 D3D4 or a D3D4-containing antibody Fab revealed a mode of RIFIN-LILRB1 D3 interaction that is similar to that of RIFIN-LAIR1. The Fab showed an unconventional triangular architecture with the inserted LILRB1 domains opening up the VH-CH1 elbow without affecting VH-VL or CH1-CL pairing. Collectively, these findings show that RIFINs bind to LILRB1 through D3 and illustrate, with a naturally selected example, the general principle of creating novel antibodies by inserting receptor domains into the VH-CH1 elbow.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Cryoelectron Microscopy , Leukocyte Immunoglobulin-like Receptor B1/chemistry , Plasmodium falciparum/chemistry , Plasmodium falciparum/immunology , Adolescent , Adult , Amino Acid Sequence , Antibodies/ultrastructure , Antibody Specificity , Antigens, Protozoan/ultrastructure , Binding Sites, Antibody , Child , Child, Preschool , Cohort Studies , Humans , Infant , Leukocyte Immunoglobulin-like Receptor B1/immunology , Mali , Models, Molecular , Plasmodium falciparum/genetics , Plasmodium falciparum/ultrastructure , Protein Domains , Young Adult
2.
Nature ; 587(7833): 309-312, 2020 11.
Article in English | MEDLINE | ID: mdl-32650338

ABSTRACT

The Plasmodium species that cause malaria are obligate intracellular parasites, and disease symptoms occur when these parasites replicate in human blood. Despite the risk of immune detection, the parasite delivers proteins that bind to host receptors on the cell surfaces of infected erythrocytes. In the causative parasite of the most deadly form of malaria in humans, Plasmodium falciparum, RIFINs form the largest family of surface proteins displayed by erythrocytes1. Some RIFINs can bind to inhibitory immune receptors, and these RIFINs act as targets for unusual antibodies that contain a LAIR1 ectodomain2-4 or as ligands for LILRB15. RIFINs stimulate the activation of and signalling by LILRB15, which could potentially lead to the dampening of human immune responses. Here, to understand how RIFINs activate LILRB1-mediated signalling, we determine the structure of a RIFIN bound to LILRB1. We show that this RIFIN mimics the natural activating ligand of LILRB1, MHC class I, in its LILRB1-binding mode. A single mutation in the RIFIN disrupts the complex, blocks LILRB1 binding of all tested RIFINs and abolishes signalling in a reporter assay. In a supported lipid bilayer system, which mimics the activation of natural killer (NK) cells by antibody-dependent cell-mediated cytotoxicity, both RIFIN and MHC are recruited to the immunological synapse of NK cells and reduce the activation of NK cells, as measured by the mobilization of perforin. Therefore, LILRB1-binding RIFINs mimic the binding mode of the natural ligand of LILRB1 and suppress the function of NK cells.


Subject(s)
Leukocyte Immunoglobulin-like Receptor B1/chemistry , Leukocyte Immunoglobulin-like Receptor B1/immunology , Malaria, Falciparum/immunology , Membrane Proteins/chemistry , Membrane Proteins/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Binding Sites/immunology , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocyte Immunoglobulin-like Receptor B1/metabolism , Ligands , Lipid Bilayers , Lymphocyte Activation , Malaria, Falciparum/parasitology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Molecular Mimicry/immunology , Mutation , Perforin/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Signal Transduction
3.
Cell Mol Immunol ; 17(9): 966-975, 2020 09.
Article in English | MEDLINE | ID: mdl-31273318

ABSTRACT

Leukocyte immunoglobulin (Ig)-like receptors (LILRs), also known as CD85 and immunoglobulin-like transcripts (ILTs), play pivotal roles in regulating immune responses. These receptors define an immune checkpoint that immune therapy can target. Through cis or trans interactions with human leukocyte antigen (HLA)-G, the two most abundantly expressed inhibitory LILRs, LILRB1, and LILRB2 (LILRB1/2, also known as CD85j/d and ILT2/4), are involved in immunotolerance in pregnancy and transplantation, autoimmune diseases, and immune evasion by tumors. Although the discrete domains of LILRB1/2 are clear, the assembly mode of the four extracellular Ig-like domains (D1, D2, D3, and D4) remains unknown. Previous data indicate that D1D2 is responsible for binding to HLA class I (HLA-I), but the roles of D3D4 are still unclear. Here, we determined the crystal structure of the four Ig-like domain LILRB2 and four-domain LILRB1 in complex with HLA-G1. The angles between adjacent domains and the staggered assembly of the four domains suggest limited flexibility and limited plasticity of the receptors during ligand binding. The complex structure of four-domain LILRB1 and HLA-G1 supports the model that D1D2 is responsible for HLA-I binding, while D3D4 acts as a scaffold. Accordingly, cis and trans binding models for HLA-I binding to LILRB1/2 are proposed. The geometries of LILRB1/2 in complex with dimeric and monomeric HLA-G1 suggest the accessibility of the dimeric receptor, which in turn, transduces more inhibitory signals. The assembly of LILRB1/2 and its binding to HLA-G1 could aid in the design of immune regulators and benefit immune interference.


Subject(s)
HLA-G Antigens/chemistry , HLA-G Antigens/metabolism , Immunoglobulins/chemistry , Leukocyte Immunoglobulin-like Receptor B1/chemistry , Leukocyte Immunoglobulin-like Receptor B1/metabolism , Alleles , Ligands , Models, Molecular , Protein Binding , Protein Domains , Structural Homology, Protein
4.
J Immunol Methods ; 464: 47-56, 2019 01.
Article in English | MEDLINE | ID: mdl-30365927

ABSTRACT

X-ray crystallographic studies of class I peptide-MHC molecules (pMHC) continue to provide important insights into immune recognition, however their success depends on generation of diffraction-quality crystals, which remains a significant challenge. While protein engineering techniques such as surface-entropy reduction and lysine methylation have proven utility in facilitating and/or improving protein crystallisation, they risk affecting the conformation and biochemistry of the class I MHC antigen binding groove. An attractive alternative is the use of noncovalent crystallisation chaperones, however these have not been developed for pMHC. Here we describe a method for promoting class I pMHC crystallisation, by exploiting its natural ligand interaction with the immunoregulatory receptor LILRB1 as a novel crystallisation chaperone. First, focussing on a model HIV-1-derived HLA-A2-restricted peptide, we determined a 2.4 ŠHLA-A2/LILRB1 structure, which validated that co-crystallisation with LILRB1 does not alter conformation of the antigenic peptide. We then demonstrated that addition of LILRB1 enhanced the crystallisation of multiple peptide-HLA-A2 complexes, and identified a generic condition for initial co-crystallisation. LILRB1 chaperone-based crystallisation enabled structure determination for HLA-A2 complexes previously intransigent to crystallisation, including both conventional and post-translationally-modified peptides, of diverse lengths. Since both the LILRB1 recognition interface on the HLA-A2 α3 domain molecule and HLA-A2-mediated crystal contacts are predominantly conserved across class I MHC molecules, the approach we outline could prove applicable to a diverse range of class I pMHC. LILRB1 chaperone-mediated crystallisation should expedite molecular insights into the immunobiology of diverse immune-related diseases and immunotherapeutic strategies, particularly involving class I pMHC complexes that are challenging to crystallise.


Subject(s)
Antigens, CD/chemistry , Crystallography, X-Ray/methods , HLA-A2 Antigen/chemistry , Leukocyte Immunoglobulin-like Receptor B1/chemistry , Molecular Chaperones/chemistry , Antigens, CD/immunology , Binding Sites , Crystallization , HIV-1/immunology , HLA-A2 Antigen/immunology , Humans , Immunodominant Epitopes , Leukocyte Immunoglobulin-like Receptor B1/immunology , Ligands , Models, Molecular , Molecular Chaperones/immunology , Phosphorylation , Protein Binding , Protein Conformation , Structure-Activity Relationship
5.
Nature ; 552(7683): 101-105, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29186116

ABSTRACT

Malaria is among the most serious infectious diseases affecting humans, accounting for approximately half a million deaths each year. Plasmodium falciparum causes most life-threatening cases of malaria. Acquired immunity to malaria is inefficient, even after repeated exposure to P. falciparum, but the immune regulatory mechanisms used by P. falciparum remain largely unknown. Here we show that P. falciparum uses immune inhibitory receptors to achieve immune evasion. RIFIN proteins are products of a polymorphic multigene family comprising approximately 150-200 genes per parasite genome that are expressed on the surface of infected erythrocytes. We found that a subset of RIFINs binds to either leucocyte immunoglobulin-like receptor B1 (LILRB1) or leucocyte-associated immunoglobulin-like receptor 1 (LAIR1). LILRB1-binding RIFINs inhibit activation of LILRB1-expressing B cells and natural killer (NK) cells. Furthermore, P. falciparum-infected erythrocytes isolated from patients with severe malaria were more likely to interact with LILRB1 than erythrocytes from patients with non-severe malaria, although an extended study with larger sample sizes is required to confirm this finding. Our results suggest that P. falciparum has acquired multiple RIFINs to evade the host immune system by targeting immune inhibitory receptors.


Subject(s)
Immune Evasion/immunology , Leukocyte Immunoglobulin-like Receptor B1/immunology , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Receptors, Immunologic/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CHO Cells , Cricetulus , Erythrocytes/immunology , Erythrocytes/parasitology , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocyte Immunoglobulin-like Receptor B1/chemistry , Ligands , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Immunologic/chemistry , Sample Size
SELECTION OF CITATIONS
SEARCH DETAIL
...