SIGLEC10 features five extracellular immunoglobulin (Ig)-like domains: one N-terminal V-set domain responsible for sialic acid binding and four C2-set domains . Its cytoplasmic tail contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs), which recruit phosphatases like SHP-1/SHP-2 to dampen cellular activation . Alternative splicing generates six isoforms, including a secreted variant lacking the transmembrane domain .
SIGLEC10 is expressed on immune cells, including B cells, monocytes, dendritic cells, and subsets of NK cells . Its roles include:
CD24-SIGLEC10 axis: Binds CD24 on tumor cells, inhibiting phagocytosis by macrophages and promoting immune evasion .
T-cell suppression: Interacts with galectin-9 on dendritic cells to suppress adaptive T-cell responses .
Neutrophil modulation: Regulates activation and degranulation in inflammatory environments .
Ligand | Function | Reference |
---|---|---|
CD52 | Targeted by Alemtuzumab; implicated in T-cell regulation | |
CD24 | Promotes tumor immune escape via macrophage inhibition | |
VAP-1 | Facilitates leukocyte adhesion and transmigration |
SIGLEC10 is a negative prognostic marker in multiple cancers, correlating with immunosuppressive microenvironments:
Immunosuppressive pathways: SIGLEC10+ TAMs upregulate PD-1, CTLA-4, and TGF-β signaling, fostering resistance to immunotherapy .
Therapeutic targeting: Blocking SIGLEC10 restores macrophage phagocytosis and enhances PD-1 inhibitor efficacy in preclinical models .
Pathway | Biological Effect | Reference |
---|---|---|
IL-6/JAK/STAT3 | Promotes pro-tumorigenic inflammation | |
TGF-β | Drives fibrosis and immune suppression | |
Phagocytosis inhibition | Mediated via CD24 binding and SHP-1 recruitment |
SIGLEC10 is a promising target for cancer immunotherapy:
SIGLEC10, PRO940, SLG2, SIGLEC-10, Sialic acid-binding Ig-like lectin 10 isoform 3, Siglec-like protein 2.
MDGRFWIRVQ ESVMVPEGLC ISVPCSFSYP RQDWTGSTPA YGYWFKAVTE TTKGAPVATN
HQSREVEMST RGRFQLTGDP AKGNCSLVIR DAQMQDESQY FFRVERGSYV RYNFMNDGFF
LKVTALTQKP DVYIPETLEP GQPVTVICVF NWAFEECPPP SFSWTGAALS SQGTKPTTSH
FSVLSFTPRP QDHNTDLTCH VDFSRKGVSA QRTVRLRVAY APRDLVISIS RDNTPALEPQ
PQGNVPYLEA QKGQFLRLLC AADSQPPATL SWVLQNRVLS SSHPWGPRPL GLELPGVKAG
DSGRYTCRAE NRLGSQQRAL DLSVQYPPEN LRVMVSQANR TVLENLGNGT SLPVLEGQSL
CLVCVTHSSP PARLSWTQRG QVLSPSQPSD PGVLELPRVQ VEHEGEFTCH ARHPLGSQHV
SLSLSVHYKK GLISTAFSNL EPKSCDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR
TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN
GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS
DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GKHHHHHH.
SIGLEC10 is broadly expressed on B cells, dendritic cells, and specific macrophage subsets. Flow cytometry analysis reveals that all B cells express SIGLEC10, along with subsets of human leukocytes including eosinophils, monocytes, and a minor population of natural killer cells. Northern blot analysis demonstrates high mRNA expression levels in peripheral blood leukocytes, spleen, and liver . Detection can be performed using anti-human SIGLEC10 antibodies with phycoerythrin-conjugated secondary antibodies in flow cytometry protocols .
Human SIGLEC10 is a transmembrane protein containing an N-terminal V-set immunoglobulin-like domain responsible for sialic acid binding, followed by two C2-set immunoglobulin-like domains . The protein's structural model reveals:
A critical arginine residue in the V-set domain mediating sialic acid recognition
Unlike the typical 7 β-strand structure of C2-type immunoglobulin folds, SIGLEC10's second domain contains only 6 β-strands (ABE and CFG)
The seventh β-strand (C') is replaced by a highly flexible loop between strands C and E (CE loop) that forms two one-turn helices
Structural modeling approaches using related Siglecs as templates (such as Siglec-5) can be employed to study SIGLEC10's conformation and binding interfaces.
SIGLEC10 expression is dynamically regulated during pathological states. Research shows that SIGLEC10 levels are elevated in peritoneal cells following RNA virus infections . In glioma studies, SIGLEC10 expression correlates with poor survival outcomes and acts as an immunosuppressor . Methodologically, researchers can:
Use qPCR to quantify expression changes during disease progression
Apply single-sample Gene Set Enrichment Analysis (ssGSEA) to identify correlations between SIGLEC10 and immune-related genes
Utilize immunohistochemistry to visualize expression patterns in tissue sections from different pathological states
SIGLEC10 demonstrates selective binding to specific sialylated structures. Methodologically, this can be investigated using:
Cell-based glycan arrays comprised of isogenic human cells displaying diverse sialic acid configurations
Binding assays using recombinant SIGLEC10-Fc fusion proteins
Competitive inhibition experiments with synthetic sialylated compounds
Research shows SIGLEC10 binds strongly to CD24, a glycosyl-phosphatidylinositol-anchored sialoprotein, in a sialylation-dependent manner . This binding depends on the critical arginine residue in SIGLEC10's V-set domain that mediates sialic acid recognition .
SIGLEC10 has been identified as a leukocyte ligand for vascular adhesion protein-1 (VAP-1), a glycoprotein expressed on inflamed endothelium with dual roles as an amine oxidase enzyme and an adhesion molecule . This interaction:
Mediates lymphocyte adhesion to endothelium during inflammation
Forms a transient Schiff base between SIGLEC10 and VAP-1 during catalytic reactions
Produces hydrogen peroxide that modulates additional adhesion molecules, signaling molecules, and transcription factors
The binding mechanism involves the WVLQNRVLSSS peptide from SIGLEC10, which docks into the active site of VAP-1, with the arginine (R293) forming a covalent bond with topaquinone . This interaction can be studied using:
Ex vivo frozen section adhesion assays with SIGLEC10-expressing lymphocytes
Molecular docking simulations with peptides derived from the CE loop
Hydrogen peroxide production assays to quantify enzymatic activity
The structural basis of SIGLEC10-VAP-1 interaction provides crucial insights for drug development. Docking studies reveal that:
The peptide WVLQNRVLSS from SIGLEC10's CE loop fits optimally into VAP-1's active site cavity
The arginine residue within this peptide is essential for covalent binding to topaquinone in VAP-1
The mouse ortholog (Siglec-G) contains a glutamine instead of arginine in this critical position, preventing similar interaction with VAP-1
These findings suggest design parameters for potential therapeutic agents:
Small molecules mimicking the WVLQNRVLSS peptide to competitively inhibit SIGLEC10-VAP-1 binding
Agents targeting the topaquinone site in VAP-1 to prevent interaction with SIGLEC10
Modified peptides with enhanced stability and bioavailability for anti-inflammatory applications
SIGLEC10 plays a critical role in B-cell tolerance and self-nonself discrimination of the immune system . This function is based on the recognition of sialylated glycans, which are:
Ubiquitously expressed on host tissues
Largely absent from most microbes (except some pathogenic strains)
Key molecular patterns distinguishing self from nonself
Experimentally, this can be investigated by:
Testing B cell responses to synthetic T-independent type 2 (TI-2) antigens harboring sialic acid motifs
Comparing binding of sialylated antigens between wild-type and Siglec-modified B cells
Analyzing B cell activation thresholds in the presence of sialylated self-antigens
SIGLEC10 exhibits significant correlations with immune checkpoint molecules in cancer contexts. Analysis of glioma datasets reveals that SIGLEC10 expression positively correlates with:
Programmed cell death 1 (PDCD1)
Cytotoxic T-lymphocyte-associated protein 4 (CTLA4)
Lymphocyte activating 3 (LAG3)
These correlations suggest SIGLEC10 may function as part of the immunosuppressive network in tumors. Methodologically, researchers can:
Perform correlation analyses between SIGLEC10 and immune checkpoint gene expression in cancer datasets
Conduct ssGSEA to identify relationships with immune-related gene sets
Investigate combinatorial blockade of SIGLEC10 and established checkpoint molecules in cancer models
Several complementary approaches can be used to identify and characterize SIGLEC10 binding partners:
Cell-based glycan arrays: Using isogenic human cell libraries with combinatorial loss/gain of sialyltransferase genes to present diverse sialoglycans in their natural context
Protein capture assays: Testing if recombinant SIGLEC10-Fc fusion proteins can capture potential binding partners (e.g., CD24) expressed on various cell types
Docking simulations: Generating structural models of SIGLEC10 domains based on crystal structures of related Siglecs and performing in silico docking with potential ligands
Ex vivo binding assays: Using frozen tissue sections to assess binding of SIGLEC10-expressing cells under conditions that mimic physiological interactions
To assess the functional outcomes of SIGLEC10 binding to its ligands, researchers can employ:
Hydrogen peroxide production assays: Measuring H₂O₂ generation following SIGLEC10-VAP-1 interaction to quantify enzymatic activity
B cell tolerance assays: Analyzing B cell activation thresholds in response to sialylated versus non-sialylated antigens
Gene expression profiling: Performing RNA-seq or microarray analysis following SIGLEC10 engagement to identify downstream signaling pathways
Functional immune assays: Measuring cytokine production, cellular proliferation, or cytotoxic activity following manipulation of SIGLEC10 signaling
Based on current understanding, several strategies for therapeutic targeting of SIGLEC10 show promise:
Blocking antibodies: Developing antibodies that specifically interfere with SIGLEC10 binding to its ligands
Competitive inhibitors: Designing peptides or small molecules based on the WVLQNRVLSS sequence that competes for binding to VAP-1
Enzymatic modification: Altering the sialylation patterns of SIGLEC10 ligands to modulate binding strength
Combination therapies: Co-targeting SIGLEC10 with established immune checkpoint inhibitors (anti-PD1, anti-CTLA4) based on their correlated expression patterns
SIGLEC10 expression has been identified as a negative predictor of survival in glioma patients . Comprehensive analysis strategies include:
Survival analysis: Kaplan-Meier plots stratifying patients by SIGLEC10 expression levels
Multivariate Cox regression: Determining whether SIGLEC10 is an independent prognostic factor when controlling for other clinical variables
Immune infiltration correlation: Using ssGSEA to assess relationships between SIGLEC10 expression and immune cell infiltration patterns
Pathway enrichment analysis: Performing GO and KEGG analyses to identify biological processes and pathways associated with SIGLEC10 expression in tumors
Siglec-10 is expressed on various immune cells, including dendritic cells, monocytes, B cells, natural killer (NK) cells, and T cells . It plays a crucial role in negatively regulating both innate and adaptive immune responses. This regulation is achieved through its interaction with CD24, which suppresses immune responses to danger-associated molecular patterns (DAMPs) by associating with the tyrosine phosphatase SHP-1, a negative regulator of nuclear factor-kappa B (NF-κB) .
Siglec-10 binds to sialic acids, which are commonly found on the surface of cells. This binding can inhibit the activation of immune cells, thereby preventing an overactive immune response. For instance, Siglec-10 has been shown to bind soluble CD52, leading to the impairment of phosphorylation of the T cell receptor-associated kinases Lck and Zap70, and subsequently, T cell activation . This mechanism is thought to be involved in maintaining T cell homeostasis and preventing autoimmune diseases such as type I diabetes .
Siglec-10’s role in immune regulation makes it a potential target for therapeutic interventions in various diseases. For example, its interaction with CD24 on tumor cells can inhibit the activation of immune cells, allowing tumors to evade the immune system . Blocking Siglec-10 has been proposed as a strategy to reactivate anti-tumor immunity . Additionally, Siglec-10 has been identified as the leukocyte ligand for vascular adhesion protein-1 (VAP-1), which plays a key role in leukocyte trafficking .
Human recombinant Siglec-10 is produced using recombinant DNA technology, which involves inserting the gene encoding Siglec-10 into a suitable expression system, such as bacteria or mammalian cells. This allows for the production of large quantities of the protein for research and therapeutic purposes. Recombinant Siglec-10 is used in various applications, including studying its function in immune regulation and developing potential therapeutic agents targeting Siglec-10 .