Fact-checked by Grok 2 weeks ago

Siglec

Siglecs, or sialic acid-binding immunoglobulin-like , constitute a family of cell surface receptors that specifically recognize sialic acid-containing glycans on glycoproteins and glycolipids, primarily expressed on hematopoietic cells of the to regulate innate and adaptive immune responses. In humans, there are 14 functional Siglec genes, encoding proteins such as Siglec-1 through Siglec-4 and Siglec-5 through Siglec-11, as well as Siglec-14 through Siglec-16, with additional pseudogenes reflecting evolutionary dynamics. Structurally, Siglecs are type I transmembrane glycoproteins belonging to the , featuring an extracellular region with a variable number of immunoglobulin-like domains—typically a membrane-distal V-set domain responsible for binding, followed by varying numbers of C2-set domains for interaction and . Intracellularly, most Siglecs contain immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit phosphatases like SHP-1 and SHP-2 upon binding, delivering inhibitory signals to dampen immune ; however, a subset associates with DAP12, an adaptor protein bearing an (ITAM), enabling activating functions. This dual inhibitory-activating potential allows Siglecs to fine-tune immune signaling in response to self-associated molecular patterns (SAMPs) on cells versus pathogen-associated molecular patterns. Siglecs are classified into two main subsets based on evolutionary conservation and sequence similarity: the conserved Siglecs (Siglec-1/sialoadhesin, , Siglec-4/MAG, and Siglec-15), which are ancient and found across tetrapods with relatively stable structures, and the rapidly evolving CD33-related Siglecs ( and Siglec-5 to -11, -14, and -16), which exhibit species-specific duplications, deletions, and polymorphisms driven by -host coevolution. Expression patterns are cell-type specific; for instance, Siglec-1 is predominantly on macrophages and dendritic cells, on B lymphocytes, on myeloid cells including monocytes and , and Siglec-7 and -9 on natural killer cells and subsets of T cells. These patterns underscore their roles in diverse immune contexts, from self/non-self to recognition. Functionally, Siglecs primarily mediate immune inhibition to promote and prevent by recognizing sialylated glycans on healthy cells, thereby setting thresholds for immune ; for example, Siglec-2 on B cells attenuates BCR signaling to control production, while Siglec-3 on myeloid cells modulates in tissues like . Activating Siglecs, such as paired like Siglec-14 (counterpart to inhibitory Siglec-5), can enhance responses against pathogens by promoting or release. Beyond immunity, Siglecs influence , migration, and survival, with implications in diseases including , cancer, neurodegeneration, and infections, where dysregulated sialic acid-Siglec interactions contribute to immune evasion or chronic .

Discovery and History

Initial Identification

The discovery of the first Siglec family member, sialoadhesin (Siglec-1), occurred in when it was identified as a sialic acid-dependent receptor on resident in , specifically recognizing glycoconjugates on sheep erythrocytes. This receptor was characterized as a large, macrophage-restricted glycoprotein that mediated sialic acid-specific hemagglutination, marking it as a novel involved in interactions with sialylated cells. Initial studies highlighted its expression on stromal macrophages in lymphohematopoietic tissues, distinguishing it from other known macrophage markers. In the early 1990s, (Siglec-2) was identified as a B-cell-specific surface that mediated homotypic and heterotypic , particularly to monocytes and erythrocytes. Cloned from a B-lymphocyte , was noted for its seven immunoglobulin-like domains and its role in facilitating cell-cell interactions during B-cell activation. Subsequent investigations in the mid-1990s revealed its function as an inhibitory receptor that modulates B-cell signaling, with studies demonstrating hyperresponsive B cells lacking this regulatory control. CD33 (Siglec-3) was recognized in 1988 through the cloning of its cDNA from a myeloid line, establishing it as a differentiation expressed on myeloid progenitor cells and maturing monocytes but absent on mature granulocytes and non-myeloid lineages. This marker was particularly valuable for identifying early myeloid commitment in hematopoiesis, with monoclonal antibodies like My9 confirming its restricted expression pattern on leukemic blasts in . By the early 1990s, further characterization emphasized its two immunoglobulin-like domains and potential involvement in myeloid , though its precise ligand interactions remained under exploration at the time. Initial studies in the late 1990s linked sialoadhesin, , and through shared structural features, including N-terminal V-set immunoglobulin-like domains that bind sialic acids and multiple C2-set domains in their extracellular regions. These commonalities, along with their expression on hematopoietic cells and roles in sialic acid-dependent recognition, prompted Paul Crocker to propose the unified "Siglec" in 1998, denoting sialic acid-binding immunoglobulin-like as a distinct subfamily of I-type . This classification laid the groundwork for recognizing Siglecs as a cohesive group prior to broader family expansion.

Evolution of the Siglec Family

The Siglec family emerged in the common ancestor of vertebrates, with orthologs of conserved members such as Siglec-4 (myelin-associated glycoprotein, MAG) identified in cartilaginous and , indicating an ancient role in vertebrate immunity and neural function. This phylogenetic origin traces back over 500 million years, predating the diversification of tetrapods, and suggests that Siglecs arose as sialic acid-binding receptors to modulate early immune responses in vertebrates. An ancestral gene is proposed as the progenitor for the rapidly evolving CD33-related (CD33r) subfamily, forming an initial through duplication events that allowed to pressures and host changes. The Siglec family divides into two main subfamilies based on evolutionary conservation and sequence similarity: the conserved group, including Siglec-1 (sialoadhesin), Siglec-2 (), Siglec-4 (MAG), and Siglec-15, which are present across most mammals with stable and consistent sialic acid-binding preferences; and the CD33r subfamily (Siglec-3/ and Siglec-5 through -14 and -16), characterized by rapid evolution, high interspecies variability, and species-specific expansions or contractions. The conserved subfamily maintains core functions in and signaling, while the CD33r group exhibits dynamic changes driven by gene duplications, conversions, and pseudogenizations, reflecting an with pathogens that mimic host sialic acids. For instance, placental mammals display 5–20 CD33r Siglecs, with mice possessing approximately 7 (Siglec-D, -E, -F, -G, -H, -I) compared to 10–13 in s (Siglec-3, -5 to -12, -14, -16), highlighting lineage-specific diversification. These duplication events, often occurring in tandem clusters on s (e.g., chromosome 19q13), have generated functional diversity, including paired activating and inhibitory receptors that fine-tune immune activation. The evolution of Siglecs has been intimately linked to the co-evolution of sialic acids, the primary ligands that enable self-recognition in the immune system. Sialic acids, enriched on vertebrate cell surfaces, serve as "self-associated molecular patterns" (SAMPs) that Siglecs detect to inhibit overzealous immune responses and prevent autoimmunity, a mechanism that likely originated in early jawed vertebrates to distinguish host glycans from microbial mimics. Pathogen exploitation of sialic acid-Siglec interactions has driven selective pressures, leading to expansions in the CD33r subfamily and adaptations like human-specific loss of Neu5Gc (N-glycolylneuraminic acid), which altered ligand availability and Siglec binding specificities to enhance self/non-self discrimination. This co-evolutionary dynamic underscores Siglecs' role in balancing tolerance and defense across vertebrate lineages.

Structure and Classification

Overall Molecular Architecture

Siglecs constitute a family of type I transmembrane glycoproteins within the , characterized by a modular extracellular domain that facilitates recognition and cell-cell interactions. The extracellular region typically comprises an N-terminal V-set immunoglobulin-like domain, responsible for binding, followed by 1 to 16 C2-set immunoglobulin-like domains that provide and extend the receptor to its s. This domain organization varies across family members; for instance, the CD33-related Siglecs generally feature 1 V-set and 1-4 C2-set domains, while sialoadhesin (Siglec-1) possesses 1 V-set and 16 C2-set domains. The V-set domain shares conserved features with other I-type lectins, including a characteristic residue essential for interaction. Anchoring the protein to the plasma membrane is a single-pass , typically a hydrophobic alpha-helix of approximately 20-25 , located membrane-proximally to the extracellular Ig-like domains. This region often includes a basic residue in certain activating Siglecs, such as Siglec-14, Siglec-15, and Siglec-16, which enables non-covalent association with adaptor proteins like DAP12 for . The cytoplasmic tail, generally short (40-50 ), harbors key signaling motifs: most Siglecs contain one or two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) with the (I/V/L/S)-x-Tyr-x-x-(L/V/I), which, upon , recruit Src homology 2 domain-containing phosphatases such as SHP-1 and SHP-2 to dampen immune responses. Notably, Siglec-16 deviates from this inhibitory paradigm, lacking functional ITIMs and instead relying on DAP12 association to transduce activating signals via the ITAM motif in the adaptor. Siglecs are extensively post-translationally modified by N-linked at multiple residues within the extracellular Ig-like domains, contributing to , stability, and protection from . These glycosylation patterns, which include both sialylated and non-sialylated glycans, modulate the overall conformation of the extracellular region, thereby influencing ligand accessibility and preventing unwanted cis-interactions with self-glycans on the same cell surface.

Ligand Binding Mechanism

Siglecs primarily recognize and bind -containing through their amino-terminal V-set immunoglobulin-like () domain, which contains a conserved residue that forms a with the carboxylate group of , such as (Neu5Ac). This interaction anchors the sialic acid in a binding pocket, with additional hydrophobic and polar contacts stabilizing the . The V-set domain's specificity arises from variations in surrounding residues, allowing differential recognition of sialic acid modifications and linkages. Different Siglecs exhibit preferences for specific linkages to underlying , influencing their binding affinities. For instance, Siglec-1 (sialoadhesin) shows a strong preference for α2-3-linked over α2-6-linked ones, enabling selective interactions with certain cell surface glycoconjugates. In contrast, Siglec-2 () favors α2-6 linkages, while other family members like Siglec-7 display affinity for both α2-6 and α2-8 disialylated structures. These linkage preferences, often with dissociation constants in the millimolar range for monovalent , can be enhanced by multivalency in natural contexts. Binding is further modulated by and interactions, where refers to Siglec-ligand engagement on the same cell surface and involves interactions between different cells. ligands, abundant on immune cells expressing Siglecs, can occupy sites and reduce , establishing a for intercellular . This regulatory mechanism helps maintain immune by preventing excessive self-recognition while allowing activation upon encountering hypersialylated ligands, such as on pathogens or tumors. Crystallographic studies have elucidated the structural basis of these interactions, particularly for Siglec-7 bound to Neu5Ac derivatives. High-resolution structures reveal that the sialic acid's forms the key with the conserved (Arg107 in Siglec-7), while the acetamido group at and at C6-9 engage in bonds and van der Waals contacts within the V-set pocket. For example, in complexes with α2-8-linked disialic acids, an additional sialic residue extends into a secondary , enhancing affinity through cooperative interactions. These insights from underscore the molecular determinants of specificity across the Siglec family.

Family Classification

Siglecs are classified into two primary subfamilies based on evolutionary conservation, , and structural characteristics: the conserved subfamily and the CD33-related (CD33r) subfamily. This division reflects distinct phylogenetic branches, with the conserved group representing ancient origins stable across mammals and the CD33r group showing rapid diversification. The conserved subfamily includes Siglec-1 (sialoadhesin), Siglec-2 (), Siglec-4 (myelin-associated glycoprotein), and Siglec-15, which exhibit high sequence similarity to sialoadhesin and are preserved in most mammalian lineages without major expansions or losses. These members feature sialoadhesin-like extracellular architectures, including a prominent N-terminal V-set immunoglobulin domain essential for recognition, and are distinguished by their limited variability across species. The CD33r subfamily encompasses Siglec-3 (), Siglec-5 to Siglec-11, and Siglec-14 to Siglec-16, forming a larger, dynamically evolving cluster marked by interspecies diversity, including pseudogenes (such as human and ), gene duplications, and species-specific variants that contribute to immune adaptation. This subfamily's expansion is particularly evident in , where additional members like Siglec-14 and Siglec-16 arose through recent duplications of Siglec-5 and Siglec-11, respectively. Key classification criteria include the number of extracellular immunoglobulin-like domains, with examples in conserved Siglecs ranging from 2 (Siglec-15) to 17 (Siglec-1), while CD33r members have 2–5 domains; the composition of cytoplasmic tails, where CD33r Siglecs often harbor immunoreceptor tyrosine-based inhibitory motifs (ITIMs) or ITIM-like sequences for potential signaling; and differential preferences for sialic acid α2,3- versus α2,6-linkages in their V-set binding sites. In human nomenclature, SIGLEC genes are designated with numerical identifiers (SIGLEC1–SIGLEC16), predominantly clustered on 19q13.3–q13.4 for the CD33r subfamily and Siglec-2/Siglec-4, while SIGLEC1 localizes to 20p13 and SIGLEC15 to 18q12.3.

Biological Functions

Signaling Pathways

Siglecs primarily exert their regulatory effects through intracellular signaling pathways that can be inhibitory or activating, depending on their cytoplasmic motifs. The majority of Siglecs, such as those in the CD33-related subfamily, contain immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in their cytoplasmic tails. Upon ligand binding and tyrosine phosphorylation by Src family kinases, these ITIMs recruit Src homology 2 (SH2) domain-containing protein tyrosine phosphatases, including SHP-1 and SHP-2. This recruitment leads to of key signaling molecules, such as immunoreceptor tyrosine-based motifs (ITAMs) on associated receptors, thereby dampening immune cell . A prominent example is (Siglec-2) on B cells, where ITIM-mediated SHP-1 recruitment dephosphorylates the (BCR) complex, inhibiting downstream pathways like calcium mobilization and proliferation. In contrast, a of Siglecs lacks ITIMs but features a charged residue in the , enabling association with the adaptor protein DAP12, which contains an ITAM. engagement phosphorylates the DAP12 ITAM, recruiting and activating spleen (Syk), which initiates proinflammatory signaling cascades, including MAPK and activation. Siglec-15 exemplifies this activating mode, pairing with DAP12 to enhance Syk-dependent responses in macrophages and monocytes, such as increased TGF-β upon recognition. Siglec signaling often involves crosstalk with other immune receptors to fine-tune responses. For instance, CD22 interacts with β7 integrin on B cells, where SHP-1 recruitment inhibits integrin endocytosis and modulates cell homing. Similarly, Siglec-9 on macrophages attenuates Toll-like receptor (TLR) signaling by reducing LPS-induced MAPK phosphorylation, thereby limiting excessive inflammation. Cis-interactions with sialylated glycans on the same cell surface further modulate Siglec signaling by masking receptor availability and dampening trans-ligand responses. In B cells, cis-ligands bind to maintain it in a clustered, inactive state, requiring high-avidity trans-interactions to trigger inhibitory signaling and prevent . This mechanism ensures balanced immune thresholds across Siglec family members.

Phagocytosis and Cell Adhesion

Siglec-1, also known as sialoadhesin or CD169, is prominently expressed on macrophages and plays a key role in by binding to sialic acid-containing glycans on host cells and pathogens, facilitating tethering and initial capture. This interaction enables macrophages to adhere to sialylated surfaces, such as those on apoptotic cells or microbial pathogens like group B Streptococcus, promoting subsequent engulfment without triggering strong inflammatory responses. For instance, Siglec-1 enhances the and of sialylated particles in cooperation with other receptors like Fcγ receptors and TIM-4 on alveolar macrophages, thereby supporting efficient pathogen clearance in the lungs. In phagocytosis, specific Siglecs exhibit both facilitatory and inhibitory functions depending on the cellular context. On neutrophils, Siglec-5 and the paired activating receptor Siglec-14 interact with sialic acids on target cells, inhibiting activation (e.g., CD11b/CD18) and thereby suppressing neutrophil-mediated and of apoptotic or opsonized targets. This inhibitory mechanism limits excessive phagocytic activity, as demonstrated by enhanced upon blockade of Siglec-5/14 with sialidase treatment or antibodies, which significantly increases neutrophil-tumor cell conjugate formation in donor studies. Conversely, Siglec-E on murine macrophages and dendritic cells modulates apoptotic cell clearance by suppressing (ROS) production during engulfment, which prevents oxidative damage and promotes efficient while maintaining anti-inflammatory homeostasis; deficiency in Siglec-E leads to impaired ROS regulation and reduced clearance efficiency in models of neurodegeneration and . Counter-receptors such as and MUC1 serve as cis-ligands that modulate Siglec-mediated strength on the same cell surface, fine-tuning immune cell interactions. , a glycosylphosphatidylinositol-anchored protein rich in sialic acids, engages Siglec-10 in cis to dampen activation and enhance tolerance during , reducing pro-inflammatory release in response to damaged tissues. Similarly, MUC1, a with extensive O-linked sialylation, acts as a cis-ligand for Siglec-9 on myeloid cells, altering dynamics and promoting tumor-associated differentiation while inhibiting trans-interactions that could drive strong . Siglecs contribute to immune synapse formation by localizing to the contact interface between immune cells and targets, stabilizing and modulating activation thresholds. For example, inhibitory Siglecs like Siglec-G and (Siglec-2) are recruited to the B cell via sialylated ligands on antigen-presenting cells, inhibiting signaling and inducing BIM-dependent of self-reactive s, which is essential for as evidenced by reduced B cell depletion in double-knockout models. In natural killer cells, Siglec-7 similarly clusters at the with tumor targets, where cis-ligand masking limits activation, but trans-engagement with hypersialylated surfaces can suppress cytotoxic granule release, thereby regulating stability and preventing overactivation.

Insights from Knockout Studies

Knockout studies in mice have provided key insights into the roles of Siglecs, particularly in regulating immune and preventing pathological responses. In Siglec-1-deficient mice, macrophages exhibit reduced of sialylated pathogens, leading to impaired clearance of such as Campylobacter jejuni and group B Streptococcus, which highlights Siglec-1's function in facilitating uptake of sialylated microbes by myeloid cells. These mice show attenuated severity in experimental autoimmune encephalomyelitis (EAE), with increased regulatory T cells and reduced Th17 cells, suggesting Siglec-1 may promote pro-inflammatory responses in certain autoimmune contexts. CD22 knockout mice demonstrate hyperactive s with enhanced BCR signaling, characterized by increased calcium influx and proliferation in response to antigens, underscoring CD22's role as a negative regulator of activation. These mice develop elevated autoantibodies and lupus-like symptoms upon aging or in autoimmune-prone backgrounds, including high-affinity anti-DNA antibodies and immune complex-mediated , indicating CD22's essential function in maintaining tolerance and preventing systemic . Siglec-G-deficient mice, the murine ortholog of human Siglec-10, exhibit expanded populations with heightened responsiveness, elevated serum IgM levels, and increased B cells and cells in aging animals, reflecting Siglec-G's inhibitory control over innate-like B cell expansion and . These changes contribute to enhanced formation and potential amplification of antibody-mediated responses, though single knockouts do not typically develop spontaneous on standard backgrounds. Studies of double knockouts reveal functional redundancy among CD33rSiglecs in B cell regulation; CD22/Siglec-G double-deficient mice show massively expanded , reduced B2 cells, hyperproliferative responses to TLR ligands, and overt systemic with anti-nuclear and anti-DNA , far exceeding phenotypes in single knockouts. This redundancy emphasizes the compensatory inhibitory roles of and Siglec-G in suppressing aberrant B cell activation and production across the CD33r subfamily.

Siglecs Across Mammals

Human and Primate Siglecs

express 14 functional Siglecs, which are primarily expressed on cells of the and a few other types, such as neurons and glial cells. These include Siglec-1 (also known as sialoadhesin or CD169), which is predominantly found on macrophages; Siglec-2 (), restricted to B cells; and Siglec-3 (), expressed on myeloid progenitors, monocytes, macrophages, dendritic cells, mast cells, and . Other notable examples are Siglec-7 and Siglec-9, both present on natural killer (NK) cells, monocytes, macrophages, neutrophils, and subsets of T cells; Siglec-8, specific to , mast cells, and ; Siglec-10, on B cells and monocytes; Siglec-11, on macrophages and ; Siglec-14, on neutrophils, monocytes, and macrophages; Siglec-15, primarily on osteoclasts and some macrophages; and Siglec-16, on macrophages. Additionally, Siglec-4 (myelin-associated glycoprotein or MAG) is expressed on and Schwann cells in the , while Siglec-5 is found on neutrophils and monocytes, and Siglec-6 on placental trophoblasts and B cells. The Siglec family in , particularly great apes, exhibits rapid , with notable expansions and variations in the CD33-related (CD33r) subgroup, which includes Siglec-3, -5 through -11, and -14 through -16. This expansion is thought to reflect pathogen-driven selective pressures, leading to increased gene diversity in great apes compared to humans. A key human-specific change is the complete loss of Siglec-13 through an Alu-mediated deletion, rendering it absent in modern humans but functional in chimpanzees and baboons, where it is expressed on monocytes and epithelial cells. Furthermore, Siglec-12 exists as a in humans due to a abolishing binding, a feature fixed in both modern and but retained in great apes. Siglec-16 is primate-specific, while Siglec-11 shows human-specific upregulation in brain . These variations highlight the dynamic of Siglecs in , potentially influencing immune recognition and self-tolerance. Human Siglecs display preferences for specific linkages, primarily α2-3, α2-6, or α2-8, and vary in the number of extracellular immunoglobulin-like () domains, which contribute to ligand avidity. The following table summarizes these features for the 14 functional human Siglecs:
SiglecExtracellular Ig DomainsPreferred Sialic Acid Linkages
Siglec-117α2-3
Siglec-27α2-6
Siglec-32α2-3, α2-6
Siglec-45α2-3
Siglec-54α2-3, α2-6
Siglec-64α2-6, α2-3 (with )
Siglec-73α2-8, branched α2-6
Siglec-83α2-3 (with 6'-sulfate)
Siglec-93α2-3, α2-6, α2-8
Siglec-105α2-3, α2-6 (prefers α2-6)
Siglec-114α2-8
Siglec-143α2-3, α2-6 (similar to Siglec-5)
Siglec-152α2-3
Siglec-163α2-8 (similar to Siglec-11)
These binding preferences influence cellular interactions, with most Siglecs featuring an N-terminal V-set domain for sialic acid recognition.

Siglecs in Non-Primate Mammals

In non-primate mammals, the Siglec family exhibits notable diversity, particularly through expansions in the CD33-related (CD33r) subgroup, which contrasts with the more conserved repertoire in . Rodents, such as mice, possess nine Siglec genes in total, including five CD33r members: (CD33), Siglec-E, Siglec-F, Siglec-G, and Siglec-H. These expansions, with Siglec-E, -F, -G, and -H being unique to , likely arose from events that enhanced immune regulation in these species. This larger repertoire allows to fine-tune sialic acid-mediated interactions in innate immunity, making them valuable model organisms for studying Siglec functions despite differences from human orthologs. A prominent example of functional divergence is mouse Siglec-F, which serves as an inhibitory receptor on eosinophils, promoting their apoptosis upon ligand engagement and thereby dampening allergic inflammation. This parallels human Siglec-8, its closest functional analog, but Siglec-F lacks the exact sequence motifs for certain antibody-induced signaling pathways observed in humans, highlighting species-specific adaptations in eosinophil regulation. Similarly, mouse Siglec-E modulates macrophage and neutrophil responses to bacterial infections, suppressing excessive inflammation, while Siglec-G on B cells influences self-nonself discrimination in adaptive immunity. These rodent-specific Siglecs underscore the role of evolutionary expansions in tailoring immune homeostasis to environmental pressures like pathogens. In species, Siglec expression supports surveillance and immune modulation, with implications for veterinary health. In , Siglec-1 (sialoadhesin) is expressed on macrophages and dendritic cells, where it facilitates recognition and uptake of sialylated pathogens, such as viruses, enhancing in immunity. Bovine dendritic cells preferentially utilize Siglec-1 over Siglec-10 for binding enveloped viruses like , illustrating its role in bridging innate recognition to adaptive responses. Porcine CD33r Siglecs, including Siglec-3, Siglec-5, and Siglec-10, are expressed on myeloid cells and contribute to sialic acid-dependent interactions that influence viral tropism and immune evasion. These receptors can bind sialylated viral envelopes, potentially increasing susceptibility to like porcine reproductive and respiratory virus (PRRSV), where engagement modulates activation and viral entry. In pigs, variations in CD33r Siglec expression may underlie breed-specific differences in outcomes, informing strategies for health management. The diversity of Siglecs in non-primate mammals highlights their adaptability in veterinary , where models elucidate basic mechanisms, and studies reveal applications in disease resistance breeding and design. For instance, targeting Siglec-F in mice has informed eosinophil-targeted therapies, while bovine and porcine Siglecs offer insights into zoonotic pathogen control. This comparative perspective emphasizes Siglecs' conserved yet expanded roles across mammals, beyond primate-centric views.

Therapeutic Potential

Siglec Mimetics and Synthesis

mimetics (SAMs) are synthetic analogs of (Neu5Ac), the primary in mammalian glycans, engineered to bind Siglecs with greater potency and specificity than natural ligands. Key modifications target the side chain at the C6 position of Neu5Ac, often replacing it with aromatic groups such as benzyl or benzoyl moieties to enhance hydrophobic interactions within the Siglec V-set . These alterations exploit structural insights from Siglec-ligand complexes to optimize binding while maintaining the core scaffold responsible for recognition. The production of SAMs relies on two principal synthesis strategies: chemoenzymatic and . Chemoenzymatic approaches leverage sialyltransferases to incorporate modified onto acceptors, combining of the sialic acid analog with enzymatic for scalable assembly of multivalent ligands. involves iterative protection-deprotection cycles in chemistry to build the ring and substituents from non-carbohydrate starting materials, enabling precise control over and substitution patterns. Notable examples of SAMs include GT1b-lactone derivatives tailored for Siglec-2 (), which mimic polysialylated motifs to achieve selective B-cell targeting, and benzoyl-modified sialic acids optimized for Siglec-7 engagement on natural killer cells. These compounds demonstrate binding enhancements of up to 1000-fold over unmodified Neu5Ac, as measured by and cell-based assays, primarily due to stabilized interactions at the C9 position and multivalent presentation.

Clinical Applications and Targeting

Siglecs have emerged as promising therapeutic targets in various diseases due to their roles in modulating immune responses, with (Siglec-3) being the most clinically advanced example. In (AML), is overexpressed on leukemic blasts, making it a key target for antibody-drug conjugates. (GO), a -targeted immunoconjugate approved by the FDA in 2017 for CD33-positive AML, delivers the cytotoxic agent to CD33-expressing cells, leading to improved overall survival when combined with standard in newly diagnosed patients. Siglec-15 has been implicated in promoting , particularly in , where its elevated expression in the metastatic niche enhances tumor-induced osteoclastogenesis and suppresses T-cell activity. A 2024 study demonstrated that blocking Siglec-15 with a reduced osteoclast formation and tumor burden in preclinical models of , highlighting its potential as a glyco-immune checkpoint. Recent preclinical as of 2025 has explored Siglec-15 –GM-CSF chimeras, which suppress tumor progression in models of Siglec-15-overexpressing cancers by enhancing immune activation. Therapeutic strategies targeting Siglecs include blocking antibodies to inhibit immunosuppressive functions in cancer and agonists to dampen hyperinflammation in . For Siglec-3 in cancer, non-conjugate blocking antibodies have shown preclinical efficacy in disrupting CD33-mediated inhibition of immune cells, enhancing antitumor responses when combined with checkpoint inhibitors. In , Siglec-9 agonists, such as synthetic derivatives, inhibit activation and NETosis, reducing inflammatory damage in models of hyperinflammation akin to bacterial . Challenges in Siglec targeting arise from functional among family members and cis-inhibition, where Siglecs bind endogenous sialic acids on the same cell, limiting accessibility for therapeutics. To overcome these, bispecific antibodies that simultaneously engage multiple Siglecs or pair Siglec with immune activators have demonstrated superior in preclinical cancer models by bypassing redundancy and enhancing immune engagement. Emerging applications focus on Siglec-8 for disorders, where antagonists like lirentelimab () induce and reduce tissue infiltration.

References

  1. [1]
    Discovery, Classification, Evolution and Diversity of Siglecs - PMC
    Siglecs are a family of glycan-recognizing proteins belonging to the Ig superfamily (ie, I-type lectins). Siglecs are expressed on various leukocyte types.
  2. [2]
    Siglecs and their roles in the immune system - Nature
    Siglecs are sialic-acid-binding immunoglobulin-like lectins that are mostly expressed by cells of the immune system. Siglecs can be divided into two subsets ...
  3. [3]
    Sialic acid–binding immunoglobulin-like lectins (Siglecs) detect self ...
    Sia-binding immunoglobulin-like lectins (Siglecs) are a large family of I-type lectins, immune-modulatory receptors within the mammalian immune system with a ...
  4. [4]
    The B-cell antigen CD22 mediates monocyte and erythrocyte ...
    May 3, 1990 · The B-cell antigen CD22 mediates monocyte and erythrocyte adhesion. Ivan Stamenkovic &; Brian Seed. Nature volume 345, pages 74 ...
  5. [5]
    The B-cell antigen CD22 mediates monocyte and erythrocyte ...
    Here we show that CD22 has a structure closely related to myelin-associated glycoprotein (MAG, a neuronal adhesion protein), and mediates ...
  6. [6]
    Siglecs: A family of sialic-acid binding lectins - Oxford Academic
    Siglecs are a subfamily of sialic-acid binding cell surface receptors, a subset of I-type lectins, that bind sialylated glycans.
  7. [7]
    Evolution of CD33-related siglecs: regulating host immune functions ...
    An ancestral siglec-11 gene existed before rodents and primates. This gene underwent an inverse duplication in early primates creating two inhibitory siglec-11- ...
  8. [8]
    Siglec receptors impact mammalian lifespan by modulating ... - eLife
    Apr 7, 2015 · Our data suggest that CD33rSiglecs co-evolved in mammals to achieve a better management of oxidative stress during inflammation, which in turn reduces ...Missing: ancestral | Show results with:ancestral<|separator|>
  9. [9]
  10. [10]
  11. [11]
    Siglec Ligands - PMC - PubMed Central - NIH
    May 20, 2021 · Humans express 14 Siglecs, of which 13 are found on overlapping cell types of the immune system and one (Siglec-4) on myelinating cells in the ...
  12. [12]
    A Second Uniquely Human Mutation Affecting Sialic Acid Biology
    In particular, all Siglec V-set domains have a conserved arginine residue that forms a salt bridge with the carboxylate group of sialic acids. Experimental ...
  13. [13]
    Siglecs—the major subfamily of I-type lectins - Oxford Academic
    S. , Hartnell, A., Mattei, M.-G., Gordon, S., and Crocker, P.R. (. 1995. ) Sialoadhesin (Sn) maps to mouse chromosome 2 and human chromosome 20 and is not ...
  14. [14]
    New Aspects of Siglec Binding Specificities, Including the ...
    Siglec-1 is reported to prefer α2–3-linked sialyllactos(amin)e over α2–6-linked sialyllactos(amin)e and showed some binding to α2–8-linked sialic acids on ...
  15. [15]
    Targeting Siglec–Sialylated MUC1 Immune Axis in Cancer - PMC
    Mar 29, 2024 · Most Siglecs have a unique specificity profile for a particular sialic acid linkage. Siglec-2 has a clear preference for α2,6-linkages; Siglec-1 ...
  16. [16]
    Modulation of Immune Tolerance via Siglec-Sialic Acid Interactions
    We highlight how the sialic acid-Siglec axis can be utilized for clinical applications to induce or inhibit immune tolerance.
  17. [17]
    Interactions between Siglec-8 and endogenous sialylated cis ...
    Oct 19, 2023 · Cis ligands may hinder Siglecs from interacting with trans ligands and thereby create an affinity/avidity threshold for such interactions (“ ...
  18. [18]
    Roles for Siglec-glycan interactions in regulating immune cells
    The architecture of Siglecs. Siglecs are found in all jawed vertebrates and, from an evolutionary perspective, are divided into two groups: the conserved ...
  19. [19]
    Modulation of immune cell reactivity with cis-binding Siglec agonists
    High-affinity Siglec ligands achieved by derivatizing sialic acids at the C5 and/or C9 positions have been previously reported (26, 37–42). These ligands have ...<|control11|><|separator|>
  20. [20]
    High resolution crystal structures of Siglec-7. Insights into ligand ...
    In the Siglec-7 structure, the ligand-binding pocket is occupied by a loop of a symmetry-related molecule, mimicking the interactions with sialic acid.
  21. [21]
    The structure of siglec-7 in complex with sialosides - NIH
    Derivatization of Neu5Ac at the C9 position by hydrophobic moieties has yielded increased binding to other siglecs, namely human and murine CD22 and CD33, and ...
  22. [22]
    Discovery of a new sialic acid binding region that regulates Siglec-7
    May 26, 2020 · Siglec-7 has a preferred binding specificity towards the Neu5Acα2,8Neu5Acα- (diSia) sequence found in certain gangliosides such as the ...
  23. [23]
  24. [24]
    Large-scale sequencing of the CD33-related Siglec gene cluster in ...
    These data indicate that CD33rSiglec genes are undergoing rapid evolution via multiple genetic mechanisms, possibly due to an evolutionary “arms race ...
  25. [25]
    Siglec Signaling in the Tumor Microenvironment - Frontiers
    Siglecs bind their ligand using the V-set sialic acid-binding Ig domain, which is positioned away from the membrane by a variable number of C2-set Ig domains.
  26. [26]
    SIGLEC16 encodes a DAP12-associated receptor ... - PubMed
    SIGLEC16 encodes a DAP12-associated receptor expressed in macrophages that evolved from its inhibitory counterpart SIGLEC11 and has functional and non- ...
  27. [27]
    Sialoadhesin (CD169/Siglec-1) is an extended molecule that ... - NIH
    Feb 4, 2023 · Sialoadhesin (CD169/Siglec-1, Sn) is a macrophage receptor that interacts with sialic acids on both host cells and pathogens.
  28. [28]
    Siglecs as modulators of macrophage phenotype and function
    Some Siglecs form a multimeric complex with either DAP10 or DAP12 dimers, thereby providing ITAM-mediated signaling downstream of Siglec ligation. (Right): ...
  29. [29]
    SIGLEC-5/14 Inhibits CD11b/CD18 Integrin Activation and ... - PMC
    Dec 5, 2023 · The interaction between SIGLEC-5/14 and the sialic acids was shown to inhibit the CD11b/CD18-regulated binding between neutrophils and antibody- ...
  30. [30]
    Siglecs Modulate Activities of Immune Cells Through Positive and ...
    Nov 2, 2021 · Siglecs modulate immune activation and can promote or inhibit ROS generation under different contexts. Siglecs promote ROS-dependent cell death ...
  31. [31]
    CD24-Siglec interactions in inflammatory diseases - Frontiers
    May 8, 2023 · Nearly 15 years ago, CD24 was shown to interact with Siglec G/10 to selectively inhibit inflammatory response to tissue injuries. Subsequent ...Missing: MUC1 | Show results with:MUC1
  32. [32]
    The intriguing roles of Siglec family members in the tumor ...
    Apr 13, 2022 · Recent studies have shown that Siglecs could play diverse immune and nonimmune regulatory roles in the tumor microenvironment (TME) and participate in tumor ...
  33. [33]
    Siglecs induce tolerance to cell surface antigens by BIM-dependent ...
    These results demonstrate that the siglecs are recruited to an immunological synapse through interactions with sialoside ligands on the antigen-expressing cell.
  34. [34]
    Siglec-7 represents a glyco-immune checkpoint for non-exhausted ...
    The formation of an immune synapse (IS) involves the spatio-temporal organization of molecular events at the interface between effector and target cells.
  35. [35]
    Impact of Siglecs on autoimmune diseases - PMC - PubMed Central
    In this review, we highlight potential of Siglecs in suppressing immune responses causing autoimmune diseases.
  36. [36]
    Deficiency in CD22, a B Cell–specific Inhibitory Receptor, Is ...
    The results demonstrate that a single gene defect exclusive to B lymphocytes is, without additional contrivance, sufficient to trigger autoantibody development.Missing: hyperactivation | Show results with:hyperactivation
  37. [37]
    Siglec-G Deficiency Leads to Autoimmunity in Aging C57BL/6 Mice
    Jul 1, 2015 · We find that aging Siglec-G-deficient and Siglec-G × FcγRIIb double-deficient mice develop an autoimmune phenotype with elevated autoantibody levels and mild ...
  38. [38]
    CD22 x Siglec-G double-deficient mice have massively ... - PubMed
    Apr 1, 2010 · These results show that Siglec-G and CD22 have partly compensatory functions and together are crucial in maintaining the B cell tolerance.
  39. [39]
    Basic and Clinical Immunology of Siglecs - PMC - PubMed Central
    Siglecs characteristically contain an N-terminal V-set (or variable set) domain for binding of sialic acid–containing glycan ligands, and within this domain is ...
  40. [40]
    Rapid evolution of binding specificities and expression patterns of ...
    We hypothesized that these opposing selection forces have accelerated CD33rSiglec evolution. We address this by comparative analysis of major CD33rSiglec ( ...Missing: CD33r | Show results with:CD33r
  41. [41]
    Probing the binding specificities of human Siglecs by cell-based ...
    Apr 23, 2021 · Insight into the fine binding specificities of Siglecs will advance understanding their diverse biological functions and benefit therapeutic ...
  42. [42]
    Siglec - an overview | ScienceDirect Topics
    The human genome encodes up to 15 Siglecs (Siglec-13 is absent in humans), whereas the mouse genome contains nine Siglecs.
  43. [43]
    Defining the in vivo function of Siglec-F, a CD33 ... - ASH Publications
    Feb 1, 2007 · We studied in vivo functions of Siglec-F, the CD33rSiglec expressed on mouse eosinophils, which are prominent in allergic processes. Induction ...Eosinophil Siglec-F... · Siglec-F Expression Is... · Siglec-F Mice Show Enhanced...
  44. [44]
    Siglec-8 Signals Through a Non-Canonical Pathway to Cause ...
    Oct 10, 2021 · These results show that Siglec-8 signals through an unanticipated set of signaling molecules in IL-5–primed eosinophils to induce cell death.
  45. [45]
    Siglec-G/10 in self–nonself discrimination of innate and adaptive ...
    The Siglecg gene is found in a cluster with genes of the related proteins Siglec-E, CD33 and Siglec-F on mouse chromosome 7 (Angata et al. 2001).
  46. [46]
    Preferential use of Siglec-1 or Siglec-10 by type 1 and type 2 ...
    Jul 18, 2018 · Siglec-1 is expressed to higher levels on the surface of the cells than Siglec-10. This might partly explain the more efficient binding and ...
  47. [47]
    Molecular cloning of porcine Siglec-3, Siglec-5 and Siglec-10 ... - NIH
    In humans, CD33-related Siglecs interact with several sialylated pathogens, such as Campylobacter jejuni, Neisseria meningitides, group B streptococcus, ...Missing: susceptibility | Show results with:susceptibility
  48. [48]
    An Intact Sialoadhesin (Sn/SIGLEC1/CD169) Is Not Required ... - NIH
    Surface expression of SIGLEC1, also known as sialoadhesin or CD169, is considered a primary determinant of the permissiveness of porcine alveolar macrophages ...Creation Of Siglec1 Knockout... · Expression Of Cd169... · Prrsv Viremia And Antibody
  49. [49]
  50. [50]
    Gemtuzumab ozogamicin for acute myeloid leukemia | Blood
    Nov 30, 2017 · Clinical studies demonstrate as much as a 2 log-fold variation in CD33 expression by AML blasts with lower levels of CD33 seen in AML cases with ...
  51. [51]
    Gemtuzumab ozogamicin for treatment of newly diagnosed CD33 ...
    Gemtuzumab ozogamicin (GO), a CD33-directed monoclonal antibody linked to cytotoxic calicheamicin is a re-emerging agent with great potential in AML treatment.
  52. [52]
    Siglec-15/sialic acid axis as a central glyco-immune checkpoint in ...
    Jan 22, 2024 · One prominent Siglec, Siglec-2 (CD22), is involved in cell adhesion and negatively regulates the B cell receptor (20–27). Due to its exclusive ...
  53. [53]
    Therapeutic Targeting of Siglecs using Antibody- and Glycan-based ...
    Antibodies that target Siglecs can deplete the Siglec-expressing cells via recruitment of effector cells of the immune system, delivery of a drug or toxin, or ...<|control11|><|separator|>
  54. [54]
    Synthetic Siglec-9 Agonists Inhibit Neutrophil Activation Associated ...
    Thus, Siglec-9 is a therapeutic target to inhibit potentially fatal hyperinflammation associated with COVID-19 in an analogous fashion to the highly effective ...Introduction · Results and Discussion · Supporting Information · References
  55. [55]
    Sialic acid and Siglec receptors in tumor immunity and immunotherapy
    Notably, treatment with blocking antibodies targeting Siglec-7 and Siglec-9 effectively reversed the tumor growth mediated by these receptors [66].
  56. [56]
    Anti–Siglec-8 Antibody for Eosinophilic Gastritis and Duodenitis
    Oct 21, 2020 · The aim of our trial (ENIGMA) was to assess the efficacy and safety of AK002 in adult patients with eosinophilic gastritis, eosinophilic ...Missing: antagonists II
  57. [57]
    365 Phase 2 trial in progress—lirentelimab in adults with moderate ...
    Jun 20, 2023 · Phase 2 proof-of-concept, randomized, double-blind, placebo-controlled study of lirentelimab SC in adults with moderate-to-severe AD inadequately controlled by ...