CD33 Human

CD33 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Immune Regulation

CD33 dampens inflammatory responses by:

  • Inhibiting phagocytosis and cytokine release via ITIM-SHP phosphatase signaling .

  • Modulating cross-talk with activatory receptors like TREM2 .

Alzheimer’s Disease (AD)

  • The rs3865444(A) and rs12459419(T) SNPs reduce full-length CD33 (CD33M) expression by promoting exon 2 skipping (CD33 ΔE2) .

  • CD33M suppresses amyloid-beta clearance by microglia, whereas the ΔE2 variant is protective .

Reporter Cell Systems

Studies using chimeric CD33-DAP12 reporter cells revealed:

  • Antibodies P67.6 and 1c7/1 activate CD33 signaling, evidenced by SYK phosphorylation and calcium flux .

  • CD33 ΔE2 fails to bind antibodies targeting the V-set domain (e.g., WM53, P67.6) .

Antibody Clones and Applications

CloneEpitope TargetReactivityKey Findings
WM53V-set domain (exon 2)Full-length CD33 onlyFails to detect CD33 ΔE2 .
P67.6Proximal V-set domainFull-length CD33 onlyActivates CD33 signaling; used in calcium imaging and phagocytosis assays .
1c7/1C2-set domainAll isoformsDetects CD33 ΔE2; used in flow cytometry and functional studies .

Evolutionary and Population Genetics

  • The protective rs3865444(A) allele is human-specific and absent in Neanderthals/Denisovans .

  • Population frequencies: 5% in Africans, 48% in Native Americans .

  • Evolutionary trade-off: Elevated CD33M expression in humans increases AD risk, countered by derived protective alleles .

Therapeutic Challenges and Future Directions

  • Current limitations: Splice variants (e.g., CD33 ΔE2) evade antibody-based therapies .

  • Opportunities: Develop antibodies targeting conserved C2-set domains (e.g., 1c7/1) or small molecules modulating CD33-TREM2 interactions .

Product Specs

Introduction
CD33 is a putative adhesion molecule found on myelomonocytic-derived cells. It facilitates sialic-acid dependent binding to other cells, showing a preference for alpha-2,6-linked sialic acid. CD33's sialic acid recognition site can be masked by cis interactions with sialic acids present on the same cell surface. In the context of immune responses, CD33 acts as an inhibitory receptor upon tyrosine phosphorylation triggered by ligands. This is achieved by recruiting cytoplasmic phosphatases through its SH2 domains, which in turn, dephosphorylate signaling molecules and block signal transduction. Furthermore, CD33 has been shown to induce apoptosis in acute myeloid leukemia.
Description
Recombinant human CD33, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 265 amino acids (specifically, amino acids 18-259) and has a molecular weight of 29.1 kDa. The CD33 protein is fused to a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile filtered solution.
Formulation
The CD33 protein solution has a concentration of 1 mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the CD33 protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
CD33Molecule, CD33Antigen (Gp67), Sialic Acid Binding Ig-Like Lectin 3, Sialic Acid-Binding Ig-Like Lectin 3, SIGLEC3, gp67, SIGLEC-3, Myeloid Cell Surface AntigenCD33, p67, Siglec-3, CD33Antigen.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSDPNFWLQ VQESVTVQEG LCVLVPCTFF HPIPYYDKNS PVHGYWFREG AIISGDSPVA TNKLDQEVQE ETQGRFRLLG DPSRNNCSLS IVDARRRDNG SYFFRMERGS TKYSYKSPQL SVHVTDLTHR PKILIPGTLE PGHSKNLTCS VSWACEQGTP PIFSWLSAAP TSLGPRTTHS SVLIITPRPQ DHGTNLTCQV KFAGAGVTTE RTIQLNVTYV PQNPTTGIFP GDGSGKQETR AGVVH.

Q&A

What is CD33 and what is its role in human biology?

CD33, also known as Sialic acid-binding Ig-like lectin 3 (SIGLEC3), is an immunoregulatory receptor primarily expressed on myeloid cells. It functions as an inhibitory receptor that mediates signaling via tyrosine phosphatases . CD33 plays critical roles in modulating innate immune responses through interaction with sialic acid-containing ligands. In the central nervous system, CD33 expression on microglia influences amyloid beta peptide clearance, which has significant implications for neurodegenerative diseases such as Alzheimer's disease .

To study CD33 function, researchers typically employ techniques including:

  • Flow cytometry for expression analysis

  • Western blotting for protein detection (appearing at approximately 55 kDa)

  • Immunofluorescence for localization studies

  • Reporter assays for functional assessment

What are the major isoforms of CD33 in humans and their functional differences?

Humans express two primary CD33 isoforms:

  • CD33M (full-length): Contains the complete extracellular domain with the functional IgV domain that mediates sialic acid binding.

  • CD33 ΔE2 (D2-CD33): Lacks exon 2, which partially encodes the IgV domain, resulting in loss of sialic acid binding capability .

The ratio between these isoforms varies among individuals and is influenced by genetic polymorphisms, particularly rs12459419, which affects splicing efficiency of exon 2 . Notably, CD33 ΔE2 shows reduced surface expression on cells compared to CD33M and is associated with enhanced amyloid beta clearance in the brain .

Experimental validation of these isoforms can be performed using specific antibody clones: while antibody clone 1c7/1 recognizes both isoforms, clones WM53 and P67.6 bind only to CD33M and not to CD33 ΔE2 .

How can different CD33 isoforms be detected and distinguished experimentally?

Distinguishing between CD33 isoforms requires specific methodological approaches:

MethodApplicationIsoform Differentiation
Flow cytometryQuantitative expression analysisUse of isoform-specific antibodies: clone 1c7/1 detects both isoforms (92.80% detection for CD33M, 75.72% for CD33 ΔE2), while WM53 and P67.6 detect only CD33M
RT-PCRTranscript analysisPrimers spanning exon 2 can differentiate between isoforms
Western blotProtein detectionDifferent molecular weights for CD33M (~55 kDa) vs. CD33 ΔE2
ELISAQuantitative protein analysisAntibody selection determines isoform specificity
ImmunofluorescenceCellular localizationCan be combined with isoform-specific antibodies to visualize distribution patterns

For optimal results, researchers should validate antibody specificity using both CD33-positive cell lines (e.g., U937, MV4-11) and CD33-negative controls (e.g., RS4;11, CHO) .

What are the optimal cell models for studying CD33 function in vitro?

Several validated cell models are available for CD33 research:

  • Myeloid cell lines:

    • U937 (human histiocytic lymphoma): Expresses high levels of CD33, suitable for localization and functional studies

    • HL-60 (promyelocytic leukemia): CD33-positive model for leukemia research

    • MOLM-14 and MV4-11 (acute myeloid leukemia): Express CD33 and used in targeted therapy research

  • Negative controls:

    • RS4;11 and Reh (acute lymphocytic leukemia): CD33-negative controls

    • CHO (Chinese hamster ovary): Non-human cell line used as negative control

  • Reporter systems:

    • CD33-DAP12 chimeric reporter cell lines have been developed to shift CD33 signaling from inhibitory to activatory for easier detection

    • These systems can incorporate calcium-sensitive fluorescent proteins (e.g., GCaMP6m) for visualizing signaling events

When selecting a model system, consider the specific CD33 isoform expression pattern and experimental readout requirements.

How do CD33 polymorphisms affect protein function and disease susceptibility?

Several key polymorphisms influence CD33 function and disease associations:

PolymorphismFunctional EffectDisease Association
rs3865444(A)Associated with increased CD33 ΔE2 to CD33M ratioProtective against Alzheimer's disease
rs12459419(T)Modulates splicing efficiency of exon 2Co-inherited with rs3865444(A), increases CD33 ΔE2 expression

Individuals homozygous for the rs3865444C (risk allele) exhibit greater cell surface expression of CD33M compared to those with rs3865444A (protective allele) . The mechanisms behind these effects include:

To study these polymorphisms, researchers can use genotyping assays, transcript analysis to measure isoform ratios, and functional studies to assess the impact on cellular processes like phagocytosis.

What evolutionary insights have emerged from studying human-specific CD33 variants?

Human CD33 shows distinctive evolutionary features compared to other primates:

  • Expression differences: Humans show higher expression of CD33M compared to chimpanzees, suggesting upregulation in the human lineage after divergence from common ancestors

  • Human-specific protective alleles: The protective rs3865444(A) allele is derived and unique to humans, despite weak direct selection on older individuals

  • Selection pressures: The evolution of protective CD33 alleles may be driven by inclusive fitness effects, where maintaining cognitive function in older individuals provides benefits to related younger kin

These findings suggest that selection may have favored alleles protecting against cognitive decline in postreproductive humans, maximizing their contributions through care for offspring, foraging assistance, and knowledge transmission .

Research methods to investigate evolutionary aspects include comparative genomics, population genetics, and functional studies comparing human and non-human primate CD33 variants.

What mechanisms link CD33 to Alzheimer's disease pathology?

CD33 contributes to Alzheimer's disease (AD) pathology through several interconnected mechanisms:

  • Inhibition of microglial phagocytosis: CD33M suppresses microglial uptake and clearance of amyloid beta peptides, leading to increased amyloid accumulation

  • Expression-pathology correlation: CD33 expression levels positively correlate with amyloid beta levels and plaque load in AD patient brains

  • Isoform-specific effects: The CD33 ΔE2 variant, which lacks the sialic acid binding domain, does not inhibit microglial phagocytosis as effectively as CD33M, resulting in enhanced amyloid clearance

Experimental approaches to study these mechanisms include:

  • Ex vivo analysis of human brain tissue for CD33 expression and amyloid load correlation

  • In vitro phagocytosis assays using microglia expressing different CD33 variants

  • Animal models with modified CD33 expression or humanized CD33 to assess effects on amyloid pathology

How do protective CD33 alleles influence microglial function in relation to amyloid clearance?

The protective effect of certain CD33 alleles against AD operates through specific microglial pathways:

  • The rs3865444(A) allele (protective) is co-inherited with rs12459419(T), which alters exon 2 splicing efficiency

  • This genetic variation results in:

    • Increased expression of CD33 ΔE2 (lacking sialic acid binding)

    • Decreased expression of inhibitory CD33M

    • Reduced CD33 surface expression on microglia

  • Functional consequences include:

    • Enhanced microglial phagocytic activity toward amyloid beta

    • Reduced amyloid deposition in brain tissue

    • Lower risk of developing Alzheimer's disease

To quantify these effects, researchers employ techniques including:

  • Single-cell RNA sequencing to analyze microglial heterogeneity

  • Live-cell imaging to track amyloid phagocytosis rates

  • Analysis of CD33 isoform ratios in different genetic backgrounds

How is CD33 utilized as a target for leukemia immunotherapy?

CD33 serves as an important target in acute myeloid leukemia (AML) therapy due to its expression pattern on leukemic cells. Current research focuses on several approaches:

  • Chimeric Antigen Receptor (CAR) T-cell therapies:

    • CAR33VH: Utilizes a human immunoglobulin heavy chain variable domain as targeting sequence

    • My96CAR: Based on the scFv from My96 antibody

  • CAR design considerations:

    • Tumor-targeting domain (CD33-binding region)

    • Linker and transmembrane domains (often derived from CD8)

    • Intracellular signaling components (4-1BB costimulatory domain and CD3 zeta)

  • Expression and efficacy metrics:

    • CAR33VH shows expression ranging from 27-49% across different donors

    • My96CAR exhibits expression levels of 61-89%

    • Expression increases with higher viral transduction (MOI)

These therapies demonstrate specific killing of CD33-positive tumors both in vitro and in vivo, with no activity against CD33-negative cell lines .

What methodological challenges exist in developing CD33-targeted therapeutic approaches?

Developing effective CD33-targeted therapies presents several methodological challenges:

  • Target heterogeneity:

    • Variable CD33 expression levels between patients

    • Heterogeneous expression within the same patient's disease

    • Different isoform ratios affecting targeting efficacy

  • Specificity validation:

    • Requires testing against CD33-positive cell lines (MV4-11, U937)

    • Must include CD33-negative controls (RS4;11, CHO)

    • Antibody clone selection affects recognition of specific CD33 variants

  • Therapeutic window:

    • On-target/off-tumor effects on normal CD33-expressing myeloid cells

    • Need for controlled activity or cellular engineering approaches

  • Technical optimization:

    • CAR expression levels vary with transduction methods and MOI

    • Binding domain selection affects CAR surface expression and function

Addressing these challenges requires comprehensive validation using multiple experimental systems and careful consideration of CD33 biology in both normal and malignant contexts.

How can CD33 reporter systems be designed and validated for functional studies?

Designing effective CD33 reporter systems involves strategic engineering approaches:

  • Chimeric receptor strategy:

    • Replace inhibitory signaling domains with activatory components

    • Example: CD33-DAP12 chimera with D50A mutation to eliminate interactions with other receptors

  • Readout mechanisms:

    • SYK phosphorylation detection via AlphaLISA

    • Calcium flux visualization using GCaMP6m (calcium-sensitive GFP variant)

  • Validation parameters:

    • Surface expression confirmation (92.80% for CD33M-DAP12-GCaMP6m reporter, 75.72% for CD33ΔE2-DAP12-GCaMP6m using appropriate antibodies)

    • Isoform-specific detection (1c7/1 detects both isoforms, WM53 and P67.6 only detect CD33M)

    • Functional validation through ligand-induced signaling

  • Controls:

    • Reporter cells lacking CD33 expression

    • Use of isotype-matched antibody controls

    • Dose-response testing with known ligands or antibodies

This approach allows for real-time monitoring of CD33 activation in response to various stimuli and can be adapted to study different CD33 variants and mutations.

What single-cell approaches are most effective for studying CD33 expression heterogeneity?

Single-cell technologies provide powerful tools to investigate CD33 expression heterogeneity:

TechnologyApplicationKey Advantages
Single-cell RNA-seqTranscriptional profilingReveals isoform ratios and co-expression patterns with other genes
Mass cytometry (CyTOF)Protein expression analysisSimultaneous detection of CD33 with dozens of other markers
Imaging mass cytometrySpatial expression analysisPreserves tissue architecture while quantifying expression
Multiplexed immunofluorescenceCellular localizationVisualizes CD33 distribution on cell surface and intracellularly
Single-cell ATAC-seqChromatin accessibilityIdentifies regulatory elements controlling CD33
expression

Methodological considerations include:

  • Sample preparation to maintain cellular integrity

  • Antibody selection for specific CD33 isoform detection

  • Computational analysis approaches for identifying cell clusters

  • Integration of multiple data modalities for comprehensive characterization

These approaches have revealed important insights about CD33 expression heterogeneity in both healthy tissues and disease states, including variable expression patterns in different myeloid cell subsets.

How can structure-function relationships of CD33 be investigated experimentally?

Investigating CD33 structure-function relationships requires multifaceted experimental approaches:

  • Domain mapping studies:

    • The IgV domain (partially encoded by exon 2) mediates sialic acid binding

    • Differential antibody recognition reveals functional domains (e.g., antibodies WM53 and P67.6 bind the V-set domain absent in CD33 ΔE2)

  • Mutagenesis approaches:

    • Site-directed mutagenesis of key residues

    • Creation of domain-swapped chimeric proteins

    • Generation of truncated variants to isolate functional domains

  • Binding and functional assays:

    • ELISA-based binding studies with recombinant CD33 variants

    • Flow cytometry to assess binding to cell-surface CD33

    • Reporter systems to measure signaling outcomes

  • Structural biology techniques:

    • X-ray crystallography of CD33 domains

    • Cryo-EM for larger complexes

    • Molecular dynamics simulations to predict conformational changes

These approaches can elucidate how specific structural features contribute to CD33 function, including sialic acid binding, signaling capabilities, and interactions with other molecules, providing insights for therapeutic targeting and understanding disease mechanisms.

What are the emerging therapeutic approaches targeting CD33 beyond traditional antibodies?

Current research is exploring several innovative approaches beyond conventional antibody therapies:

  • Next-generation CAR designs:

    • Inducible or switchable CARs to control toxicity

    • Dual-targeting CARs combining CD33 with other myeloid markers

    • VH-only binding domains that show efficacy comparable to scFv-based CARs

  • RNA-based therapeutics:

    • Antisense oligonucleotides targeting CD33 splicing

    • siRNA approaches to modulate CD33 expression

    • RNA editing to alter CD33 function

  • Small molecule modulators:

    • Compounds affecting CD33 glycosylation

    • Inhibitors of CD33 downstream signaling

    • Agents that modify CD33 surface expression

  • Genetic approaches:

    • CRISPR-based editing to modify CD33 variants

    • Engineered cellular therapies with modified CD33 signaling

Each approach requires specific validation strategies, including in vitro functional assays, animal models, and eventually clinical testing to determine efficacy and safety profiles.

How might integrating CD33 research with broader immunological and neurological contexts advance understanding?

Integrating CD33 research with broader contexts offers several promising research avenues:

  • Immune-CNS interactions:

    • Investigating how peripheral immune CD33 expression influences central nervous system function

    • Exploring blood-brain barrier models to study myeloid cell trafficking

    • Examining how systemic inflammation affects brain CD33 function

  • Multi-omics approaches:

    • Integrating genomics, transcriptomics, and proteomics data related to CD33

    • Mapping CD33 interaction networks in different cellular contexts

    • Identifying novel regulatory mechanisms of CD33 expression

  • Therapeutic synergies:

    • Combining CD33-targeting approaches with other immune modulators

    • Exploring interactions between CD33 and other Siglec family members

    • Investigating how CD33 modulation affects response to standard therapies

  • Evolutionary medicine perspective:

    • Further exploring how human-specific CD33 variants evolved

    • Investigating CD33 function across different species to identify conserved mechanisms

    • Understanding why protective alleles emerged specifically in humans

These integrative approaches require collaborative research spanning immunology, neuroscience, oncology, and evolutionary biology to fully elucidate CD33's complex roles in human health and disease.

Product Science Overview

Structure

CD33 is a single-pass type I membrane protein that contains two immunoglobulin-like domains: one V-set domain and one C2-set domain . The extracellular portion of CD33 is responsible for binding sialic acids, while the intracellular portion contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that are involved in the inhibition of cellular activation .

Function

The primary function of CD33 is to modulate the immune response. It acts as an inhibitory receptor that dampens the activation of immune cells, thereby preventing excessive inflammation and autoimmunity . CD33 achieves this by recruiting phosphatases to its ITIMs, which then dephosphorylate key signaling molecules involved in cell activation .

Recombinant CD33

Recombinant CD33 is a form of the protein that is produced using recombinant DNA technology. This involves inserting the gene encoding CD33 into a suitable expression system, such as HEK293 cells, to produce the protein in large quantities . Recombinant CD33 is often tagged with a polyhistidine tag to facilitate purification and detection .

Applications

Recombinant CD33 is widely used in research and therapeutic applications. It is utilized in studies investigating the role of CD33 in immune regulation and its potential as a therapeutic target for diseases such as acute myeloid leukemia (AML) . CD33 is the target of gemtuzumab ozogamicin (Mylotarg®), an antibody-drug conjugate used in the treatment of AML .

Stability and Storage

Recombinant CD33 is typically provided as a lyophilized powder and should be stored at -20°C to -80°C to maintain its stability . It is recommended to aliquot the protein to avoid repeated freeze-thaw cycles, which can degrade the protein .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.