SIGLEC5 Human Recombinant produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 667 amino acids (17-441a.a.) and having a molecular mass of 74.2kDa (Molecular size on SDS-PAGE will appear at approximately 70-100kDa). SIGLEC5 is expressed with a 239 amino acid hIgG-His Tag at C-Terminus and purified by proprietary chromatographic techniques.
Sialic Acid Binding Ig Like Lectin 5, Obesity-Binding Protein 2, OB-Binding Protein 2, CD33 Antigen-Like 2, SIGLEC-5, CD33L2, OB-BP2, OBBP2, Sialic Acid-Binding Immunoglobulin-Like Lectin 5, Sialic Acid Binding Ig-Like Lectin 5, Sialic Acid-Binding Ig-Like Lectin 5, CD170 Antigen, CD170, Sialic acid-binding Ig-like lectin 5, Siglec-5, CD33 antigen-like 2, Obesity-binding protein 2, OB-BP2, OB-binding protein 2, CD170.
Sf9, Baculovirus cells.
ADLEKPVYEL QVQKSVTVQE GLCVLVPCSF SYPWRSWYSS PPLYVYWFRD GEIPYYAEVV ATNNPDRRVK PETQGRFRLL GDVQKKNCSL SIGDARMEDT GSYFFRVERG RDVKYSYQQN KLNLEVTALI EKPDIHFLEP LESGRPTRLS CSLPGSCEAG PPLTFSWTGN ALSPLDPETT RSSELTLTPR PEDHGTNLTC QMKRQGAQVT TERTVQLNVS YAPQTITIFR NGIALEILQN TSYLPVLEGQ ALRLLCDAPS NPPAHLSWFQ GSPALNATPI SNTGILELRR VRSAEEGGFT CRAQHPLGFL QIFLNLSVYS LPQLLGPSCS WEAEGLHCRC SFRARPAPSL CWRLEEKPLE GNSSQGSFKV NSSSAGPWAN SSLILHGGLS SDLKVSCKAW NIYGSQSGSV LLLQGRSNLG TGVVPAALLE PKSCDKTHTC PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN AKTKPREEQY NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP QVYTLPPSRD ELTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG KHHHHHH.
SIGLEC5 is a transmembrane protein belonging to the CD33-related Siglec family. It contains an N-terminal V-set immunoglobulin-like domain that mediates sialic acid binding, followed by variable numbers of C2-set domains, a transmembrane region, and a cytoplasmic tail containing immunoreceptor tyrosine-based inhibitory motifs (ITIMs) .
The SIGLEC5 V-set domain contains a critical conserved arginine residue essential for sialic acid recognition - a feature shared with other Siglec family members. Interestingly, evolutionary analysis reveals that this arginine residue underwent mutation in non-human hominids but was restored in humans, suggesting human-specific adaptations in SIGLEC5 function .
Unlike some other Siglecs that show strong preference for particular sialic acid linkages, SIGLEC5 binds equally to alpha2,3-linked and alpha2,6-linked sialic acid residues . This broader binding profile may reflect its evolved functions in human immune regulation.
Researchers employ multiple complementary approaches to distinguish between membrane-bound and soluble SIGLEC5 (sSIGLEC5):
Flow cytometry analysis: Using fluorophore-conjugated anti-SIGLEC5 antibodies (e.g., SIGLEC5-PE) to detect surface expression on intact cells, particularly monocytes and neutrophils .
ELISA quantification: Solid-phase sandwich ELISA assays specifically designed to measure sSIGLEC5 in liquid biopsies (serum, plasma) or cell culture supernatants .
Western blotting: To differentiate between the full-length membrane-bound form (~140kDa) and the cleaved soluble form (~100kDa) based on molecular weight.
Cell fractionation studies: To confirm membrane localization versus cytosolic/secreted distribution.
Recent research in sepsis has shown that sSIGLEC5 levels are clinically relevant, with higher concentrations observed in non-survivors compared to survivors, establishing sSIGLEC5 as a potential prognostic biomarker .
SIGLEC5 shows a distinct tissue distribution pattern with highest expression in:
Peripheral blood leukocytes
Spleen
Bone marrow
Lower expression levels are found in:
At the cellular level, SIGLEC5 is primarily expressed by myeloid lineage cells, including monocytes and neutrophils, but is notably absent from leukemic cell lines representing early stages of myelomonocytic differentiation .
Transcriptional regulation of SIGLEC5 involves Hypoxia-Inducible Factor 1-alpha (HIF1α), which binds to specific hypoxia response elements (HREs) in the SIGLEC5 promoter. Chromatin Immunoprecipitation (ChIP) assays have confirmed HIF1α binding to three distinct HREs within the SIGLEC5 promoter region . This regulation mechanism links SIGLEC5 expression to cellular stress responses and inflammatory conditions.
Experimental evidence shows that:
HIF1α-transfected monocytes exhibit increased SIGLEC5 expression at both mRNA and protein levels
The HIF1α inhibitor PX-478 reduces SIGLEC5 expression on monocytes
Hypoxic conditions upregulate SIGLEC5, consistent with the HIF1α regulatory mechanism
During immune activation and inflammatory conditions such as sepsis, SIGLEC5 expression undergoes significant changes:
Increased soluble SIGLEC5: Higher quantities of sSIGLEC5 are detected in plasma from septic patients compared to healthy volunteers, with levels further increasing in septic shock compared to sepsis without shock .
Correlation with disease severity: sSIGLEC5 levels stratify patients according to disease severity, with higher levels correlating with poorer outcomes.
Cellular expression changes: While soluble levels increase, membrane-bound SIGLEC5 expression patterns can vary depending on the inflammatory stimulus and cell type.
Regulatory mechanisms: The HIF1α pathway appears central to SIGLEC5 upregulation during inflammation, linking cellular stress responses to SIGLEC5 expression .
These findings suggest SIGLEC5 represents a dynamically regulated immune checkpoint molecule whose expression reflects and potentially influences the inflammatory state of the host.
Studying SIGLEC5-ligand interactions requires multiple complementary approaches:
Binding assays with recombinant proteins:
Surface Plasmon Resonance (SPR) to measure binding kinetics
ELISA-based binding assays with immobilized potential ligands
Glycan array screening to identify specific sialic acid structures recognized by SIGLEC5
Cellular interaction studies:
Flow cytometry-based binding assays using SIGLEC5-Fc fusion proteins
Cell adhesion assays between SIGLEC5-expressing cells and cells displaying potential ligands
Proximity ligation assays (PLA) to visualize protein-protein interactions in situ
Functional validation:
Blocking experiments using anti-SIGLEC5 antibodies or siRNA knockdown
Site-directed mutagenesis of the sialic acid binding domain
CRISPR-Cas9 gene editing to create SIGLEC5 knockout cell lines
Specialized techniques for SIGLEC5-PSGL1 interactions:
The most informative approach combines binding studies with functional readouts, such as measuring CD8+ T cell proliferation or cytokine production following modulation of SIGLEC5-ligand interactions.
When measuring sSIGLEC5 in clinical samples, researchers should implement these critical controls and validation steps:
Pre-analytical considerations:
Standardized sample collection procedures (timing, anticoagulants)
Consistent processing protocols with minimal freeze-thaw cycles
Proper storage conditions (-80°C for long-term storage)
Analytical validation:
Standard curve with recombinant SIGLEC5 covering the expected concentration range
Spike-and-recovery experiments to assess matrix effects
Precision assessment (intra- and inter-assay CV typically <10%)
Linearity of dilution to confirm accurate quantification across the measurement range
Reference controls:
Age- and sex-matched healthy control samples processed identically
Positive controls from patients with known elevated sSIGLEC5 levels
Internal quality control samples to monitor assay performance over time
Complementary measurements:
Parallel assessment of membrane-bound SIGLEC5 on circulating monocytes when feasible
Measurement of related inflammatory biomarkers to contextualize sSIGLEC5 results
Assessment of potential confounding factors (renal function, medication effects)
Statistical considerations:
Implementing these controls ensures reliable and clinically meaningful sSIGLEC5 measurements.
SIGLEC5 shows several human-specific evolutionary changes with significant functional implications:
Arginine residue restoration: The critical arginine residue in the V-set domain appears to have been mutated in non-human hominids but was restored in humans, potentially altering sialic acid binding preferences .
Sialic acid preference shift: While ancestral Siglecs preferentially recognized Neu5Gc (N-glycolylneuraminic acid), human SIGLEC5 binds preferentially to Neu5Ac (N-acetylneuraminic acid). This shift occurred following the human-specific mutation in the CMAH gene that eliminated Neu5Gc production in humans .
Expression pattern differences: Human SIGLEC5 shows expression patterns that may differ from its non-human primate orthologs, potentially reflecting adaptation to human-specific immune challenges.
These evolutionary changes have important functional implications:
Pathogen interaction: Human-specific SIGLEC5 binding preferences may influence interactions with pathogens that have co-evolved with humans and express Neu5Ac.
Immune regulation: Changes in SIGLEC5 binding properties likely alter its role in discriminating between "self" and "non-self," potentially contributing to unique aspects of human immune regulation.
Disease susceptibility: Human-specific SIGLEC5 properties may influence susceptibility to certain infectious or inflammatory diseases not observed in non-human primates .
These evolutionary differences highlight the importance of careful interpretation when extrapolating from animal models to human SIGLEC5 biology.
Multiple lines of evidence support SIGLEC5's role as an immune checkpoint molecule:
Structural homology: Sequence alignment analysis reveals that human SIGLEC5 shares canonical Ig-like-V-type domains with established immune checkpoint molecules, with identity ranges of 14-25% and similarity ranges of 22-38% - comparable to homology observed among established checkpoint molecules like the B7 family proteins .
Functional effects on T cells:
Receptor-ligand interaction: SIGLEC5 binds to CD8+ T cells via P-selectin glycoprotein ligand-1 (PSGL1) in a sialic acid-dependent manner. PSGL1 had previously been identified as an immune checkpoint receptor that promotes CD8+ T cell exhaustion .
Reversibility of effects: Blockade of the SIGLEC5/PSGL1 interaction reverses the impaired CD8+ proliferation and blocks apoptosis, consistent with checkpoint inhibition mechanisms .
Clinical relevance: Elevated sSIGLEC5 levels in sepsis correlate with increased mortality and impaired T cell function, suggesting pathophysiological relevance of this checkpoint pathway .
This evidence collectively establishes SIGLEC5 as a biologically significant immune checkpoint molecule with potential therapeutic implications.
SIGLEC5 plays a significant role in sepsis pathophysiology through several mechanisms:
Biomarker of disease severity and prognosis:
Immune dysregulation mechanisms:
Regulatory pathways:
Potential therapeutic approaches targeting SIGLEC5 in sepsis include:
Blocking antibodies: Antibodies disrupting the SIGLEC5/PSGL1 interaction have shown promise in reversing T cell dysfunction in experimental models.
HIF1α inhibition: Agents like PX-478 that target HIF1α could reduce SIGLEC5 expression.
Soluble decoy receptors: These could neutralize circulating sSIGLEC5.
Peptide inhibitors: Rationally designed peptides targeting the SIGLEC5-PSGL1 binding interface.
Challenges in therapeutic development include potential off-target effects and the need to balance immune modulation without compromising pathogen clearance .
While sepsis represents the most extensively studied disease context for SIGLEC5, emerging evidence suggests its involvement in several other conditions:
Inflammatory disorders:
Chronic inflammatory conditions may show altered SIGLEC5 expression patterns
The HIF1α-SIGLEC5 axis may be activated in inflammatory microenvironments
Infectious diseases:
Autoimmune conditions:
Cancer immunobiology:
Given its immune checkpoint properties, SIGLEC5 may influence anti-tumor immune responses
Hypoxic tumor microenvironments might upregulate SIGLEC5 via HIF1α stabilization
Neurodegenerative diseases:
Research in these areas remains preliminary, representing important opportunities for further investigation.
When confronting contradictory findings about SIGLEC5 function, researchers should implement this systematic approach:
Methodological reconciliation:
Compare detection antibodies and epitopes recognized
Evaluate assay sensitivity and specificity differences
Consider differences in sample processing that might affect SIGLEC5 integrity
Standardize functional readouts when comparing across studies
Biological context considerations:
Cell-type specific effects may explain divergent findings
Membrane-bound versus soluble SIGLEC5 may have distinct functions
Post-translational modifications might differ between experimental systems
Presence of competing ligands in different systems
Experimental validation strategies:
Perform side-by-side comparisons using standardized protocols
Include multiple complementary readouts of SIGLEC5 function
Use both gain-of-function and loss-of-function approaches
Validate findings across different donor samples to account for genetic variation
Integrative analysis approaches:
Systems biology methods to contextualize SIGLEC5 within broader signaling networks
Multi-parameter analysis correlating SIGLEC5 expression with functional outcomes
Machine learning approaches to identify patterns explaining apparent contradictions
Meta-analysis of published data with careful attention to methodological differences
By implementing these strategies, researchers can better resolve contradictions and develop a more nuanced understanding of context-dependent SIGLEC5 functions.
Analyzing SIGLEC5 as a biomarker requires rigorous statistical approaches:
In the sepsis study, ROC curve analysis successfully identified sSIGLEC5 as a survival marker with a specific cut-off value (≤523.6 ng/mL), demonstrating the utility of these approaches in clinical biomarker validation .
Several cutting-edge tools are emerging to advance SIGLEC5 research:
Advanced genetic engineering approaches:
CRISPR-Cas9 engineered cellular models with SIGLEC5 modifications
Domain-specific mutations to dissect structure-function relationships
Inducible expression systems to study temporal aspects of SIGLEC5 function
Humanized mouse models expressing human SIGLEC5
Improved detection and visualization methods:
Super-resolution microscopy techniques to study SIGLEC5 clustering and distribution
Multiparameter flow cytometry panels incorporating SIGLEC5 with other immune markers
Mass cytometry (CyTOF) for high-dimensional analysis of SIGLEC5 in complex cell populations
Ultrasensitive SIGLEC5 detection methods for limited biological samples
Functional screening platforms:
High-throughput screens for small molecule modulators of SIGLEC5 function
Glycan array technologies with expanded sialic acid diversity
CRISPR activation/repression libraries to identify regulators of SIGLEC5 expression
Single-cell functional assays to capture heterogeneity in SIGLEC5 responses
Computational and structural biology tools:
Molecular dynamics simulations of SIGLEC5-ligand interactions
AI-assisted prediction of SIGLEC5 binding partners
Structural biology approaches (cryo-EM, X-ray crystallography) to resolve SIGLEC5 complexes
Systems immunology frameworks to position SIGLEC5 within immune networks
These emerging tools will enable researchers to address previously intractable questions about SIGLEC5 biology and accelerate therapeutic development targeting this pathway.
The SIGLEC5/PSGL1 axis offers promising opportunities for immunotherapeutic development:
Checkpoint blockade strategies:
Monoclonal antibodies blocking SIGLEC5/PSGL1 interaction
Engineered soluble SIGLEC5 decoy receptors to prevent native SIGLEC5 binding
Small molecule inhibitors targeting the binding interface
Peptide-based competitive inhibitors derived from binding site structures
Translational development considerations:
Timing of intervention may be critical (early vs. late sepsis)
Combination with existing sepsis treatments
Patient stratification based on sSIGLEC5 levels or genetic factors
Biomarker-guided therapy to identify optimal responders
Clinical application scenarios:
Sepsis and septic shock
Chronic T cell exhaustion states
Adjunctive therapy for antimicrobial treatment
Potential applications in cancer immunotherapy
Preclinical-to-clinical translation pathway:
Validation in human cell systems and organoids
Humanized mouse models expressing human SIGLEC5/PSGL1
Ex vivo testing on patient samples
Phase 1 safety studies in healthy volunteers before patient trials
Monitoring considerations:
sSIGLEC5 levels as pharmacodynamic biomarkers
T cell functional recovery metrics
Systems-level immune monitoring
Microbiological clearance assessment
The evidence that blocking SIGLEC5/PSGL1 interaction reverses impaired CD8+ proliferation and prevents apoptosis provides strong mechanistic support for therapeutic development, though careful evaluation of potential adverse effects will be essential .
Tissue/Cell Type | Relative Expression Level | Detection Method |
---|---|---|
Peripheral blood leukocytes | High | Flow cytometry, immunoblotting |
Monocytes | High | Flow cytometry |
Neutrophils | High | Flow cytometry |
Spleen | High | Immunohistochemistry, RT-qPCR |
Bone marrow | High | Immunohistochemistry, RT-qPCR |
Lymph nodes | Moderate | Immunohistochemistry |
Lung | Moderate | Immunohistochemistry |
Appendix | Moderate | Immunohistochemistry |
Placenta | Low | Immunohistochemistry, RT-qPCR |
Pancreas | Low | Immunohistochemistry |
Thymus | Low | Immunohistochemistry |
Early myelomonocytic leukemic cell lines | Absent | Flow cytometry, RT-qPCR |
Clinical Group | Mean sSIGLEC5 Level | Clinical Correlation | Statistical Significance |
---|---|---|---|
Healthy volunteers | Baseline | Reference group | - |
Non-infectious SIRS | Moderately elevated | Inflammatory response | p < 0.05 vs. healthy |
Sepsis | Significantly elevated | Organ dysfunction | p < 0.01 vs. healthy |
Septic shock | Highly elevated | Multi-organ failure | p < 0.001 vs. healthy, p < 0.05 vs. sepsis |
Survivors | Lower levels | Better prognosis | p < 0.05 vs. non-survivors |
Non-survivors | Higher levels (>523.6 ng/mL) | Poor prognosis | p < 0.05 vs. survivors |
Siglec-5 is a protein-coding gene that encodes a putative adhesion molecule. This molecule mediates sialic-acid dependent binding to cells, binding equally to alpha-2,3-linked and alpha-2,6-linked sialic acid . The sialic acid recognition site may be masked by cis interactions with sialic acids on the same cell surface .
Siglec-5 plays a crucial role in the immune system. It is a member of the CD33-related subset of Siglecs and inhibits the activation of several cell types, including monocytes, macrophages, and neutrophils . This inhibition is significant for immune regulation and preventing overactivation of immune responses.
Recombinant human Siglec-5 is used in research to study its role in cell-cell interactions and immune modulation. When expressed on COS cells or as a recombinant protein fused to the Fc region of human IgG1, Siglec-5 can mediate sialic acid–dependent binding to human erythrocytes and soluble glycoconjugates .