Fact-checked by Grok 2 weeks ago

CD33

CD33, also known as -3 or sialic acid-binding immunoglobulin-like lectin 3, is a transmembrane that serves as an inhibitory receptor primarily expressed on cells of the myeloid lineage, including monocytes, macrophages, granulocytes, and myeloid precursors. As a member of the family within the , CD33 recognizes sialic acid-containing glycans on cell surfaces and components, thereby mediating cell-cell interactions and modulating immune signaling through recruitment of tyrosine phosphatases such as SHP-1 and SHP-2 via its cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Structurally, CD33 consists of an extracellular region with one V-set immunoglobulin-like domain followed by a C2-set domain, a single transmembrane helix, and a short cytoplasmic tail containing two ITIMs that enable negative regulation of cellular activation. The protein is encoded by the CD33 gene located on chromosome 19q13.41 in humans, producing multiple isoforms through alternative splicing, with the full-length form featuring the sialic acid-binding V-set domain essential for ligand interaction. Expression of CD33 is broad but predominantly high in hematopoietic tissues, with the highest levels observed in spleen (RPKM 7.5) and bone marrow (RPKM 6.6), reflecting its role in myeloid differentiation and immune homeostasis. It localizes to the external side of the plasma membrane, Golgi apparatus, and peroxisomes, where it influences processes like endocytosis and sialic acid-dependent adhesion. Biologically, CD33 functions to dampen innate immune responses by inhibiting production, activation, and immune signaling pathways, thereby preventing excessive and promoting . It binds sialic acids in cis (on the same cell) or trans (on other cells), which can mask its activity or facilitate inhibitory signaling, and it enables activation to regulate downstream pathways like those involving suppressor of cytokine signaling 3 (SOCS3). In normal physiology, CD33 contributes to maintaining myeloid cells in a resting state and modulating their proliferation and survival during immune challenges. CD33 holds significant clinical relevance as a therapeutic target in (AML), where it is expressed on 85-90% of leukemic blasts and some leukemia stem cells, but absent from normal hematopoietic stem cells, allowing selective targeting. The antibody-drug conjugate (GO), first approved in 2000 but withdrawn in 2010 and re-approved in 2017, delivers the cytotoxic agent to CD33-positive cells and is used as a CD33-directed therapy for AML, demonstrating improved survival in favorable- and intermediate-risk patients when combined with , though resistance mechanisms like drug efflux and isoform variations can limit efficacy (as of 2025). Additionally, genetic variants in CD33, such as the rs3865444 polymorphism, are associated with increased risk of late-onset by enhancing microglial expression of CD33, which inhibits amyloid-β clearance and promotes plaque accumulation in the . As of 2025, emerging CD33-targeted immunotherapies, including CAR-NK cells and bispecific ADCs, are in clinical development for AML, alongside efforts to target CD33 in .

Gene and Expression

Genomic Location and Variants

The CD33 gene is located on the long (q) arm of human at cytogenetic band 19q13.41. The spans 28,941 bp (approximately 29 ), from 51,211,076 to 51,240,016 on the forward strand, and comprises 7 exons that encode the protein's functional domains. This genomic organization supports the production of a primary transcript that undergoes post-transcriptional processing to generate mature mRNA species. Alternative splicing of the CD33 pre-mRNA primarily involves exon 2, which encodes the extracellular immunoglobulin V-set (IgV) domain responsible for recognition. Inclusion of exon 2 yields the full-length isoform CD33M, a with sialic acid-binding capability, whereas exclusion of exon 2 produces the truncated isoform CD33m, which lacks the IgV domain and is predominantly intracellular. These isoforms arise from tissue-specific splicing regulation, with CD33m being more prevalent in certain myeloid contexts, and their ratio influences overall CD33 function in immune modulation. Several single nucleotide polymorphisms () in CD33 have functional consequences on transcript processing and protein expression. The intronic rs12459419 (C>T), located near the 3' splice site of 2, promotes 2 skipping in the T , thereby increasing the proportion of CD33m relative to CD33M and reducing cell-surface CD33 levels. Recent studies (as of November 2025) have further detailed the impact of rs12459419 on microglial splicing and expression. The (MAF) of rs12459419 T is approximately 30-32% in European-ancestry populations and 15-44% across global superpopulations. Another key variant, rs2455069 (A>G) in 3, introduces a missense change (Arg69Gly) that alters the protein's structural conformation, potentially affecting interactions and overall expression; the G has a frequency of about 47% in cohorts and is linked to modulated CD33 levels. The CD33 gene exhibits strong evolutionary conservation across mammals, with orthologs present in primates like chimpanzees and rodents like mice, reflecting its ancient role in innate immunity. However, the alternative splicing mechanism generating the CD33m isoform is largely human-specific, as non-human primates predominantly express the full-length CD33M form, highlighting adaptive changes in human microglial regulation.

Tissue and Cellular Expression

CD33 is a sialic acid-binding immunoglobulin-like lectin (Siglec) primarily expressed on cells of the myeloid lineage, serving as a key marker for immature myeloid cells such as hematopoietic progenitors, monocytes, macrophages, dendritic cells, and granulocytes. Expression is notably high on multipotent myeloid precursors and unipotent colony-forming cells in the bone marrow, with detection on normal myeloid progenitors and myelocytes. In contrast, CD33 shows low or absent expression on mature lymphocytes, erythrocytes, platelets, and non-hematopoietic tissues, highlighting its specificity to the myeloid compartment. During myeloid cell development, CD33 expression is dynamically regulated, exhibiting high levels on early multi-lineage hematopoietic progenitors such as , CFU-GM, CFU-G, and E-BFU in the , and progressively downregulated as cells mature into peripheral granulocytes and tissue macrophages. This maturation-associated decrease results in low-level surface expression on mature granulocytes, while monocytes and macrophages retain moderate levels. Gene variants, such as the promoter rs3865444, can influence expression by modulating of exon 2, thereby altering isoform ratios and overall protein levels. In the , CD33 is expressed predominantly on , the resident myeloid-derived immune cells of the brain, where it correlates with microglial markers like CD11b and AIF-1. immunostaining confirms this localization, with no significant expression observed in neurons or . CD33 exists in two main isoforms: the full-length CD33M (membrane-bound, including 2 for binding) and the shorter D2-CD33 isoform (lacking 2, with reduced ligand-binding capacity), both present on and other myeloid cells, though their relative abundance varies by genetic background. CD33 expression can be modulated under pathological conditions, such as , where it is upregulated on infiltrating myeloid cells in inflamed tissues, contributing to immune regulation in these environments.

Protein Structure

Domain Organization

CD33 is a type I transmembrane with a molecular weight of approximately 40-67 kDa, varying by isoform and extent of . The protein architecture includes an extracellular domain, a single transmembrane helix, and a short cytoplasmic . The extracellular region consists of two immunoglobulin-like domains: an N-terminal V-set Ig domain responsible for sialic acid recognition and a membrane-proximal C2-set Ig domain. These domains form the ligand-binding portion of the receptor, with the V-set domain at the distal end. The transmembrane domain is a 21-residue alpha-helix that anchors CD33 in the plasma membrane and appears to maintain a monomeric configuration. The cytoplasmic tail contains two immunoreceptor tyrosine-based inhibitory motifs (ITIMs), characterized by sequences LxYxxL (around Tyr340) and TxYxxV (around Tyr358), which facilitate recruitment of SH2 domain-containing phosphatases upon phosphorylation. Recent structural studies, including crystal structures of the extracellular domains (PDB IDs 5IHB, 5J06, and 9bet), have revealed a monomer-dimer equilibrium influenced by the C2-set domain, with the transmembrane and cytosolic regions showing limited oligomerization propensity. These analyses indicate minimal conformational shifts in the core domains under physiological conditions.

Sialic Acid Binding and Interactions

CD33, a member of the family, exhibits binding specificity for s, particularly recognizing both α2,3- and α2,6-linked s through its N-terminal V-set immunoglobulin domain. This interaction is mediated by conserved key residues within the V-set domain, including Arg99, which forms electrostatic contacts with the carboxylate group of , and Trp106, which contributes hydrophobic interactions essential for accommodation. The binding affinity is influenced by the glycosylation state of CD33 itself, with N-linked s at Asn69 and Asn114 playing regulatory roles; specifically, the at Asn69 masks the -binding in its sialylated form, and desialylation or at this enhances recognition. CD33 engages in both cis- and trans-interactions with sialylated glycans. In cis-binding, CD33 interacts with sialylated glycans on the same surface, often leading to masking of its and of receptor . Trans-binding occurs when CD33 on one recognizes sialylated glycoproteins on adjacent cells, facilitating - and inhibitory signaling in myeloid cells. These interactions underscore CD33's role as a self-recognition receptor, contributing to by dampening activation signals upon engagement. Among key interacting partners, CD33 forms a sialic acid-dependent cis-interaction with CD45, a receptor tyrosine phosphatase on immune cells, which suppresses CD45's phosphatase activity and thereby influences downstream immune regulation. Additionally, CD33 biophysically interacts with clusterin, a chaperone protein, through sialylated glycan recognition, with genetic associations linking this partnership to altered microglial function. CD33 also binds SHP-1 (PTPN6), a protein tyrosine phosphatase, in a genotype-dependent manner; for example, the Alzheimer's disease risk-associated rs3865444 CC genotype enhances this interaction, promoting inhibitory signaling in cells expressing full-length CD33. These molecular associations highlight CD33's integration into broader inhibitory networks on myeloid cells.

Biological Functions

Role in Myeloid Cell Regulation

CD33 serves as an inhibitory receptor on myeloid progenitors, where it suppresses the and of these cells during hematopoiesis. Engagement of CD33 by monoclonal antibodies has been shown to induce and inhibit in normal hematopoietic progenitors, thereby modulating the expansion of myeloid lineages. This regulatory function helps maintain balance in myeloid cell development, preventing excessive production of mature myeloid cells. Recent studies on individuals with germline CD33 loss-of-function variants have shown mild increases in + monocytes and decreases in CD3+ T cells, along with subtle reductions in overall leukocyte and counts, but no significant clinical manifestations or overt , underscoring CD33's modulatory rather than essential role in myeloid regulation. In macrophages and , CD33 modulates by inhibiting the uptake of apoptotic cells, cellular debris, pathogens, and synapses. The full-length CD33 isoform (CD33M) specifically reduces phagocytic activity, limiting the clearance of these targets and thereby restraining microglial responses in the . This inhibition is mediated through CD33's recognition of ligands on target surfaces, which transduces negative signals to dampen phagocytic efficiency. CD33 also plays a key role in controlling by dampening release from activated monocytes. Reduced CD33 expression correlates with increased production of pro-inflammatory , such as IL-6 and TNF-α, in response to stimuli like or high glucose conditions, indicating that CD33 normally attenuates these responses to prevent excessive . Expression of CD33 is dynamically regulated during myeloid cell maturation and immune , with high levels on immature progenitors and progressive downregulation as cells mature into monocytes, macrophages, or granulocytes. Upon by T-cell contact, phorbol esters, or Fcγ receptor cross-linking, CD33 surface expression decreases on monocytes, fine-tuning immune responses by reducing inhibitory signaling at later stages. This pattern allows CD33 to exert stronger control over early myeloid expansion while permitting robust effector functions in mature cells.

Signaling Mechanisms

CD33 functions primarily as an inhibitory receptor through its intracellular immunoreceptor -based inhibitory motifs (ITIMs). Upon engagement by sialic acid-containing ligands, CD33 undergoes on its ITIMs, mediated by family kinases such as Lyn or . This creates docking sites for the Src homology 2 (SH2) domain-containing protein phosphatases SHP-1 (PTPN6) and SHP-2 (), which are recruited to the phosphorylated ITIMs. The differential binding affinities of the ITIMs for SHP-1 and SHP-2 contribute to fine-tuned inhibitory signaling, with the membrane-proximal ITIM preferentially associating with SHP-1 and the distal one with SHP-2. The recruited SHP-1 and SHP-2 exert their inhibitory effects by dephosphorylating key substrates in activating pathways, thereby dampening downstream signaling in myeloid cells. For instance, these phosphatases target phosphorylated residues on adaptor proteins like DAP12, which is associated with activating receptors such as TREM2, leading to reduced activation of spleen tyrosine kinase (Syk). This inhibition propagates to suppress phosphatidylinositol 3-kinase (PI3K)/Akt and (MAPK) pathways, which are critical for cellular activation, proliferation, and production. Consequently, CD33 signaling attenuates overall myeloid cell responsiveness, promoting a baseline repressive state. CD33 exists in two main isoforms with distinct signaling capabilities: the full-length CD33M and the truncated CD33m. CD33M, which includes both ITIMs, fully mediates inhibitory signaling as described, repressing processes like upon engagement. In contrast, CD33m lacks the V-set -binding due to but retains both ITIMs in the cytoplasmic tail. Due to its lack of ligand-binding capability and predominant intracellular localization, CD33m promotes gain-of-function effects, such as enhanced , through ITIM-mediated signaling. CD33 engages in cross-talk with other receptors to modulate immune responses, notably dampening Fcγ receptor (FcγR)-mediated activation. Ligand-induced CD33 signaling inhibits FcγRI (CD64)-triggered calcium mobilization and downstream effector functions in monocytes and macrophages by recruiting SHP-1/2 to counteract activating signals. This inhibitory cross-talk helps maintain immune homeostasis by preventing excessive activation in response to immune complexes.

Role in Alzheimer's Disease

Genetic Risk Factors

The (SNP) rs2455069 (G>A) in the CD33 gene represents a primary genetic risk factor for (AD), with the minor A conferring protection against susceptibility. This protective is associated with an (OR) of approximately 0.85–0.90 for reduced AD risk and promotes skipping of 2, thereby decreasing expression of the full-length CD33M isoform in . The lead CD33 variant, rs3865444 (C>A), is the most established risk factor at this locus, with the minor protective by reducing CD33 expression and promoting 2 skipping, leading to enhanced microglial . Additional CD33 variants, such as rs12459419, contribute to risk by influencing splicing and expression levels, with the major C linked to higher CD33 expression and increased susceptibility. Meta-analyses of genome-wide studies (GWAS) conducted between 2019 and 2025 have established genome-wide for these CD33 variants, confirming their role in across large multi-ancestry cohorts totaling over 100,000 individuals. High expression of the AD-associated CD33M isoform, driven by risk alleles at these loci, correlates with impaired microglial phagocytic function, exacerbating amyloid-beta accumulation. Population-based studies indicate elevated AD risk among APOE ε4 carriers who also possess CD33 risk alleles, such as the G allele of rs2455069, with evidence of gene-gene interactions accelerating cognitive decline. Earlier meta-analyses (2015–2018) have noted ethnic variations in these associations, including protective effects of CD33 minor alleles more evident in populations compared to East Asian groups.

Pathogenic Mechanisms

CD33 contributes to (AD) progression by impairing the clearance of amyloid-β (Aβ) plaques through its inhibitory effects on microglial . As a sialic acid-binding immunoglobulin-like (), CD33 recognizes sialylated glycans on Aβ aggregates, which triggers recruitment of Src homology 2 domain-containing phosphatase-1 (SHP-1) and SHP-2 via its intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs). This signaling cascade dampens phagocytic activity in , the 's primary immune cells responsible for Aβ removal, resulting in plaque accumulation and sustained . Studies have shown that higher CD33 expression correlates with reduced Aβ uptake in human and AD , exacerbating . In AD, CD33 engages in cis-interactions with other myeloid receptors, such as CD45 (protein tyrosine phosphatase receptor type C), which suppress CD45's phosphatase activity and further hinder microglial responses. This binding disrupts dephosphorylation of key signaling molecules, impairing Aβ clearance and promoting neuronal damage, including loss of dendritic spines in amyloid-exposed neuronal cultures. Recent biophysical studies from 2025 have also identified direct interactions between CD33 and clusterin (CLU), a chaperone protein that modulates Aβ aggregation; these interactions alter CD33's sialic acid affinity and genetically influence AD risk by enhancing plaque stability in microglia-neuron co-cultures. Such molecular cross-talk underscores CD33's role in perpetuating a neurotoxic microenvironment. AD-associated risk alleles, particularly those promoting the membrane-bound long isoform CD33^M, drive microglial dysfunction by upregulating pro-inflammatory pathways while suppressing homeostatic functions. This isoform shift increases expression of inflammatory markers like nestin and reduces responses, fostering a chronic pro-inflammatory state that accelerates and neuronal loss independent of Aβ levels. Proteomic analyses of AD brains reveal altered and impaired in , contributing to inefficient debris clearance and synaptic degeneration. Emerging from 2024–2025 highlights genotype-dependent interactions between CD33 and SHP-1 (PTPN6), where variants enhance SHP-1 to CD33 ITIMs, amplifying inhibitory signaling in response to Aβ exposure. This leads to genotype-specific reductions in density in amyloid-treated neuronal models, linking microglial inhibition directly to synaptic pathology. studies confirm that these interactions correlate with increased Aβ burden and tangles in AD cohorts, providing mechanistic insight into how CD33 variants exacerbate neurodegeneration.

Clinical Applications

Targeting in Acute Myeloid Leukemia

CD33 is expressed on the surface of myeloid cells, including leukemic blasts in 85-90% of (AML) cases, which positions it as a reliable marker for detecting via . The primary therapeutic targeting of CD33 in AML involves (GO), an antibody-drug conjugate approved by the FDA in 2000 under accelerated approval for relapsed CD33-positive AML in patients over 60 years, which was voluntarily withdrawn in 2010 due to lack of confirmatory trial success but re-approved in 2017 at a fractionated lower dose for newly diagnosed CD33-positive AML in adults, with the indication extended to pediatric patients aged 1 month and older in 2020. GO consists of a humanized anti-CD33 conjugated to , a cytotoxic that induces DNA double-strand breaks upon internalization in CD33-expressing cells, leading to . Clinical trials, such as the ALFA-0701 study, demonstrated improved outcomes with GO added to standard induction chemotherapy, including a 2-year overall survival of 53% vs. 42% overall (HR 0.69; 95% CI 0.49-0.98; p=0.0368), with benefits particularly in favorable- and intermediate-risk patients. Emerging CD33-targeted immunotherapies include bispecific T-cell engager antibodies, such as AMG 330 (CD33xCD3), which redirect T cells to lyse CD33-positive blasts. Phase I/II trials of CD33xCD3 bispecifics in relapsed/ AML have reported overall response rates of 30-60%, with complete remissions in about 20-40% of patients, though remains a common managed with step-up dosing. Similarly, CD33-directed chimeric receptor (CAR) T-cell therapies, including those using second-generation constructs with CD3ζ and costimulatory domains, have shown promising early results in phase I/II studies up to 2025, achieving complete response rates of 40-60% in heavily pretreated AML patients, particularly when combined with lymphodepletion. As of 2025, phase I/II trials of CD33 CAR-T therapies continue to show complete response rates of 40-60% in relapsed/ AML, with efforts to mitigate on-target off-tumor toxicity. Resistance to CD33-targeted therapies, particularly GO, can arise from alternative splicing of CD33 transcripts that exclude exon 2, resulting in isoforms lacking the membrane-distal V-set Ig-like domain and reducing surface expression by up to 50% in some AML cells, thereby evading antibody binding. This splicing polymorphism, present in approximately 20-30% of AML cases depending on rs12459419 genotype, correlates with inferior response rates to GO and has prompted investigations into splice variant-specific targeting strategies.

Therapeutic Potential in Neurodegeneration

The therapeutic potential of targeting CD33 in neurodegeneration, particularly (AD), stems from its role in inhibiting microglial phagocytosis of amyloid-β (Aβ) plaques, a hallmark of AD pathology. Downregulating CD33 expression or function enhances microglial clearance of Aβ, reducing plaque burden and in preclinical models. For instance, genetic knockout of CD33 in AD mouse models (e.g., APP/PS1 and 5xFAD) has been shown to decrease Aβ levels by approximately 30-40% and amyloid plaque density by 20-35%, while improving cognitive outcomes. These findings position CD33 inhibition as a strategy to restore beneficial microglial activity without broadly suppressing innate immunity. Preclinical studies have explored anti-CD33 antibodies to block CD33 signaling and promote Aβ uptake. In models expressing full-length CD33, administration of monoclonal antibodies such as AL003 downregulated surface CD33 on , leading to increased Aβ and reduced plaque-associated dystrophic neurites. Similarly, approaches using (AAV) vectors to deliver CD33-specific (shRNA) in APP/PS1 mice resulted in 25-35% reductions in soluble Aβ42 and cortical plaque area when initiated early in disease progression. These interventions also attenuated by lowering pro-inflammatory levels, such as IL-1β, in the . Emerging gene editing strategies aim to address AD-associated CD33 variants, such as the rs2455069 polymorphism, which increases risk by altering CD33 splicing and surface expression. /Cas9-mediated editing of rs2455069 in induced pluripotent stem cell-derived has demonstrated potential for isoform switching from the risk-conferring full-length CD33 to the protective short isoform lacking the sialic acid-binding , thereby enhancing Aβ clearance without off-target effects. Additionally, recent 2025 investigations into the CD33-CD45 interaction have revealed that CD33 binding inhibits CD45 activity, impairing microglial signaling; small-molecule inhibitors disrupting this cis-interaction restored function and improved phagocytic efficiency in human myeloid cells, suggesting a novel therapeutic avenue for AD. Clinical translation has advanced to early-phase trials, with anti-CD33 monoclonal antibodies like AL003 entering Phase I and Ib studies between 2020 and 2022 to evaluate safety, target engagement, and biomarker changes in mild-to-moderate AD patients. These trials focused on (CSF) Aβ levels and peripheral CD33 downregulation, showing preliminary tolerability but were discontinued due to strategic shifts by collaborators. No Phase II/III trials are currently active as of 2025, though ongoing preclinical optimization emphasizes brain-penetrant formats. Key challenges include achieving sufficient blood-brain barrier penetration for large-molecule antibodies, which limits central efficacy, and mitigating off-target effects on peripheral myeloid cells, potentially increasing infection risk. Combination therapies with anti-amyloid agents, such as , are proposed to synergize plaque clearance with microglial activation, though long-term safety data remain needed.

References

  1. [1]
    945 - Gene ResultCD33 CD33 molecule [ (human)] - NCBI
    Aug 19, 2025 · CD33 expression on natural killer cells is a potential confounder for residual disease detection in acute myeloid leukemia by flow cytometry.Missing: scientific | Show results with:scientific
  2. [2]
    Siglecs and their roles in the immune system - Nature
    An important function of CD33-related Siglecs is their ability to regulate cell growth and survival, either by inhibition of proliferation or induction of ...
  3. [3]
    Acute myeloid leukemia stem cells and CD33-targeted immunotherapy
    We review studies on the nature of stem cells in AML, discuss clinical data on the effectiveness of CD33-directed therapy, and consider the mechanistic basis ...
  4. [4]
    Alzheimer's Disease Risk Gene CD33 Inhibits Microglial Uptake of ...
    Apr 25, 2013 · We have previously shown that the CD33 gene is a risk factor for Alzheimer's disease (AD). Here, we observed increased expression of CD33 in microglial cells ...
  5. [5]
    Distribution and levels of cell surface expression of CD33 and ... - PMC
    Jun 13, 2014 · CD33 was detected on blasts of 85–90% of patients presenting with AML as well as on normal myeloid progenitors and myelocytes. Interestingly, ...
  6. [6]
    Treatment of CD33-directed Chimeric Antigen Receptor-modified T ...
    Sep 23, 2014 · CD33 is primarily expressed on multipotent myeloid precursors, unipotent colony-forming cells, maturing granulocytes and monocytes, peripheral ...<|control11|><|separator|>
  7. [7]
    CD33 Expression and Gentuzumab Ozogamicin in Acute Myeloid ...
    Jun 28, 2021 · Physiologically, CD33 expression is restricted to early multi-lineage hematopoietic progenitors (CFU-GEMM, CFU-GM, CFU-G, and E-BFU), ...
  8. [8]
    CD33 Antigen - an overview | ScienceDirect Topics
    CD33 antigen expression is down-regulated with maturation of the myeloid lineage, resulting in low-level expression on peripheral granulocytes and tissue ...
  9. [9]
    CD33 Alzheimer's Risk-Altering Polymorphism, CD33 Expression ...
    Aug 14, 2013 · In summary, these results suggest a novel model wherein SNP-modulated RNA splicing modulates CD33 function and, thereby, AD risk. Introduction.
  10. [10]
    Differential gene expression of tumor-infiltrating CD33+ myeloid ...
    Sep 30, 2020 · CD33 is expressed on a heterogenous population of myeloid cells, and it does not distinguish MDSCs from TAMs or tumor-associated neutrophils ( ...
  11. [11]
    Novel CD33 antibodies unravel localization, biology and therapeutic ...
    Dec 24, 2020 · Figure 1C shows protein bands corresponding to approximately 67 kDa and approximately 40–42 kDa for the glycosylated and unglycosylated forms ...
  12. [12]
    Structure of the CD33 Receptor and Implications for the Siglec Family
    Thus, the CD33 TM helix is composed of a thin neck and a thicker belly, which are followed by a positively charged, cytosolic patch. Figure 3. Figure 3.Relevance Of Cd33... · Cd33 Forms A Monomeric... · Cd33 Signaling Hypotheses
  13. [13]
    The Myeloid-specific Sialic Acid-binding Receptor, CD33 ...
    Interestingly, the cytoplasmic tail of CD33 contains two tyrosine-based motifs (LXY340 XXL and TXY358 XXV), both potential ITIMs, although only the first ...<|control11|><|separator|>
  14. [14]
    CD33/Siglec-3 Binding Specificity, Expression Pattern, and ... - NIH
    Many CD33-related Siglecs are expressed on cells involved in innate immunity. For example, hSiglec-7/AIRM-1 is expressed on natural killer cells and monocytes ( ...
  15. [15]
  16. [16]
    Siglecs in the immune system - PMC - NIH
    ... Trp106) on the A and G strands that form hydrophobic contacts with the ... The myeloid-specific sialic acid-binding receptor, CD33, associates with the ...
  17. [17]
    Probing the cis interactions of the inhibitory receptor Siglec‐7 with ...
    Jul 20, 2006 · Its sialic acid-binding activity on NK cells is masked by cis interactions with sialylated glycans, which are likely to be important for ...
  18. [18]
    Targeting the Siglec–Sialic Acid Immune Axis in Cancer
    Siglecs are the key immune receptors that bind to cell-surface sialic acids, with interactions that ultimately lead to modulation of the immune system (10, 16, ...
  19. [19]
    Modulation of Immune Tolerance via Siglec-Sialic Acid Interactions
    (A) Siglec-2,−3, and−5 till−11 modulate TLR signaling upon binding of sialic acids and thereby dampen proinflammatory responses. Ligand binding to Siglec-3 ...<|control11|><|separator|>
  20. [20]
    CD33 and clusterin interact biophysically and genetically ... - bioRxiv
    Jul 31, 2025 · CD33 and clusterin interact biophysically and genetically to modulate Alzheimer risk. Roger B. Dodd, Masahiro Enomoto, Ye Zhou ...
  21. [21]
    CD33 and SHP-1/PTPN6 Interaction in Alzheimer's Disease - MDPI
    Sep 13, 2024 · Our findings demonstrate a genotype-dependent interaction between CD33 and SHP-1, which may functionally contribute to the AD risk associated with this CD33 ...
  22. [22]
    The inhibitory effect of anti-CD33 monoclonal antibodies on AML ...
    Functional studies revealed that engagement of CD33 induced apoptosis and inhibition of proliferation in normal myeloid cells and leukemia cells from AML and ...
  23. [23]
    Engagement of p75/AIRM1 or CD33 inhibits the proliferation of ... - NIH
    These data suggest a modulatory role of p75/AIRM1 and CD33 during myeloid differentiation and may offer clues toward novel approaches in the therapy of myeloid ...
  24. [24]
    CD33M inhibits microglial phagocytosis, migration and proliferation ...
    In the BV-2 cells, CD33M inhibited microglial phagocytosis of beads, synapses, debris and dead cells, while CD33m increased phagocytosis of beads, debris and ...
  25. [25]
    Repression of phagocytosis by human CD33 is not conserved with ...
    Dec 3, 2019 · Our findings demonstrate that expression of the long isoform of hCD33 (hCD33M) alone is sufficient to repress phagocytosis in both monocytes and ...Missing: mammals | Show results with:mammals
  26. [26]
    High glucose concentrations induce TNF-α production through the ...
    Apr 14, 2012 · CD33 is known to play a role in the regulation of cytokine production, and low levels of CD33 expression have been associated with high levels ...
  27. [27]
    CD33‐related siglecs as potential modulators of inflammatory ...
    Feb 21, 2012 · Antibody crosslinking of siglec-E on LPS-stimulated macrophages led to reduced production of proinflammatory cytokines, TNF-α and IL-6.Inhibitory Siglecs As... · Siglecs And Inflammatory... · Cd33-Related Siglecs And...
  28. [28]
    CD33 Expression and Gentuzumab Ozogamicin in Acute Myeloid ...
    Jun 28, 2021 · Physiologically, CD33 expression is restricted to early multi-lineage hematopoietic progenitors (CFU-GEMM, CFU-GM, CFU-G, and E-BFU), ...Cd33 Expression And... · 2. Cd33 Expression In Acute... · 12. Cd33 Bispecific...
  29. [29]
    Reporter cell assay for human CD33 validated by specific antibodies ...
    Jun 29, 2021 · Further, murine CD33 has a positive charged residue in its transmembrane domain, which is known to be critical for interaction with ITAM- ...Expression Of Cd33 On The... · Generation Of Cd33 Reporter... · Calcium Imaging In Cd33...
  30. [30]
    Myeloid specific human CD33 is an inhibitory receptor ... - PubMed
    Jul 15, 2000 · Human CD33 has two tyrosine residues in its cytoplasmic domain (Y340 and Y358). ... Amino Acid Sequence; Antigens, CD / chemistry; Antigens ...
  31. [31]
    Emerging roles for ITAM and ITIM receptor signaling in microglial ...
    Oct 11, 2023 · TREM2 acts downstream of CD33 in modulating microglial pathology in Alzheimer's disease. ... PI3K/AKT/FoxO3a signaling pathway in ...2.2 Microglia In Alzheimer's... · 3.1. 1 Trem2/dap12 Signaling · 3.2. 1 Siglec Receptor...
  32. [32]
    The CD33 short isoform is a gain-of-function variant that enhances ...
    Mar 25, 2021 · We find that hCD33m enhances phagocytosis. This contrasts with the human CD33 long isoform (hCD33M) that represses phagocytosis, as previously demonstrated.
  33. [33]
    CD33 Alzheimer's Risk-Altering Polymorphism, CD33 Expression ...
    Aug 14, 2013 · In summary, CD33 expression is increased in AD and in individuals with the rs3865444C allele that is associated with increased AD risk. Moreover ...
  34. [34]
  35. [35]
    Dissecting the Ability of Siglecs To Antagonize Fcγ Receptors
    Fcγ receptors (FcγRs) play key roles in the effector function of IgG, but their inappropriate activation plays a role in several disease etiologies.
  36. [36]
    CD33 rs2455069 SNP: Correlation with Alzheimer's Disease ... - MDPI
    These SNPs can affect the expression level, the structure, and the function of CD33, leading to changes in microglia-mediated clearance of amyloid β, likely ...
  37. [37]
    CD33 rs2455069 SNP: Correlation with Alzheimer's Disease and ...
    Mar 26, 2022 · We demonstrate an association between the G allele of the rs2455069 gene variant and the presence of AD in a cohort of 195 patients from southern Italy.Missing: frequency | Show results with:frequency
  38. [38]
    Alzheimer's Disease-Associated Alternative Splicing of CD33 Is ...
    Feb 13, 2023 · ... CD33, rs12459419, affects the alternative splicing of this exon. The minor allele is associated with a reduced risk of AD and promotes the ...
  39. [39]
    Multi-ancestry genome-wide meta-analysis of 56241 individuals ...
    Jul 17, 2025 · We construct and analyze a multi-ancestry GWAS dataset in the Alzheimer's Disease Genetics Consortium (ADGC) to test for novel shared and ...
  40. [40]
    New insights into the genetic etiology of Alzheimer's disease and ...
    Apr 4, 2022 · We performed a two-stage genome-wide association study totaling 111,326 clinically diagnosed/'proxy' AD cases and 677,663 controls. We found 75 ...
  41. [41]
    Alzheimer's disease associated isoforms of human CD33 ...
    May 27, 2024 · Genome-wide association studies (GWAS) suggest that approximately 70% of Alzheimer's disease (AD) risk is attributable to genetic factors [1, 2] ...
  42. [42]
    Effect of APOE and CD33 on Cognitive Decline - PMC
    Jun 23, 2015 · Conclusions. Individuals with multiple AD risk genes in addition to having one or more APOE ε4 alleles are at greater risk of cognitive decline ...
  43. [43]
    Meta-analysis of the association between CD33 and Alzheimer's ...
    Both lines of evidence indicate that CD33 gene could play an important role in susceptibility to AD. In 2011, three large-scale genome-wide association studies ...Missing: 2019-2025 | Show results with:2019-2025
  44. [44]
    Association of CD33 and MS4A cluster variants with Alzheimer's ...
    Aug 10, 2025 · In summary, our results suggest that CD33 and MS4A cluster variants are associated with LOAD susceptibility in East Asian populations.
  45. [45]
    CD33M inhibits microglial phagocytosis, migration and proliferation ...
    Mar 15, 2021 · Over-expression of Alzheimer's risk variant of CD33, CD33M, inhibits phagocytosis, proliferation and migration but increases adhesion to ...
  46. [46]
    CD33-CD45 Interaction Reveals a Mechanistic Link to Alzheimer's ...
    Jul 31, 2025 · Overexpression of CD33 or loss of CD45 contributes to impaired microglial clearance of amyloid beta and amyloid beta-induced loss of dendritic ...
  47. [47]
    CD33-CD45 Interaction Reveals a Mechanistic Link to Alzheimer's ...
    Jul 31, 2025 · Here we identify CD45 as an immune cell-specific sialic acid-dependent cis CD33 binding partner, whose phosphatase activity is inhibited by CD33 ...
  48. [48]
    Alzheimer's genes in microglia: a risk worth investigating
    Nov 20, 2023 · We note that impairment to protective phenotypes may include prolonged or insufficient microglial activation, resulting in dysregulated ...
  49. [49]
    CD33 and SHP-1/ PTPN6 Interaction in Alzheimer's Disease - PubMed
    Sep 13, 2024 · Our findings demonstrate a genotype-dependent interaction between CD33 and SHP-1, which may functionally contribute to the AD risk associated ...
  50. [50]
    Targeting CD33 for acute myeloid leukemia therapy - BMC Cancer
    Jan 3, 2022 · These findings provide a strong rationale for targeting CD33 in combination with chemotherapy, which can be considered a promising therapeutic strategy for AML.
  51. [51]
    Mylotarg (gemtuzumab ozogamicin) FDA Approval History
    Sep 7, 2020 · Mylotarg originally received accelerated approval in May 2000 as a stand-alone treatment for older patients with CD33-positive AML who had experienced a ...
  52. [52]
    T-cell-based immunotherapy of acute myeloid leukemia - Nature
    May 5, 2021 · Responders were more likely to have higher AMG 330 exposures and lower baseline leukemic burden, with no correlation between CD33 expression on ...Targeting Cd33 With... · Targeting Cd123 With T-Cell... · Chimeric Antigen Receptor...
  53. [53]
    Honing CAR T cells to tackle acute myeloid leukemia | Blood
    Mar 13, 2025 · Overall, most patients with AML treated with CD33-, CD123-, or CLEC12A-directed CAR stand-alone therapy have not achieved durable responses.
  54. [54]
    Alzheimer's Disease Risk Gene CD33 Inhibits Microglial Uptake of ...
    May 22, 2013 · We have previously shown that the CD33 gene is a risk factor for Alzheimer's disease (AD). Here, we observed increased expression of CD33 in microglial cells ...
  55. [55]
    Gene therapy for Alzheimer's disease targeting CD33 reduces ...
    CD33 is upregulated on microglial cells from post-mortem AD patient brains, and high levels of CD33 inhibit uptake and clearance of amyloid beta (Aβ) in ...<|control11|><|separator|>
  56. [56]
    A Phase 1 Study of AL003 in Healthy Volunteers and Participants ...
    May 3, 2022 · AL003 is a human anti-CD33 monoclonal antibody developed for the treatment of Alzheimer's disease (AD). CD33 regulates uptake of the amyloid ...
  57. [57]
    Application of CRISPR/Cas9 in Alzheimer's Disease - Frontiers
    CRISPR/cas9 Targets CD33. Human genetic association studies indicate that immune response is also the major pathway of AD etiology. The importance of chronic ...
  58. [58]
    First in Human Study for Safety and Tolerability of AL003 ...
    This is a multi-center, randomized, double-blind, placebo-controlled, dose escalation first in human (FIH) study in healthy adults and in patients with mild to ...
  59. [59]
    Alector Announces Initiation of Phase 1b Study of AL003 in ...
    Phase 1b study of AL003 will enroll approximately 12 Alzheimer's disease patients to assess safety, pharmacokinetics and pharmacodynamics, and monitor target ...
  60. [60]
    [PDF] Anti-CD33 - Alzheimer's Drug Discovery Foundation
    Neuroprotective Benefit: CD33 alleles that reduce CD33 surface expression on microglia are associated with decreased risk for Alzheimer's because high CD33 ...<|control11|><|separator|>
  61. [61]
    CD33 in Alzheimer's Disease – Biology, Pathogenesis, and ...
    Dec 12, 2018 · This review summarizes the recently advanced biology of CD33 and its association with AD. It also provides insights from a drug discovery perspective.