CD22 is a 140 kDa type I transmembrane protein belonging to the SIGLEC family. Its structure comprises:
Extracellular domain: Seven immunoglobulin (Ig) domains (V-set and C2-set) with a ligand-binding V-like domain at the N-terminus that recognizes α2,6-linked sialic acid residues .
Transmembrane domain: Single-spanning helix anchoring the protein to the cell membrane .
Cytoplasmic domain: Contains six tyrosine residues with immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and immunoreceptor tyrosine-based activation motifs (ITAMs) .
Therapy | Target Population | Response Rate | Reference |
---|---|---|---|
Epratuzumab | SLE, NHL | 30–40% | |
CD22-CAR FH80 | Pediatric B-ALL | 75% CR | |
Inotuzumab | Relapsed B-ALL | 81% ORR |
Recombinant CD22 proteins are critical for structural and functional studies:
Recent advances in CD22 biology include:
Structural insights: Cryo-EM reveals an extended ectodomain conformation enabling nanocluster formation and ligand avidity .
Migration defects: CD22−/− B cells show impaired homing to bone marrow and lymph nodes .
Autoantibody escape: CD22lo B cells evade peripheral tolerance checkpoints, contributing to SLE .
CD22, also called Sialic Acid-Binding Ig-Like Lectin 2, belongs to the immunoglobulin (Ig) superfamily. This protein is involved in B-cell to B-cell interactions and plays a role in positioning B-cells within lymphoid tissues. CD22 can act as a positive regulator through interactions with Src family tyrosine kinases and can also function as an inhibitory receptor by recruiting cytoplasmic phosphatases.
Produced in Sf9 Baculovirus cells, CD22 is a single, glycosylated polypeptide chain consisting of 907 amino acids (20-687a.a.). It has a molecular weight of 102.1 kDa. Note: On SDS-PAGE, the molecular size appears to be between 100-150 kDa.
This CD22 protein is expressed with a C-terminal 239 amino acid hIgG-His-tag and is purified using proprietary chromatographic methods.
This CD22 protein solution has a concentration of 0.25 mg/ml and contains 10% glycerol and Phosphate Buffered Saline (pH 7.4).
Purity is greater than 85.0% as determined by SDS-PAGE analysis.
CD22 Molecule, CD22 Antigen, Sialic Acid-Binding Ig-Like Lectin 2, B-Lymphocyte Cell Adhesion Molecule, T-Cell Surface Antigen Leu-14, SIGLEC-2, SIGLEC2, BL-CAM, Sialic Acid Binding Ig-Like Lectin 2, B-Cell Receptor CD22.
Sf9, Baculovirus cells.
DSSKWVFEHP ETLYAWEGAC VWIPCTYRAL DGDLESFILF HNPEYNKNTS KFDGTRLYES TKDGKVPSEQ KRVQFLGDKN KNCTLSIHPV HLNDSGQLGL RMESKTEKWM ERIHLNVSER PFPPHIQLPP EIQESQEVTL TCLLNFSCYG YPIQLQWLLE GVPMRQAAVT STSLTIKSVF TRSELKFSPQ WSHHGKIVTC QLQDADGKFL SNDTVQLNVK HTPKLEIKVT PSDAIVREGD SVTMTCEVSS SNPEYTTVSW LKDGTSLKKQ NTFTLNLREV TKDQSGKYCC QVSNDVGPGR SEEVFLQVQY APEPSTVQIL HSPAVEGSQV EFLCMSLANP LPTNYTWYHN GKEMQGRTEE KVHIPKILPW HAGTYSCVAE NILGTGQRGP GAELDVQYPP KKVTTVIQNP MPIREGDTVT LSCNYNSSNP SVTRYEWKPH GAWEEPSLGV LKIQNVGWDN TTIACAACNS WCSWASPVAL NVQYAPRDVR VRKIKPLSEI HSGNSVSLQC DFSSSHPKEV QFFWEKNGRL LGKESQLNFD SISPEDAGSY SCWVNNSIGQ TASKAWTLEV LYAPRRLRVS MSPGDQVMEG KSATLTCESD ANPPVSHYTW FDWNNQSLPY HSQKLRLEPV KVQHSGAYWC QGTNSVGKGR SPLSTLTVYY SPETIGRRLE PKSCDKTHTC PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN AKTKPREEQY NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP QVYTLPPSRD ELTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG KHHHHHH.
CD22 is a sialic acid-binding immunoglobulin type-lectin (Siglec) family member that functions as an inhibitory co-receptor of the B-cell receptor (BCR). It plays crucial roles in regulating B-cell activation, signaling, antibody responses, and cellular homing. The significance of CD22 lies in its restricted expression pattern on B-cells and most B-cell lymphomas, making it an important therapeutic target for B-cell-mediated diseases . CD22's inhibitory function is essential for maintaining B-cell homeostasis and preventing excessive immune responses. Understanding CD22's functions is critical for developing new therapeutic approaches for conditions such as B-cell lymphomas, autoimmune disorders, and other B-cell related pathologies, particularly as CD22-targeted therapies continue to emerge in clinical settings.
Human CD22 (hCD22) and murine CD22 (mCD22) share approximately 60% homology in amino acid sequence, with higher conservation in functionally critical regions. Specifically, greater homology is observed within the N-terminal ligand binding domain and the C-terminal cytoplasmic tail, both essential for BCR signaling regulation . Despite this conservation, there are subtle but important differences in their glycan ligand preferences . These differences have led researchers to develop ligands with differential affinity and selectivity for mCD22 and hCD22. Understanding these species-specific differences is crucial when translating findings from mouse models to human applications, particularly in the development of therapeutics targeting CD22.
CD22 contains several functional domains that work together to regulate B-cell receptor signaling. The extracellular portion includes an N-terminal domain responsible for sialic acid binding and multiple Ig-like domains . The N-terminal domain recognizes specific sialic acid-containing glycan ligands. The protein also contains a transmembrane domain and a cytoplasmic tail with multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs) . Upon BCR activation, these ITIMs become phosphorylated by Src kinases like Lyn, creating docking sites for phosphatases such as SHP-1. These recruited phosphatases then attenuate BCR signaling, effectively dampening B-cell activation. For experimental purposes, researchers can isolate and express various domains separately to study their specific functions in receptor-ligand interactions and signaling pathways .
Mouse Model | Construction Method | Expression Level | Functions Restored | Limitations |
---|---|---|---|---|
Human knock-in (Huki) hCD22 | Replacement of mouse CD22 gene with human CD22 cDNA | 10-30× lower than human B-cells | Antibody responses, endocytotic properties | Failed to restore B-cell maturation, homing to BM, BCR signaling regulation |
MB1-Cre driven hCD22 transgenic | B-cell specific expression using MB1-Cre | Comparable to human primary B-cells | Ca²⁺ flux responses, antibody responses, partial homing restoration, tolerance induction | Partial rather than complete restoration of homing to Peyer's patches |
CD22-deficient (CD22⁻/⁻) | Knockout of CD22 gene | No CD22 expression | N/A (negative control) | Impaired homing, altered antibody responses, enhanced BCR signaling |
For CD22 protein isolation and purification, researchers can use several approaches depending on their experimental needs. Soluble CD22 can be purchased commercially, but researchers frequently generate their own constructs . One common method involves expressing CD22-Fc fusion proteins, where the extracellular domain of CD22 is fused to human IgG1 Fc . This construct can be transfected into mammalian cell lines like 293T cells using lipid-based transfection reagents. The secreted fusion protein can then be purified from cell culture supernatant using protein A columns . For studying specific domains of CD22, researchers can generate constructs encoding individual domains with appropriate tags (such as His or c-Myc), express them in mammalian cells, and purify them using affinity chromatography . These purified proteins can then be used for various applications including antibody selection, binding studies, and structural analyses.
Studying CD22 localization and interactions requires multiple complementary techniques. Confocal microscopy with fluorescently labeled antibodies is effective for visualizing CD22 distribution on the cell surface and its co-localization with other proteins like the BCR . Quantitative co-localization analysis using metrics such as Manders' overlap coefficient (MOC) provides statistical measures of protein interaction . Flow cytometry allows for quantification of CD22 expression levels and can be combined with functional assays. For protein-protein interactions, co-immunoprecipitation followed by Western blotting or mass spectrometry can identify binding partners. To study CD22's interactions with sialic acid ligands, glycan arrays and surface plasmon resonance offer valuable insights into binding specificities and affinities. For functional interactions, calcium flux assays following BCR stimulation in the presence or absence of CD22 ligands or antibodies can reveal CD22's regulatory role in B-cell signaling .
CD22 regulates BCR signaling through a tyrosine phosphorylation-dependent inhibitory mechanism. When B-cells are activated through the BCR, CD22 must be in close proximity to become phosphorylated by Src family kinases, particularly Lyn . This phosphorylation occurs within multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in CD22's cytoplasmic tail. These phosphorylated motifs then serve as docking sites for Src homology 2 (SH2) domain-containing proteins, most importantly the phosphatase SHP-1 . When recruited to the BCR complex through CD22, SHP-1 dephosphorylates various components of the BCR signaling cascade, effectively dampening the signal . This negative regulation helps prevent excessive B-cell activation and plays a critical role in B-cell tolerance and prevention of autoimmunity. The regulatory function can be assessed experimentally by measuring calcium flux responses following BCR stimulation, with CD22-deficient B-cells typically showing enhanced calcium responses compared to wild-type cells .
CD22 plays a significant role in B-cell homing, particularly to gut-associated lymphoid tissues. Studies have shown that CD22-deficient B-cells have markedly impaired homing to Peyer's patches (PPs), moderately impaired homing to mesenteric lymph nodes, but normal homing to peripheral lymph nodes and spleen in short-term homing assays . The mechanism involves CD22's ability to bind sialic acid-containing glycan ligands on high endothelial venules (HEVs) in these specific locations . To study this phenomenon, researchers use complex mixing and transfer experiments where B-cells from different genotypes (e.g., wild-type, CD22-deficient, and hCD22-expressing) are labeled with different fluorescent dyes, mixed at equal ratios, transferred into recipient mice, and their relative distribution to various lymphoid organs is assessed by flow cytometry . Interestingly, transgenic expression of human CD22 in mice partially restores homing to Peyer's patches compared to CD22-deficient mice, but not to wild-type levels, suggesting potential differences in recognition of homing-related glycan ligands between human and mouse CD22 .
Lymphoid Tissue | WT B-cell Homing | CD22⁻/⁻ B-cell Homing | hCD22⁺ B-cell Homing | Mechanism |
---|---|---|---|---|
Peyer's patches | Normal (100%) | Markedly impaired | Partially restored | CD22-sialic acid ligand interactions on specialized HEVs |
Mesenteric lymph nodes | Normal (100%) | Moderately impaired | Partially restored | Similar to Peyer's patches but less pronounced |
Peripheral lymph nodes | Normal (100%) | Normal | Normal | CD22-independent homing mechanisms |
Spleen | Normal (100%) | Normal | Normal | CD22-independent homing mechanisms |
Development of anti-CD22 antibodies for therapeutic applications involves multiple sophisticated approaches. Researchers have utilized phage display technology to identify fully human anti-CD22 antibodies from naive human Fab phage libraries, avoiding the immunogenicity issues associated with murine or chimeric antibodies . These antibodies undergo rigorous characterization for specificity, affinity, epitope mapping, and functional effects on B-cells . Antibodies targeting different epitopes of CD22 can have varying effects on receptor internalization, signaling, and cell killing. For therapeutic applications, these antibodies can be used alone, conjugated to toxins (antibody-drug conjugates), or combined with other therapies . The optimization process involves engineering for enhanced binding affinity, improved pharmacokinetics, and reduced immunogenicity. Transgenic mouse models expressing human CD22 provide critical platforms for preclinical testing of these antibodies, enabling assessment of mechanisms of action and efficacy before moving to human trials .
CD22-targeted therapies show significant potential for treating both B-cell malignancies and autoimmune diseases due to CD22's restricted expression on B-cells and its role in B-cell function . For B-cell malignancies such as acute lymphoblastic leukemia, follicular lymphoma, and diffuse large B-cell lymphoma, anti-CD22 antibodies have been tested in clinical trials with promising results . These approaches include naked antibodies, antibody-drug conjugates, and bispecific antibodies. In autoimmune diseases like systemic lupus erythematosus (SLE), targeting CD22 offers a potentially more selective approach than current B-cell depletion therapies, as it could modulate rather than completely eliminate B-cell responses . The mechanism of action involves either depleting B-cells, delivering cytotoxic payloads specifically to B-cells, or modulating B-cell responses through CD22's inhibitory signaling. Transgenic mouse models expressing human CD22 provide essential tools for developing and testing these therapeutic approaches, helping bridge the gap between basic research and clinical applications .
CD22's natural role as an inhibitory co-receptor makes it an attractive target for inducing antigen-specific tolerance in autoimmune diseases and allergies . By exploiting CD22's inhibitory function, researchers have developed Siglec-engaging tolerance-inducing antigenic liposomes (STALs) that co-display both antigen and CD22 ligands . When B-cells specific for the antigen bind these liposomes, simultaneous engagement of CD22 delivers inhibitory signals that prevent activation and promote apoptosis . This approach allows for targeted elimination or anergy of antigen-specific B-cells without affecting the broader B-cell repertoire . The methodology has been validated in transgenic mouse models for both autoimmune and allergic conditions, including peanut allergy oral sensitization models . Importantly, human CD22 transgenic mice enable testing of human-specific CD22 ligands in vivo, facilitating translation to human applications . This targeted approach represents a significant advancement over current non-specific immunosuppressive therapies.
Anti-CD22 antibodies targeting different epitopes can induce markedly different biological effects through diverse molecular mechanisms . Some antibodies may block interaction with natural ligands, while others may induce receptor clustering or internalization. The epitope location relative to functional domains of CD22 significantly impacts outcomes. Antibodies binding near the ligand-binding domain may interfere with CD22's interaction with sialic acid ligands, potentially enhancing BCR signaling by preventing CD22's inhibitory function . Conversely, antibodies that crosslink CD22 may enhance its inhibitory function or trigger internalization. The internalization property is particularly important for antibody-drug conjugates, where efficient uptake is crucial for delivering cytotoxic payloads . Research addressing these questions requires advanced techniques including epitope mapping, structural studies, internalization assays, signaling studies, and functional B-cell assays. Understanding these mechanisms is essential for optimizing therapeutic antibodies and predicting their efficacy in different clinical contexts.
CD22 functions within a complex network of inhibitory and activating receptors that collectively regulate B-cell homeostasis . Advanced research questions in this area focus on how CD22 cooperates or competes with other inhibitory receptors like FcγRIIB, CD72, PD-1, and other Siglec family members. These receptors may have additive, synergistic, or redundant effects depending on the context of B-cell activation . The proximity of CD22 to the BCR is required for CD22 to become phosphorylated through the actions of the Src kinase Lyn, and this phosphorylation is crucial for recruiting phosphatases like SHP-1 that inhibit BCR signaling . Investigating these interactions requires sophisticated approaches including multi-parameter flow cytometry, CRISPR-mediated gene editing to create double or triple knockout models, phospho-proteomics to map signaling networks, and in vivo models of immune challenges. Transgenic mice expressing human CD22 in combination with other human inhibitory receptors would provide valuable models for studying these complex interactions in a humanized context .
CD22 is composed of an extracellular domain, a single transmembrane domain, and a cytoplasmic tail. The extracellular domain contains seven immunoglobulin-like domains, which are responsible for its binding to sialic acid-containing ligands . The cytoplasmic tail contains multiple tyrosine-based motifs that are involved in signal transduction .
CD22 is primarily expressed on the surface of mature B cells and is involved in modulating B-cell receptor (BCR) signaling. It acts as an inhibitory co-receptor that dampens BCR signaling, thereby preventing overactivation of B cells and maintaining immune homeostasis . Additionally, CD22 plays a role in B-cell adhesion and migration by interacting with other cell surface molecules .
Recombinant CD22 is produced using various expression systems, including mammalian cells, to ensure proper folding and post-translational modifications. The recombinant protein is often tagged with a polyhistidine tag (His tag) to facilitate purification and detection . It is commonly used in research to study B-cell biology, develop therapeutic antibodies, and investigate the role of CD22 in B-cell malignancies .
CD22 has emerged as an important target for monoclonal antibody (mAb)-based therapies for B-cell malignancies, including lymphomas and leukemias . Therapeutic antibodies targeting CD22 can induce B-cell depletion, thereby reducing the tumor burden and improving patient outcomes. Additionally, CD22-targeted therapies are being explored in the treatment of autoimmune diseases, where B-cell dysregulation plays a key role .
Recombinant CD22 is also used in various assays to study its binding interactions, signaling pathways, and functional roles in B cells. For example, it can be used in enzyme-linked immunosorbent assays (ELISAs) to measure the binding affinity of antibodies or other ligands to CD22 . Furthermore, recombinant CD22 is utilized in flow cytometry and immunohistochemistry to detect and quantify CD22 expression on B cells .