SIGLEC7 (Sialic acid-binding immunoglobulin-type lectin 7), also designated CD328, is a transmembrane protein belonging to the CD33-related Siglec family. It functions as an immune checkpoint receptor on natural killer (NK) cells, monocytes, and subsets of T cells . Structurally, SIGLEC7 contains:
An N-terminal V-set immunoglobulin (Ig) domain responsible for sialic acid (Sia) binding.
Two C2-set Ig-like domains acting as spacers.
A transmembrane region.
A cytoplasmic tail with immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that suppress immune activation .
SIGLEC7 is predominantly expressed on:
Cell Type | Expression Level | Key References |
---|---|---|
NK cells | High | |
Monocytes | Moderate | |
CD8+ T cells | Low | |
Platelets | Activation-dependent |
In platelets, SIGLEC7 localizes to α-granule membranes and translocates to the surface upon activation, correlating with CD62P expression .
SIGLEC7 recognizes sialylated glycoconjugates via two distinct binding regions:
Involves Arg124 on the F β-strand.
Preferentially binds α2,8-linked disialic acid (diSia) sequences (e.g., GD3 ganglioside) .
Discovered through in silico docking and mutagenesis.
Features Arg67 and Arg92, forming salt bridges with sialic acid carboxyl groups .
GD3 (disialoganglioside).
Branched α2,6-sialyl residues (e.g., DSLc4).
Synthetic sialoglycoconjugates with clustered diSia motifs .
Inhibitory Signaling: Engagement of sialylated ligands triggers ITIM phosphorylation, recruiting phosphatases (e.g., SHP-1/SHP-2) to dampen NK cell cytotoxicity .
Self-Recognition: Distinguishes host cells (high sialylation) from pathogens, preventing autoimmunity .
Cross-linking with gangliosides induces apoptosis via:
Antibody-Based Targeting: Anti-SIGLEC7 monoclonal antibodies (e.g., DB7.2) enhance NK cell-mediated killing of ovarian cancer (OC) cells in vitro (EC₅₀: 82.67 nM) .
In Vivo Efficacy:
CLL B cells overexpress SIGLEC7 ligands (disialyl-T structures) via ST6GalNAc-IV (biosynthesis) and GCNT1 (glycan elongation suppression) .
High ST6GALNAC4 and low GCNT1 mRNA levels correlate with poor prognosis .
Therapeutic Target | Mechanism | Outcome |
---|---|---|
ST6GalNAc-IV KO | Reduces disialyl-T | Restores NK cytotoxicity |
SIGLEC7 blockade | Prevents ITIM signaling | Enhances tumor clearance |
Sialic Acid Binding Ig Like Lectin 7, QA79 Membrane Protein, SIGLEC-7, D-Siglec, AIRM-1, CDw328, AIRM1, P75, Sialic Acid Binding Ig-Like Lectin 19, Pseudogene, Adhesion Inhibitory Receptor Molecule 1, Siglec-7, Sialic Acid Binding Ig-Like Lectin, Pseudogene 2, Sialic Acid Binding Immunoglobulin-Like Lectin 7, Adhesion Inhibitory Receptor Molecule 1, Sialic Acid Binding Ig-Like Lectin 7, Sialic Acid-Binding Ig-Like Lectin 7, CD328 Antigen, SIGLEC19P, P75/AIRM1, SIGLECP2, CD328, QA79.
Sf9, Baculovirus cells.
ADPQKSNRKD YSLTMQSSVT VQEGMCVHVR CSFSYPVDSQ TDSDPVHGYW FRAGNDISWK APVATNNPAW AVQEETRDRF HLLGDPQTKN CTLSIRDARM SDAGRYFFRM EKGNIKWNYK YDQLSVNVTA LTHRPNILIP GTLESGCFQN LTCSVPWACE QGTPPMISWM GTSVSPLHPS TTRSSVLTLI PQPQHHGTSL TCQVTLPGAG VTTNRTIQLN VSYPPQNLTV TVFQGEGTAS TALGNSSSLS VLEGQSLRLV CAVDSNPPAR LSWTWRSLTL YPSQPSNPLV LELQVHLGDE GEFTCRAQNS LGSQHVSLNL SLQQEYTGKM RPVSGVLLLE PKSCDKTHTC PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN AKTKPREEQY NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP QVYTLPPSRD ELTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG KHHHHHH.
SIGLEC7 belongs to the CD33-like siglec family and contains a V-set domain that recognizes sialic acids, followed by C2-set Ig-domains that act as spacers or regulators of oligomerization. Most importantly, SIGLEC7 has an immunoreceptor tyrosine-based inhibitory/switch motif (ITIM/ITSM) in its cytosolic region . When SIGLEC7 binds to sialic acid-containing ligands, these motifs undergo tyrosine phosphorylation by Src family kinases, which initiates transduction of inhibitory signals into SIGLEC7-expressing cells . This inhibitory function is particularly important in natural killer cells, where SIGLEC7 engagement suppresses cytotoxic activity.
Methodologically, researchers studying SIGLEC7 structure-function relationships should employ site-directed mutagenesis to identify key binding residues and signaling motifs, coupled with functional assays to assess downstream effects on immune cell activity.
The primary ligands for SIGLEC7 are sialic acid-containing glycans, particularly tri-sialylated T antigen. Research has identified specific carrier proteins for these ligand glycans, including PODXL, MUC13, and PDPN .
To identify and characterize these ligands, researchers should:
Transfect cells with sialyltransferase cDNAs to generate potential ligands
Perform mass spectrometry analysis to determine glycan structures
Use equilibrium dialysis and STD-NMR experiments to confirm binding interactions
Conduct inhibition analysis using synthetic sialoglycoconjugates and ganglioside-based glycoconjugates
Validate functional significance through co-culture experiments with SIGLEC7-expressing immune cells
This multi-faceted approach allows for comprehensive characterization of SIGLEC7 ligand structures and their functional relevance.
Recent research has identified two distinct sialic acid-binding regions in SIGLEC7: the well-known primary binding site (site 1 containing R124) and a newly discovered binding region (site 2 containing R67) . To effectively study these binding regions, researchers should employ:
In silico analysis to predict potential binding sites
Site-directed mutagenesis to confirm the functional relevance of predicted residues
Equilibrium dialysis and STD-NMR experiments to directly measure binding interactions
Inhibition analysis using synthetic glycoconjugates to assess binding specificity
Structural studies to understand how the flexible conformation of the C-C′ loop affects the relationship between the two binding sites
These approaches collectively provide a comprehensive understanding of SIGLEC7's complex binding properties and can inform the design of therapeutic agents targeting specific binding regions.
SIGLEC7 expression shows notable variation across cancer types and subtypes, with significant implications for prognosis and treatment strategies. In gliomas, SIGLEC7 is particularly upregulated in the mesenchymal subtype compared to other subtypes, with ROC curve analysis yielding AUC values of 94.6% in TCGA and 87.1% in CGGA databases . This mesenchymal enrichment is significant as this subtype is associated with more aggressive disease and poorer outcomes.
To study SIGLEC7 expression variation methodologically:
Analyze transcriptomic data from databases like TCGA and CGGA
Validate with immunohistochemistry on tissue microarrays
Correlate expression with clinical parameters and survival outcomes
Perform single-cell RNA sequencing to identify specific cell populations expressing SIGLEC7 within the tumor microenvironment
The high expression of SIGLEC7 in aggressive cancer subtypes suggests its potential as both a prognostic biomarker and therapeutic target.
SIGLEC7 appears to play a significant role in macrophage polarization towards an immunosuppressive M2 phenotype. Single-cell sequencing analysis across multiple databases has revealed a striking consistency between the distribution patterns of SIGLEC7 and CD163, a typical marker for M2 macrophages .
To investigate this relationship methodologically:
Perform single-cell RNA sequencing on tumor samples to identify co-expression patterns
Use flow cytometry to quantify SIGLEC7 expression on different macrophage subsets
Conduct in vitro polarization experiments using monocyte-derived macrophages with SIGLEC7 manipulation
Analyze the impact of SIGLEC7 blockade on macrophage phenotype and function in humanized mouse models
Gene enrichment analysis further supports this relationship, showing that SIGLEC7 predominantly participates in immune response processes in glioma, influencing its malignant progression through modification of the immune microenvironment .
SIGLEC7 shows significant positive correlations with established immune checkpoint genes, including TIM-3, HVEM, CD200R1, CD47, TIGIT, CTLA4, PD-1, and PD-L2 . This correlation suggests that SIGLEC7 may function as part of a broader immune checkpoint network.
To study these interactions methodologically:
Perform correlation analysis using transcriptomic data from cancer databases
Conduct co-expression studies at the protein level using multi-color flow cytometry
Investigate the functional consequences of co-blockade using in vitro and in vivo models
Analyze signaling pathway convergence through phospho-proteomic approaches
Understanding these interactions is crucial for developing effective combinatorial immunotherapy strategies. Gene Set Variation Analysis (GSVA) indicates that SIGLEC7 expression correlates more strongly with inhibitory immune functions than with promotive functions, particularly in regulating responses to tumor cells .
When SIGLEC7 engages with its ligands, several key signaling events occur in immune cells:
Phosphorylation of ITIM/ITSM by Src family kinases
Recruitment and activation of SHP-1 (Src homology region 2 domain-containing phosphatase-1)
Transduction of inhibitory signals that reduce cytotoxic activity
The phosphorylation levels of Siglec7-ITIM/ITSM and SHP-1 can increase more than 2- and 10-fold, respectively, compared to baseline levels when interacting with ligand-expressing cells .
To study these pathways methodologically:
Use co-immunoprecipitation to detect protein-protein interactions
Perform Western blotting to monitor phosphorylation status
Employ phospho-flow cytometry for single-cell analysis
Create mutant constructs to identify essential signaling motifs
Use specific inhibitors to block pathway components and observe functional consequences
Understanding these signaling mechanisms is essential for developing strategies to modulate SIGLEC7 activity in therapeutic contexts.
A fascinating aspect of SIGLEC7 biology is its capacity for bidirectional signaling, affecting both the SIGLEC7-expressing immune cells and the target cells bearing ligand glycans:
In SIGLEC7-expressing immune cells:
Inhibitory signaling through ITIM/ITSM phosphorylation
SHP-1 recruitment and activation
In target cells expressing ligand glycans:
Phosphorylation of AKT and ERK1/2
Cytoskeletal activation with co-localization of actin filaments
Enhanced invasion activity without significant changes in proliferation
To study this bidirectional signaling methodologically:
Set up co-culture experiments with SIGLEC7-expressing immune cells and ligand-expressing target cells
Monitor signaling events in both cell types simultaneously
Use recombinant SIGLEC7-Fc fusion proteins to stimulate cells and isolate SIGLEC7-specific events
Perform functional assays such as transwell invasion assays to assess phenotypic consequences
This dual signaling may contribute to immune evasion and enhanced malignancy in cancer contexts, making it an important area for therapeutic intervention.
Distinguishing SIGLEC7-specific signaling from other immune receptors presents methodological challenges that require careful experimental design:
Use recombinant SIGLEC7-Fc conjugated with protein A beads for selective stimulation
Generate SIGLEC7 knockout/knockdown models for comparison with wild-type cells
Employ SIGLEC7-specific blocking antibodies in complex cellular systems
Utilize receptor mutants with specific binding or signaling defects
Perform phospho-proteomic analysis to identify unique signaling signatures
When analyzing cancer cell lines in co-culture with SIGLEC7-expressing immune cells, researchers should include appropriate controls to exclude signals from other cellular interactions. For example, using "non-cytotoxic" U937-Siglec7 high cells instead of KHYG-1-Siglec-7 high cells can help isolate SIGLEC7-specific intracellular signals .
SIGLEC7 expression shows significant correlation with clinical outcomes in glioma patients:
Methodologically, researchers evaluating SIGLEC7 as a prognostic biomarker should:
Use large, well-annotated patient cohorts with long-term follow-up
Apply robust statistical methods including multivariate analysis
Validate findings across independent datasets
Consider molecular subtyping and immune infiltration metrics in their analysis
The strong correlation between SIGLEC7 expression and poor prognosis suggests its potential utility as a prognostic biomarker in cancer.
Targeting SIGLEC7 for cancer immunotherapy offers several promising approaches:
Blocking antibodies: Developing Fc-engineered antibodies that specifically block SIGLEC7-ligand interactions without depleting SIGLEC7-expressing cells .
Glycan modification: Targeting the four sialyltransferases involved in generating SIGLEC7 ligands to reduce their expression on cancer cells .
Combination therapy: Using SIGLEC7 blockade in combination with other checkpoint inhibitors, as SIGLEC7 expression correlates with established immune checkpoint molecules .
Cell-specific targeting: Designing approaches that account for tissue-specific effects, as "the impact of these Siglecs on tumor progression is highly dependent on the anatomical distribution of the tumor and, as a consequence, the local tumor microenvironment" .
Humanized mouse models expressing human SIGLEC7 provide valuable platforms for testing these therapeutic strategies. Studies have shown that SIGLEC7 blockade can significantly reduce tumor burden in vivo, supporting the use of antibodies targeting SIGLEC7 to therapeutically enhance antitumor immunity .
SIGLEC7 expression offers significant potential for improving patient stratification in clinical trials:
Molecular subtyping: SIGLEC7 shows specific enrichment in the mesenchymal subtype of gliomas, with ROC analysis demonstrating high specificity (AUC values of 94.6% in TCGA and 87.1% in CGGA databases) .
Immune landscape classification: SIGLEC7 expression correlates with specific immune cell infiltration patterns, particularly M2 macrophages, allowing for stratification based on immune microenvironment .
Response prediction: High expression of SIGLEC7 may identify patients more likely to benefit from immunotherapies targeting myeloid cells or combinatorial approaches.
Methodologically, researchers implementing SIGLEC7-based stratification should:
Develop standardized assays for SIGLEC7 expression assessment
Establish clinically relevant cutoff values
Integrate SIGLEC7 with other biomarkers for comprehensive patient profiling
Conduct retrospective analyses of existing clinical trial data stratified by SIGLEC7 expression
This approach could enhance the precision of immunotherapy clinical trials and ultimately improve treatment outcomes for cancer patients.
Developing appropriate humanized mouse models is critical for studying human SIGLEC7 function in vivo:
Expression recapitulation: Models should accurately express human SIGLEC7 with the correct tissue distribution pattern. Research has demonstrated that properly humanized mice "recapitulate the expression pattern of the human Siglec-7 and Siglec-9 on peripheral blood mononuclear" cells .
Functional validation: The humanized receptors should retain their binding specificity and signaling capabilities.
Immune competence: Models should maintain a functional immune system to study SIGLEC7's role in immune regulation.
Tumor modeling: For cancer research, the models should support the growth of human tumors in an immunocompetent background.
Methodologically, researchers can:
Use CRISPR/Cas9 to replace murine Siglec genes with human SIGLEC7
Adopt knock-in approaches for physiological expression levels
Utilize conditional expression systems for tissue-specific studies
Validate models by comparing receptor function to human immune cells
These humanized models provide valuable platforms for testing SIGLEC7-targeting therapeutics and understanding in vivo functions that cannot be fully recapitulated in vitro .
Comprehensive analysis of SIGLEC7's impact on the tumor immune microenvironment requires multi-dimensional approaches:
Single-cell RNA sequencing: To identify specific cell populations expressing SIGLEC7 and analyze their transcriptional states. This has revealed consistent distribution patterns between SIGLEC7 and M2 macrophage markers like CD163 .
Multiplex immunohistochemistry/immunofluorescence: To visualize the spatial relationships between SIGLEC7-expressing cells and other immune populations within the tumor microenvironment.
CyTOF (mass cytometry): To analyze multiple protein markers simultaneously on single cells, allowing for detailed immune phenotyping.
Spatial transcriptomics: To map gene expression patterns within the tissue context, preserving spatial information.
Functional assays: To assess the impact of SIGLEC7 manipulation on immune cell function, including cytokine production, cytotoxicity, and migration.
Comparison of immune cell infiltration between patient groups stratified by SIGLEC7 expression has revealed significant differences in multiple immune cell populations, with notable impact on macrophage subsets . This suggests SIGLEC7 plays a key role in shaping the immune landscape of tumors.
Distinguishing the effects of SIGLEC7 across diverse immune cell populations requires sophisticated experimental approaches:
Cell type-specific knockdown/knockout: Using lineage-specific promoters or conditional systems to manipulate SIGLEC7 expression in specific immune cell subsets.
Adoptive transfer experiments: Transferring defined immune cell populations with or without SIGLEC7 expression into humanized mouse models.
Ex vivo functional assays with isolated subsets: Testing various immune functions (cytotoxicity, cytokine production, proliferation, migration) in purified populations.
Multispectral flow cytometry: Simultaneously analyzing SIGLEC7 expression, activation markers, and functional readouts across multiple immune cell types.
In vitro co-culture systems: Reconstituting complex cellular interactions under controlled conditions to isolate SIGLEC7-dependent effects.
These approaches can reveal cell type-specific functions of SIGLEC7, which is particularly important given that SIGLEC7 is expressed on both NK cells and monocyte/macrophage lineages, where it may have distinct functional consequences .
Sialic acid-binding immunoglobulin-like lectin 7 (Siglec-7) is a member of the Siglec family, which are sialic acid-binding proteins predominantly expressed on immune cells. Siglec-7 is known for its role in modulating immune responses and maintaining immune homeostasis. This article delves into the structure, function, and significance of Siglec-7, particularly focusing on its recombinant form used in research and therapeutic applications.
Siglec-7 is a transmembrane protein characterized by its immunoglobulin (Ig)-like domains. It consists of an extracellular region with three Ig-like domains, a transmembrane region, and a cytoplasmic tail containing immunoreceptor tyrosine-based inhibitory motifs (ITIMs). These ITIMs are crucial for transmitting inhibitory signals within the cell .
Siglec-7 is constitutively expressed on natural killer (NK) cells, monocytes, and a subset of CD8+ T cells . Its expression can be modulated in response to various stimuli, including cytokines and interactions with other cells.
The primary function of Siglec-7 is to recognize and bind sialic acids, which are commonly found on the surface of cells and pathogens. This binding is sialic acid-dependent and plays a significant role in immune regulation. Siglec-7 preferentially binds to α(2,8)-linked disialic acid and α(2,6)-linked sialic acid .
One of the key roles of Siglec-7 is to inhibit the activation of NK cells. When Siglec-7 binds to its ligands on target cells, it transmits inhibitory signals through its ITIMs, leading to the suppression of NK cell cytotoxicity. This mechanism helps prevent excessive immune responses and maintains immune tolerance .
Siglec-7 has been implicated in various diseases, particularly those involving immune dysregulation. For instance, in HIV-1 infection, Siglec-7 expression is reduced on NK cells from viremic patients. This reduction is associated with increased susceptibility to infection and disease progression . Additionally, cancer cells often upregulate Siglec ligands to evade immune surveillance by engaging Siglec-7 and other inhibitory receptors on immune cells .
Recombinant Siglec-7 is a form of the protein produced through recombinant DNA technology. It is used extensively in research to study the protein’s structure, function, and interactions. Recombinant Siglec-7 can be produced in various expression systems, including bacterial, yeast, and mammalian cells, each offering distinct advantages in terms of yield, post-translational modifications, and functionality.
Given its role in immune regulation, Siglec-7 is a potential target for therapeutic interventions. Modulating Siglec-7 activity could enhance immune responses against infections and tumors. For example, blocking Siglec-7 interactions with its ligands may boost NK cell activity and improve anti-tumor immunity . Conversely, enhancing Siglec-7 signaling could be beneficial in conditions characterized by excessive immune activation, such as autoimmune diseases.