LILRB2 (Leukocyte Immunoglobulin-Like Receptor Subfamily B Member 2) is a transmembrane protein encoded by the LILRB2 gene in humans. It belongs to the leukocyte immunoglobulin-like receptor (LIR) family, located on chromosome 19q13.4, and functions as an inhibitory immune checkpoint receptor . LILRB2 is expressed on myeloid cells, including monocytes, macrophages, and dendritic cells, where it regulates immune responses by binding to MHC class I molecules and other ligands .
LILRB2 consists of:
Extracellular region: Four immunoglobulin (Ig)-like domains (D1–D4) responsible for ligand binding .
Transmembrane domain: Anchors the receptor to the cell membrane .
Cytoplasmic tail: Contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit phosphatases like SHP-1/2 to suppress signaling .
Crystal structures (e.g., PDB 2DYP, 6AED) reveal that LILRB2 binds HLA-G via hydrophobic interactions in the α3 domain, enabling competitive inhibition of CD8+ T cell activation .
MHC I Interaction: Competes with CD8α for binding to classical (HLA-A, -B, -C) and non-classical (HLA-G, HLA-F) MHC I molecules, suppressing T cell activation .
β-Amyloid Receptor: Binds oligomeric Aβ42 in Alzheimer’s disease, triggering synaptic loss via cofilin-mediated actin depolymerization .
Myeloid Cell Modulation: Inhibits pro-inflammatory responses in macrophages and dendritic cells by recruiting SHP-1/2, reducing ERK/p38 MAPK and NF-κB signaling .
Antibody Antagonists: Anti-LILRB2 monoclonal antibodies (e.g., MK-4830, JTX-8064) skew immunosuppressive MDSCs/M2 macrophages toward pro-inflammatory M1 phenotypes, enhancing antitumor immunity .
Combination Therapy: Co-administration with SHP-1/2 inhibitors synergistically reduces intracellular M. tuberculosis burden by 1.8 log<sub>10</sub> CFU .
Model | Outcome |
---|---|
SK-MEL-5 (Melanoma) | Anti-LILRB2 antibodies induced tumor regression in humanized mice . |
APP/PS1 (Alzheimer’s) | Aβ-LILRB2 binding reduced PSD-95 levels, impairing synaptic plasticity . |
Commercial recombinant LILRB2 proteins are used for in vitro studies:
Leukocyte Immunoglobulin Like Receptor B2 (LILRB2), a member of the leukocyte immunoglobulin-like receptor (LIR) family, is found on immune cells. It binds to MHC class I molecules on antigen-presenting cells and transmits a negative signal, thereby suppressing immune response stimulation. This receptor plays a crucial role in regulating inflammatory responses and cytotoxicity, ensuring a focused immune response and limiting autoreactivity. LILRB2 competes with CD8 alpha for MHC I binding and interacts with both classical MHC I proteins and the non-classical HLA-G1 and HLA-F molecules.
Recombinant human LILRB2, produced in HEK293 cells, is a single, glycosylated polypeptide chain. It comprises 444 amino acids (24-461 a.a), has a molecular weight of 48.3kDa, and includes a 6 amino acid His tag at the C-terminus. Purification is achieved through proprietary chromatographic techniques.
A clear, colorless solution that has been sterilized by filtration.
The LILRB2 solution is provided at a concentration of 0.5mg/ml in phosphate buffered saline (pH 7.4) with 20% glycerol.
For short-term storage (up to 2-4 weeks), keep at 4°C. For longer periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity is confirmed to be greater than 95.0% using SDS-PAGE analysis.
Leukocyte Immunoglobulin-like Receptor, CD85D, CD85d, CD85 antigen-like family member D, CD85d antigen, ILT-4, ILT4, ILT4CD85d, Ig-like transcript 4, Immunoglobulin-like transcript 4, LILRB2, LIR2, LIR-2, LIR2CD85D, Leukocyte immunoglobulin-like receptor 2, MIR10, MIR-10, MIR10LILRA6, Monocyte/macrophage immunoglobulinlike receptor 10, leukocyte immunoglobulin-like receptor subfamily B member 2, leukocyte immunoglobulin-like receptor subfamily B member 2 soluble isoform 1.
HEK293 Cells.
GTIPKPTLWA EPDSVITQGS PVTLSCQGSL EAQEYRLYRE KKSASWITRI RPELVKNGQF HIPSITWEHT GRYGCQYYSR ARWSELSDPL VLVMTGAYPK PTLSAQPSPV VTSGGRVTLQ CESQVAFGGF ILCKEGEDEH PQCLNSQPHA RGSSRAIFSV GPVSPNRRWS HRCYGYDLNS PYVWSSPSDL LELLVPGVSK KPSLSVQPGP VMAPGESLTL QCVSDVGYDR FVLYKEGERD LRQLPGRQPQ AGLSQANFTL GPVSRSYGGQ YRCYGAHNLS SECSAPSDPL DILITGQIRG TPFISVQPGP TVASGENVTL LCQSWRQFHT FLLTKAGAAD APLRLRSIHE YPKYQAEFPM SPVTSAHAGT YRCYGSLNSD PYLLSHPSEP LELVVSGPSM GSSPPPTGPI STPAGPEDQP LTPTGSDPQS GLGRHLGVHH HHHH
LILRB2 contains four immunoglobulin (Ig) domains (D1-D4), a transmembrane domain, and 3-4 immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in its cytoplasmic tail . These ITIMs interact with tyrosine phosphatases, inhibiting activating signals in immune cells . Unlike LILRB1, which is widely expressed in monocytes, dendritic cells, B cells, and subsets of NK and T cells, LILRB2 expression is restricted primarily to the myelomonocytic lineage . LILRB2 binds to HLA molecules through its two N-terminal extracellular domains (D1 and D2), with D3 and D4 appearing to function as scaffolds for the D1-D2 binding . A distinctive feature of LILRB2 compared to LILRB1 is that it does not require the presence of β2m when binding to HLA molecules .
For comprehensive characterization of LILRB2 genetic diversity, next-generation sequencing (NGS) combined with specialized bioinformatics approaches is most effective . The bioinformatics strategy using tools like hla-mapper software is crucial for achieving reliable read mapping of LILRB2, particularly due to its high sequence similarity with other LILR family members . This approach is especially important since LILRB2 arose from intergenic recombination and gene duplication, with identity levels among the LILR D1-D4 domains ranging from 63% to 84% when aligned to LILRB1 . Full exon sequencing is necessary to capture all potential variants, as demonstrated in studies that identified numerous SNVs arranged into multiple haplotypes .
Researchers can differentiate between LILRB2 isoforms using a combination of techniques. Alternative mRNA processing gives rise to eight different protein isoforms from LILRB2, each with distinct cell expression profiles . Methodologically, this requires:
RT-PCR with isoform-specific primers to distinguish between alternative splice variants
Western blotting with antibodies targeting unique epitopes in different isoforms
Flow cytometry to detect surface expression patterns on different cell types
Mass spectrometry for precise protein identification and characterization
Reporter systems with isoform-specific constructs to monitor functional differences
These approaches can help distinguish between the various LILRB2 isoforms that may have distinct biological functions in different cellular contexts.
Both positive and purifying selection patterns have been detected in LILRB2 coding regions . Methodologically, several approaches have proven effective:
FUBAR (Fast Unconstrained Bayesian AppRoximation) analysis has identified residues under positive selection, suggesting amino acid replacements resulting in beneficial functional changes .
FoldX4 stability analysis has classified specific SNPs (like rs1128646 and rs7247538) as stabilizing mutations, while others (such as rs386056*G) cause slight destabilizing effects .
Population genetics approaches examining haplotype structure and frequency in diverse populations reveal evidence of selection .
Comparative genomics showing that LILRB2, like other LIR family members, is rapidly evolving with larger interspecies differences compared to the genome average .
These findings collectively provide strong evidence supporting the directional selection regime hypothesis for LILRB2 evolution.
Linkage disequilibrium (LD) patterns in LILRB2 are highly population-specific, being affected by factors such as non-random mating, selection, mutation, migration, admixture, genetic drift, and effective population size . Research in admixed populations like Brazilians has revealed that:
Variants differentiating specific LILRB2 haplogroups (a, c, d) are part of the same LD block and are in high or complete LD with each other (r² > 0.5) .
These population-specific LD patterns may not be directly extrapolated to worldwide populations without comprehensive evaluation .
The implications for research include:
Need for population-specific genetic association studies
Potential for uncovering novel variants in admixed populations
Importance of considering ancestral background when interpreting LILRB2 genetic data
Necessity for tailored approaches to identify disease associations in different populations
LILRB2 polymorphisms can significantly affect protein stability and function, with both structural and evolutionary implications . Research has shown:
SNP ID | Position | Effect on Protein | Selection Pattern |
---|---|---|---|
rs1128646 | Haplogroup a | Stabilizing mutation | Positive selection |
rs7247538 | Haplogroup a | Stabilizing mutation | Positive selection |
rs7247451 | Haplogroup a | Associated with function | Positive selection |
rs386056*G | D2 domain | Slight destabilizing effect | Not specified |
rs383369*G | Not specified | Associated with expression levels | Not specified |
Some polymorphisms directly affect the interaction with ligands. For example, non-conserved residues in the binding region on D1D2 (such as Q76, R80, and R84 in LILRB2 compared to Y76, D80, and R84 in LILRB1) contribute to the higher affinity of LILRB2 for HLA-G compared to LILRB1 . Additionally, residues T36 and A38 in LILRB2 recognize the 195–197 loop of HLA-G, demonstrating how specific polymorphisms can affect ligand recognition .
The molecular mechanism of Angptl2 binding to LILRB2 involves:
Angptl2 expressed in mammalian cells forms high-molecular-weight (HMW) species, which is required for activation of LILRB2 .
A specific motif in the first and fourth Ig domains of LILRB2 is critical for Angptl2 binding and signaling activation .
While individual Ig domains don't bind Angptl2, Ig1 and Ig2 in combination display about 50% of the maximal binding, while Ig3 and Ig4 in combination display about 10%, indicating the major binding site resides in Ig1 and Ig2 .
Researchers can experimentally validate this interaction using:
Chimeric receptor reporter systems that fuse the LILRB2 extracellular domain with transmembrane/intracellular domains containing immunoreceptor tyrosine-based activation motifs linked to reporter genes .
Flow cytometry-based cell-surface ligand binding assays with domain- and site-specific mutations to map critical binding regions .
Confocal microscopy to visualize receptor clustering upon ligand binding, as evidenced by the change from ring-like to spot-like distribution of LILRB2 on cell membranes .
Ligand multimerization is crucial for LILRB2 activation and subsequent signaling . Key findings include:
Angptl2 expressed in mammalian cells exists as high-molecular-weight species necessary for LILRB2 activation .
Immobilized anti-LILRB2 antibodies induce more potent activation than soluble antibodies or Angptl2, suggesting that ligand presentation in a multimerized form is critical .
Receptor clustering appears to be the mechanism of activation, as demonstrated by the change in LILRB2-ECD distribution from a ring-like to a spot-like pattern after treatment with immobilized antibodies .
Methodological approaches to assess this include:
Gel filtration chromatography to characterize the molecular weight of ligands
Chemical cross-linking studies to analyze ligand multimerization
Surface plasmon resonance to measure binding kinetics of monomeric versus multimeric ligands
Confocal microscopy with fluorescently labeled receptor to visualize clustering
FRET (Förster Resonance Energy Transfer) analysis to measure receptor proximity upon activation
LILRB2 signaling differs significantly between Angptl2 and HLA-G binding:
Angptl2 binding to LILRB2 is more potent than and not completely overlapped with HLA-G binding .
In reporter cell assays, Angptl2-treated LILRB2 reporter cells induced significantly greater GFP expression (18.95% ± 0.95%) than control cells (5.34% ± 1.19%), while even high concentrations of HLA-G (130 μg/mL) failed to activate GFP expression .
Angptl2 binding to LILRB2 induces activation of SHP-2 and Ca²⁺/calmodulin-dependent kinase, which are critical for supporting HSC activity .
Techniques to measure these differences include:
Chimeric receptor reporter systems with quantifiable outputs (e.g., GFP expression)
Phosphorylation assays for downstream signaling molecules (SHP-1, SHP-2)
Calcium flux assays to measure intracellular calcium mobilization
Competitive binding assays to determine relative affinities
Functional assays specific to cell types of interest (e.g., HSC expansion assays)
LILRB2 plays crucial roles in hematopoietic stem cell (HSC) maintenance and expansion through several mechanisms:
LILRB2 is expressed on human HSCs and its activation supports ex vivo expansion of these cells .
Binding of Angptls to LILRB2 induces activation of SHP-2 and Ca²⁺/calmodulin-dependent kinase, types of factors known to be critical for supporting HSC activity .
LILRB2 signaling inhibits differentiation and promotes self-renewal, helping to maintain the stem cell state .
The interaction with Angptls leads to specific downstream signaling events that support HSC expansion while preserving stemness properties .
These findings have led to the development of specialized culture systems containing defined cytokines and immobilized anti-LILRB2 antibodies that support stable and reproducible ex vivo expansion of repopulating human cord blood HSCs .
Optimization of serum-free culture conditions for LILRB2-mediated HSC expansion requires attention to several key factors:
Ligand presentation: Immobilized anti-LILRB2 antibodies induce more potent activation than soluble antibodies or Angptl2 . The immobilization approach is critical as it supports receptor clustering for optimal signaling activation .
Cytokine combination: A defined mixture of cytokines must be combined with LILRB2 activation to support HSC expansion while maintaining stemness .
Culture duration: Determining the optimal culture period to balance expansion with preservation of repopulating potential.
Antibody concentration: Titration of immobilized anti-LILRB2 concentration to achieve optimal receptor activation without inducing negative effects.
Substrate selection: The surface used for antibody immobilization can affect both antibody orientation and HSC interaction.
Medium formulation: Beyond cytokines, other medium components must be optimized to support HSC metabolism and prevent differentiation.
Research has revealed both similarities and contradictions regarding LILRB2 function in normal versus leukemic stem cells:
LILRB2 supports self-renewal in both normal HSCs and leukemic stem cells .
Both cell types utilize LILRB2 signaling for maintenance and expansion .
Similar downstream signaling pathways (SHP-2 and Ca²⁺/calmodulin-dependent kinase) are activated in both contexts .
While LILRB2 activity is regulated in normal HSCs, it appears to be dysregulated in leukemic cells .
The balance between self-renewal and differentiation mediated by LILRB2 differs between normal and leukemic contexts.
The requirement for LILRB2 in leukemia development suggests potential therapeutic targeting approaches that must spare normal HSCs.
These contradictions present both challenges and opportunities for developing targeted therapies that disrupt LILRB2 function in leukemic cells while preserving normal HSC activity.
Effective bioinformatic pipelines for analyzing LILRB2 sequence data must address several challenges, particularly for diverse or admixed populations:
Specialized mapping tools: Software like hla-mapper is essential for reliable read mapping of LILRB2 due to its high sequence similarity with other LILR family members (63-84% identity) .
Pipeline components:
Quality control and read trimming
Specialized alignment algorithms for genes with homologs
Variant calling optimized for polymorphic regions
Haplotype phasing
Selection analysis (FUBAR, FoldX4)
Population structure analysis
Linkage disequilibrium assessment
Population-specific considerations:
Validation strategies:
Cross-platform validation of identified variants
Functional prediction of missense variants
Conservation analysis across species
Domain-swapping experiments are valuable for studying LILRB2 binding specificity. Based on research findings, an effective experimental design should:
Target key domains: Focus on D1 and D2 domains as they display about 50% of maximal binding to Angptl2, while D3 and D4 in combination display only about 10% .
Create chimeric constructs:
LILRB2 D1-D2 + LILRB1 D3-D4
LILRB1 D1-D2 + LILRB2 D3-D4
LILRB2 D1 + LILRB1 D2-D4
LILRB1 D1 + LILRB2 D2-D4
Include site-directed mutagenesis:
Utilize appropriate assay systems:
Include both ligands:
Test Angptl2 binding and activation
Compare with HLA-G binding patterns
Evaluate differences in downstream signaling
To accurately study LILRB2 function, researchers must carefully consider experimental systems that recapitulate physiological conditions:
Isolated CD34+ hematopoietic stem/progenitor cells from cord blood or bone marrow
Primary monocytes and dendritic cells
Ex vivo expanded HSCs using defined conditions with immobilized anti-LILRB2
Patient-derived leukemia samples
Chimeric receptor reporter systems using appropriate cell backgrounds
CRISPR-engineered cell lines with modified LILRB2 loci
Inducible expression systems to control LILRB2 levels
Ligand presentation: Immobilized versus soluble ligands produce different outcomes
Multimerization status: Ensure ligands form appropriate high-molecular-weight species
Physiological expression levels: Avoid overexpression artifacts
Signaling readouts: Include multiple downstream pathways (SHP-1/2, calcium flux)
Functional endpoints: Measure effects on differentiation, proliferation, and cellular function
Use primary cells for final validation of mechanisms identified in cell lines
Implement single-cell approaches to account for heterogeneity in primary populations
Consider 3D culture systems to better mimic in vivo microenvironments
Employ multiple ligands to compare signaling outcomes
Include genetic diversity analysis when using primary cells from different donors
LILRB2 genetic diversity likely influences susceptibility to hematological malignancies through several mechanisms:
Functional polymorphisms in LILRB2 may influence immune response diversity among individuals and consequently, their susceptibility to diseases .
Specific haplotypes and SNPs may affect LILRB2's ability to regulate HSC self-renewal versus differentiation, potentially predisposing to leukemia in certain genetic backgrounds .
LILRB2 is required for leukemia development as it inhibits differentiation and promotes self-renewal of leukemic progenitors . Genetic variants that enhance or diminish these functions could modify leukemia risk.
Selection signatures detected in LILRB2 coding regions suggest functional importance of certain polymorphisms that may influence disease susceptibility.
Population-specific patterns of LILRB2 genetic diversity may contribute to differences in hematological malignancy incidence across ethnic groups.
Future research should focus on large-scale association studies correlating LILRB2 genotypes with disease incidence and outcomes across diverse populations.
LILRB2's role in HSC expansion has significant implications for clinical cell therapy applications:
Development of improved ex vivo expansion protocols: Immobilized anti-LILRB2 antibodies induce more potent activation than Angptl2, leading to better HSC expansion . This approach can help overcome limited cell numbers in cord blood transplantation.
Serum-free culture systems: The development of defined, serum-free cultures containing immobilized anti-LILRB2 antibodies supports stable expansion of repopulating human cord blood HSCs , reducing variability and regulatory concerns for clinical applications.
Preservation of stemness: LILRB2 activation supports expansion while maintaining repopulating potential , addressing a key challenge in HSC expansion protocols.
Targeted approaches: Understanding the specific motif in LILRB2's Ig domains critical for activation enables the development of more specific agonists for clinical use.
Quality control: Knowledge of LILRB2 signaling provides molecular markers to assess the quality of expanded HSCs before clinical use.
Therapeutic targeting of LILRB2 would require fundamentally different approaches in autoimmune conditions versus cancer contexts:
Goal: Enhance LILRB2 inhibitory function to suppress overactive immune responses
Approach: Develop agonistic antibodies or multimeric ligands that activate LILRB2 signaling
Target cells: Myeloid cells, dendritic cells
Considerations: Avoid excessive immunosuppression that might increase infection risk
Biomarkers: Monitor SHP-1/2 activation in myeloid cells
Goal: Block LILRB2 inhibitory signals to enhance anti-tumor immunity and/or inhibit leukemic stem cell self-renewal
Approach: Develop antagonistic antibodies or small molecules that prevent LILRB2 signaling
Target cells: Tumor-associated macrophages, leukemic stem cells
Considerations: Potential effects on normal HSCs must be carefully evaluated
Biomarkers: Assess differentiation markers in leukemic cells, immune activation in tumor microenvironment
These divergent approaches highlight the context-dependent nature of LILRB2 function and the need for careful therapeutic design based on disease mechanism.
Leukocyte Immunoglobulin-Like Receptor B2 (LILRB2), also known as ILT4, is an immunoinhibitory protein expressed on the surface of myeloid cells. This receptor plays a crucial role in regulating immune responses and has garnered significant interest in the field of immuno-oncology due to its potential as a therapeutic target .
LILRB2 is a member of the leukocyte immunoglobulin-like receptor (LILR) family, which includes both activating and inhibitory receptors. LILRB2 contains long cytoplasmic tails with immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Upon activation, these ITIMs recruit SHP protein tyrosine phosphatases, which subsequently impact various cellular processes such as growth, migration, invasion, differentiation, survival, and cellular trafficking .
LILRB2 binds to major histocompatibility complex (MHC) class I molecules, such as HLA-G and HLA-A. This binding inhibits myeloid cell activation and promotes an anti-inflammatory state, often referred to as an M2-like state. This immunosuppressive effect is crucial for maintaining immune homeostasis and preventing excessive inflammation .
Given its role in immune regulation, LILRB2 has been identified as a promising target for cancer immunotherapy. For instance, JTX-8064 is a humanized monoclonal antibody that specifically binds to and antagonizes LILRB2. By blocking LILRB2, JTX-8064 induces a pro-inflammatory, anti-tumor state in macrophages, which can enhance the body’s immune response against tumors .
Research on LILRB2 has primarily focused on its role in cancer and its potential as a therapeutic target. Studies have shown that antagonizing LILRB2 can lead to significant pharmacodynamic responses, including macrophage polarization, interferon-gamma signaling, and T cell inflammation. These findings suggest that targeting LILRB2 could be an effective strategy for enhancing anti-tumor immunity .