LILRA6 protein, a single-pass type I membrane protein, has its N-terminus in the extracellular space and its C-terminus in the cytoplasm. It contains 481 amino acids . The protein has a calculated molecular weight of 48.2 kDa but migrates at 64-70 kDa under reducing conditions on SDS-PAGE due to glycosylation . LILRA6 may act as a receptor for class I MHC antigens . It is expressed in avian macrophages and modulates cytokine production by activating multiple signaling pathways .
LILRA6 interacts with several proteins, including:
SYK: Tyrosine-protein kinase SYK, which regulates innate and adaptive immunity, cell adhesion, osteoclast maturation, platelet activation, and vascular development .
PTPN6: Tyrosine-protein phosphatase non-receptor type 6, which modulates signaling by tyrosine phosphorylated cell surface receptors .
RPL5: 60S ribosomal protein L5, a component of the ribosome responsible for protein synthesis .
HLA-G, HLA-E, HLA-A, HLA-F, HLA-C: Various HLA class I histocompatibility antigens involved in immune regulation and antigen presentation .
LILRB3: Leukocyte immunoglobulin-like receptor subfamily B member 3 .
Copy number variation in the LILRA6 gene correlates with susceptibility to atopic dermatitis (AD) . The presence of one copy of LILRA6 on each chromosome may be a risk factor for AD . LILRA6 interacts with a novel ligand exposed on necrotic epithelial cells .
Table 1: LILRA3 and LILRA6 copy number variation in atopic dermatitis (AD)
| Marker | Number of Copies | Total | AD | P value |
|---|---|---|---|---|
| Unaffected | Affected | |||
| LILRA3 | 0 | 613 (20.7) | 401 (20.9) | 0.714 |
| 1 | 2339 (78.9) | 1512 (78.7) | ||
| 2 | 12 (0.4) | 9 (0.5) | ||
| LILRA6 | 0 | 217 (7.3) | 143 (7.4) | 0.948 |
| 1 | 2107 (71.2) | 1370 (71.3) | ||
| 2 | 617 (20.8) | 395 (20.6) | ||
| 3 | 19 (0.6) | 12 (0.6) | ||
| 4 | 4 (0.1) | 2 (0.1) |
Number of copies denotes copies per haplotype. Frequencies are given in parentheses.
Table 2: Transmission of LILRA3 and LILRA6 among families in relation to atopic dermatitis (AD)
| Number of Copies | Trans | Untrans | T-Freq | U-Freq | P value |
|---|---|---|---|---|---|
| LILRA3 | 0 | 157 | 432 | 0.2224 | 0.2006 |
| 1 | 547 | 1715 | 0.7748 | 0.7962 | |
| 2 | 2 | 7 | 0.00283 | 0.00325 | |
| LILRA6 | 0 | 44 | 200 | 0.06358 | 0.09325 |
| 1 | 498 | 1467 | 0.7197 | 0.6842 | |
| 2 | 145 | 462 | 0.2095 | 0.20155 | |
| 3 | 4 | 14 | 0.00578 | 0.00653 | |
| 4 | 1 | 1 | 0.00145 | 0.000466 |
LILRA6, also known as ILT8 and CD85b, is a transmembrane molecule belonging to the Leukocyte Immunoglobulin-like Receptor (LILR) family of immune regulatory proteins. The mature human LILRA6 protein consists of a 424 amino acid extracellular domain (ECD) containing two immunoglobulin-like domains, a 21 amino acid transmembrane segment, and a 13 amino acid cytoplasmic domain. The protein contains a positively charged arginine residue in the transmembrane segment that likely mediates association with the signaling protein Fc epsilon RI gamma. Alternative splicing can generate a shorter isoform that is truncated near the middle of the ECD .
LILRA6 and LILRB3 represent a pair of activating/inhibitory receptors with nearly identical extracellular domains (94% amino acid sequence identity). They are considered "paired receptors" as they have almost identical extracellular domains but differ significantly in their signaling activities. While LILRB3 is inhibitory and signals through immunoreceptor tyrosine-based inhibition motifs (ITIMs) in its cytoplasmic tail, LILRA6 is activating and signals through association with FcRγ, which contains an immunoreceptor tyrosine-based activation motif (ITAM). This structural arrangement allows these receptors to deliver opposing signals despite recognizing the same ligands .
LILRA6 is primarily expressed on cells of myelomonocytic lineage, with monocytes showing the highest expression levels. This expression pattern is similar to that of LILRB3, which is detected on monocytes, macrophages, dendritic cells, granulocytes, and some T cells. Importantly, previous studies have had difficulty distinguishing between LILRB3 and LILRA6 expression due to their structural similarity, but recent gene-specific analyses have confirmed monocytes as the primary cell type expressing LILRA6. The expression level of LILRA6 on these cells correlates directly with the gene copy number .
LILRA6 exhibits extensive genetic polymorphism and copy number variation (CNV). In a study of Caucasian populations, approximately 32% of individuals possessed more than 2 copies of LILRA6, while 4% had only one copy per diploid genome. Copy numbers can range from one to six per individual. When designing experiments with LILRA6, researchers should consider:
Genotyping subjects for LILRA6 copy number using quantitative PCR methods
Sequencing to identify specific allelic variants
Including appropriate controls that account for CNV diversity
The CNV status can significantly affect expression levels and potentially influence receptor function and signaling capability. Researchers should therefore stratify experimental cohorts based on LILRA6 copy number when analyzing functional outcomes .
When analyzing LILRA6 expression, researchers should implement a comprehensive approach that accounts for both polymorphism and copy number variation:
Use gene-specific primers that distinguish LILRA6 from the highly similar LILRB3
Employ multiple reference genes (e.g., ZNF80, GPR15, or TERT) for accurate normalization
Combine quantitative PCR with genomic DNA analysis to correlate expression with copy number
Consider analyzing multiple gene regions (e.g., 3'UTR and intronic fragments)
When possible, perform allele-specific expression analysis to identify differential expression of variants
A study examining LILRA6 mRNA expression in peripheral blood mononuclear cells found that expression levels correlate with copy number of the gene, suggesting a gene dosage effect that should be accounted for in experimental design and data interpretation .
Differentiating between LILRA6 and LILRB3 presents a significant challenge due to their 94% sequence identity in the extracellular domain. Researchers should consider:
Gene-specific PCR primers targeting divergent regions, particularly in the 3'UTR
Allele-specific oligonucleotide probes for quantitative PCR
Genomic DNA assays to determine copy number alongside expression analysis
Custom antibodies targeted to the few divergent extracellular residues
Functional assays that distinguish between activating (LILRA6) and inhibitory (LILRB3) activity
It is critical to note that most commercially available antibodies cannot distinguish between these receptors, and many previous studies reporting "LILRB3 expression" likely detected both LILRB3 and LILRA6. Researchers should explicitly acknowledge this limitation when interpreting published data .
LILRA6 engages several key signaling pathways that contribute to immune cell activation:
| Signaling Pathway | LILRA6 Effect | LILRB3 Effect | Key Intermediates |
|---|---|---|---|
| Src kinases | Phosphorylation | Inhibition | FcεRIγ, Lyn |
| JAK-STAT | Activation | Suppression | STAT1, STAT3, STAT5 |
| NF-κB | Activation | Inhibition | IKK complex, p65 |
| PI3K/AKT | Activation | Suppression | PDK1, mTOR |
| ERK1/2 MAPK | Activation | Inhibition | Raf, MEK1/2 |
LILRA6 associates with the FcεRIγ adaptor protein through its transmembrane arginine residue, leading to ITAM phosphorylation and downstream signal propagation. In contrast, LILRB3 contains cytoplasmic ITIMs that recruit phosphatases like SHP-1 and SHP-2, resulting in negative regulation of the same pathways. This molecular arrangement allows these paired receptors to fine-tune myeloid cell responses based on the relative expression levels and engagement of each receptor .
Several potential ligands for LILRA6 have been identified:
Cytokeratins 8, 18, and 19 - exposed on necrotic epithelial cells
Angiopoietin-like 7 - demonstrated in binding assays
MHC class I molecules - suggested by functional studies
To investigate novel binding partners, researchers can employ:
Immunoprecipitation followed by mass spectrometry
Recombinant LILRA6-Fc fusion proteins for pull-down assays
Reporter cell assays (e.g., 2B4 reporter system) to validate functional binding
Surface plasmon resonance to quantify binding affinities
Cell-based binding assays using allele-specific LILRA6 variants
Importantly, different LILRA6 alleles show varying binding affinities to ligands. For instance, research on the closely related LILRB3 found that specific alleles (e.g., LILRB312) displayed particularly strong binding to necrotic cells compared to others (e.g., LILRB301). Given the extensive sequence similarity in the extracellular domains, similar allelic binding variation likely exists for LILRA6 .
LILRA6 has been shown to associate with and activate MHC class I molecules and β2-microglobulin. This interaction appears to induce the expression of transporters associated with antigen processing. The functional consequences include:
Enhanced antigen presentation capacity in myeloid cells
Modulation of CD8+ T cell responses
Regulation of NK cell education through altered MHC-I presentation
Potential cross-presentation of exogenous antigens
Unlike LILRB1 and LILRB2, which recognize a broad range of MHC-I molecules through conserved α3 domain and β2-microglobulin interactions, LILRA6 binding appears more selective and may be influenced by specific peptide-MHC complexes. This interaction represents a mechanism by which innate immune receptors can modulate adaptive immune responses .
LILRA6 has been implicated in several autoimmune and inflammatory conditions:
Atopic dermatitis - Human LILRA6 polymorphisms correlate with disease susceptibility
Inflammatory bowel disease - Altered expression observed in patient samples
Rheumatoid arthritis - Potential role in pathogenesis
The activating nature of LILRA6 suggests that enhanced signaling through this receptor could contribute to inappropriate inflammatory responses. Additionally, the variable copy number of LILRA6 may influence disease susceptibility by altering the balance between activating (LILRA6) and inhibitory (LILRB3) signals. Individuals with higher LILRA6 copy numbers might experience enhanced inflammatory responses upon receptor engagement .
Genome-wide association studies have shown that duplications at the LILRA6 locus are associated with high-grade serous ovarian cancer susceptibility. The mechanisms potentially linking LILRA6 CNV to cancer may include:
Altered immune surveillance of transformed cells
Modified tumor microenvironment through cytokine regulation
Enhanced recognition of damage-associated molecular patterns (DAMPs) released by tumor cells
Interaction with cytokeratins exposed on necrotic cancer cells
Researchers investigating this connection should consider stratifying cancer cohorts by LILRA6 copy number and examining functional differences in myeloid cells from individuals with varying copy numbers. Higher LILRA6 expression may lead to altered immune responses against nascent tumors, potentially influencing cancer progression and therapeutic outcomes .
LILRA6 appears to play significant roles in antimicrobial immunity:
Activation of macrophage-mediated immune responses
Induction of Th1-, Th2-, and Th17-type cytokines
Upregulation of Toll-like receptors
Potential recognition of pathogen-associated molecular patterns
LILRA6 activation in the context of infections could enhance inflammatory responses and antimicrobial effector functions. This suggests potential therapeutic applications:
Development of LILRA6 agonists to enhance immunity against specific pathogens
Targeted modulation of LILRA6 signaling to boost vaccine responses
Integration of LILRA6 polymorphism analysis in personalized infection risk assessment
The receptor may be particularly relevant in the context of pathogens like Mycoplasma hyorhinis, Streptococcus pneumonia, Candida albicans, and Legionella pneumophila, where LILR family members have been implicated in host defense mechanisms .
For producing functional recombinant LILRA6:
Expression Systems:
HEK293 cells for mammalian glycosylation patterns
CHO cells for stable high-yield production
E. coli for non-glycosylated fragments (e.g., single domains)
Construct Design:
Full extracellular domain (aa 1-424) for most applications
Fc-fusion proteins for enhanced stability and detection
Inclusion of polyhistidine or other purification tags
Validation Methods:
ELISA binding assays with known ligands (e.g., Angiopoietin-like 7)
Flow cytometry to confirm binding to cellular targets
Reporter cell assays to verify functional activity
Western blotting under reducing and non-reducing conditions
When studying LILRA6, typical ED50 values for functional binding assays range from 0.1-0.6 μg/mL when coated at 2 μg/mL concentration, providing a benchmark for experimental design .
To distinguish LILRA6 functions from other LILR family members:
Use gene-specific knockdown/knockout approaches:
CRISPR-Cas9 targeting unique LILRA6 sequences
siRNA/shRNA designed against divergent regions
Validation with allele-specific PCR
Employ blocking antibodies with verified specificity:
Pre-validation against recombinant proteins
Cross-adsorption to remove antibodies recognizing shared epitopes
Isotype-matched controls
Utilize receptor chimeras:
Swap extracellular domains with cytoplasmic domains
Create LILRA6/LILRB3 chimeras to isolate signaling effects
Compare with wild-type receptors
Implement allele-specific functional assays:
Express distinct LILRA6 alleles in reporter systems
Compare binding and activation profiles
Correlate with genetic variations
When interpreting results, researchers should account for the inherent challenges in distinguishing LILRA6 from LILRB3 due to their high sequence similarity and presence on the same cell populations .
To address the complex relationship between LILRA6 CNV, expression, and function:
Integrated Genomic and Transcriptomic Analysis:
Digital PCR for absolute copy number determination
RNA-seq with allele-specific analysis
Long-read sequencing to resolve structural variations
Single-Cell Approaches:
scRNA-seq to correlate expression with cell states
Mass cytometry for protein-level analysis
Imaging mass cytometry for spatial context
Systems Biology Integration:
Pathway enrichment analysis based on CNV status
Network modeling of signaling differences
Machine learning to identify functional patterns
Longitudinal Studies:
Track expression changes during immune responses
Correlate with disease progression
Monitor in response to therapeutic interventions
Functional Genomics:
CRISPR-based screens in relevant cell types
Isogenic cell lines with defined LILRA6 copy numbers
Synthetic biology approaches to control expression levels
These advanced approaches can help resolve how LILRA6 copy number influences receptor density on the cell surface, potentially affecting signaling thresholds upon ligand engagement and ultimately modulating immune cell function in both health and disease contexts .
The most promising areas for LILRA6 research include:
Precision medicine applications based on LILRA6 genotyping:
Prediction of autoimmune disease susceptibility
Cancer risk stratification
Personalized immunotherapy approaches
Development of LILRA6-targeted therapeutics:
Agonistic or antagonistic antibodies
Small molecule modulators of signaling
Engineered ligands with controlled receptor activation properties
Understanding the evolutionary significance of LILRA6 CNV:
Population genetics across diverse ethnic groups
Selection pressures maintaining polymorphism
Comparative immunology in non-human primates
Elucidation of the complete "ligandome":
Systematic screening for additional binding partners
Structural biology of receptor-ligand complexes
Context-dependent ligand recognition