CD33 Human, Sf9

CD33 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production and Purification

  • Expression: Optimized in Sf9 cells, which enable post-translational modifications absent in prokaryotic systems .

  • Purification: Proprietary chromatographic techniques yield a sterile, colorless solution in phosphate-buffered saline (pH 7.4) .

  • Stability: Requires storage at -20°C with carrier proteins (e.g., 0.1% HSA/BSA) to prevent aggregation .

Functional Properties

  • Ligand Binding: Preferentially binds α-2,6-linked sialic acid, though cis interactions with cell-surface sialoglycans mask its binding site .

  • Immune Modulation:

    • Inhibitory receptor signaling via cytoplasmic SHP-1/SHP-2 phosphatases .

    • Reduces SYK phosphorylation and calcium signaling in microglia, impacting phagocytosis .

  • Disease Relevance:

    • Alzheimer’s Disease: Higher CD33 expression correlates with amyloid-beta accumulation .

    • Acute Myeloid Leukemia (AML): Splice variants (e.g., CD33 ΔE2) evade antibody-based therapies like gemtuzumab ozogamicin .

Table 2: Functional Studies Using CD33 Human, Sf9

Study FocusKey FindingSource
Antibody ValidationClone P67.6 activated CD33 signaling, reducing TREM2-mediated phagocytosis
Splice VariantsCD33 ΔE2 lacks the V-set domain, escaping therapeutic antibody targeting
Reporter AssaysCD33-Fc fusion proteins validated ligand binding to RPTPζ in human brain

Research Applications

  • Therapeutic Development: Used to screen anti-CD33 antibodies (e.g., WM53, P67.6) for AML and Alzheimer’s therapeutics .

  • Ligand Interaction Studies: Identified RPTPζ as a high-affinity ligand in brain sialoglycans .

  • Structural Biology: Facilitated epitope mapping of CD33 isoforms to optimize antibody specificity .

Comparative Analysis with Other Expression Systems

ParameterCD33 (Sf9)CD33 (E. coli)
GlycosylationYesNo
Molecular Weight74.2 kDa29.1 kDa
TagHis-IgGHis-tag
Functional AssaysSuitable for ligand-binding studiesLimited to non-glycosylated domains

Critical Challenges

  • Splice Variants: CD33 ΔE2 and CD33 ΔE2,E7a lack critical epitopes, complicating therapeutic targeting .

  • Autoinhibition: Cis sialic acid interactions limit ligand accessibility in vitro .

Future Directions

  • Isoform-Specific Therapeutics: Develop antibodies targeting conserved domains (e.g., C2-set Ig-like region) .

  • Microglial Targeting: Explore CD33’s role in neuroinflammation using recombinant protein-based assays .

Product Specs

Introduction
CD33, also known as Siglec-3, is a transmembrane receptor expressed on myeloid progenitor cells, mature monocytes, macrophages, and microglial cells. It plays a role in cell adhesion and signaling within the immune system. CD33 recognizes and binds to sialic acid residues on glycans, with a preference for alpha-2,6-linked sialic acid. This interaction can mediate cell-cell adhesion. CD33 also functions as an inhibitory receptor. Upon ligand binding, CD33 becomes tyrosine phosphorylated, leading to the recruitment of cytoplasmic phosphatases. These phosphatases then dephosphorylate downstream signaling molecules, effectively blocking signal transduction pathways. Dysregulation of CD33 function has been implicated in various diseases, including cancer and neurodegenerative disorders. In acute myeloid leukemia (AML), for instance, CD33 is often overexpressed and contributes to disease progression. Notably, CD33 is a therapeutic target in AML, with antibody-drug conjugates like gemtuzumab ozogamicin used to specifically target and eliminate CD33-positive leukemia cells.
Description
CD33, a single-pass type I transmembrane protein, is produced in Sf9 insect cells using baculovirus expression system. This recombinant protein encompasses amino acids 18 to 259 of the extracellular domain of human CD33, fused to a C-terminal His-tag and a human IgG-Fc domain. The resulting protein has a total of 484 amino acids and a molecular weight of approximately 54 kDa. SDS-PAGE analysis under reducing conditions reveals multiple bands within the 50-70 kDa range, likely representing glycosylated forms of the protein. The purification process involves proprietary chromatographic methods to ensure high purity.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The CD33 protein is supplied at a concentration of 0.25 mg/ml in a buffer consisting of phosphate-buffered saline (PBS) at pH 7.4 and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the CD33 protein should be kept refrigerated at 4°C. For long-term storage, it is recommended to freeze the protein at -20°C. To prevent protein degradation during freezing and thawing, it is advisable to add a carrier protein such as albumin (HSA or BSA) at a concentration of 0.1%. Repeated freeze-thaw cycles should be avoided to maintain protein integrity and activity.
Purity
The purity of the CD33 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms

Myeloid cell surface antigen CD33 isoform 1, CD33, FLJ00391, p67, SIGLEC-3, SIGLEC3, gp67.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADLDPNFWLQ VQESVTVQEG LCVLVPCTFF HPIPYYDKNS PVHGYWFREG AIISGDSPVA TNKLDQEVQE ETQGRFRLLG DPSRNNCSLS IVDARRRDNG SYFFRMERGS TKYSYKSPQL SVHVTDLTHR PKILIPGTLE PGHSKNLTCS VSWACEQGTP PIFSWLSAAP TSLGPRTTHS SVLIITPRPQ DHGTNLTCQV KFAGAGVTTE RTIQLNVTYV PQNPTTGIFP GDGSGKQETR AGVVHLEPKS CDKTHTCPPC PAPELLGGPS VFLFPPKPKD TLMISRTPEV TCVVVDVSHE DPEVKFNWYV DGVEVHNAKT KPREEQYNST YRVVSVLTVL HQDWLNGKEY KCKVSNKALP APIEKTISKA KGQPREPQVY TLPPSRDELT KNQVSLTCLV KGFYPSDIAV EWESNGQPEN NYKTTPPVLD SDGSFFLYSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK SLSLSPGKHH HHHH.

Q&A

What is CD33 Human expressed in Sf9 cells and why is it important in research?

CD33 produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain (amino acids 18-259) typically fused to a 239 amino acid hIgG-His Tag at the C-terminus. The complete protein contains 484 amino acids with a molecular mass of approximately 54kDa, though it shows multiple bands between 50-70kDa on SDS-PAGE under reducing conditions due to glycosylation patterns . CD33 is significant in research because it belongs to the sialic acid-binding Ig-like lectin (Siglec) family and is genetically linked to Alzheimer's disease (AD) susceptibility through differential expression of its isoforms in microglia . The protein plays a critical role in modulating inflammatory responses and monocyte activation, making it relevant for both neuroinflammation and immune response studies .

How does the baculovirus expression system benefit CD33 production compared to other expression platforms?

The baculovirus expression vector system (BEVS) utilizing Sf9 insect cells offers several methodological advantages for CD33 production:

  • Post-translational modifications: The system enables proper folding and glycosylation patterns that more closely resemble mammalian modifications than bacterial systems

  • Scale flexibility: The system can be optimized for both small-scale research applications and larger production needs

  • Protein integrity: BEVS produces full-length proteins with appropriate disulfide bond formation and tertiary structure

  • Expression efficiency: When optimized, the system yields higher concentrations of functionally active CD33 compared to mammalian cell expression

For optimal expression, critical parameters include incubation temperature, cell count at infection, multiplicity of infection (MOI), and feeding percentage. Supplementary factors such as cholesterol, polyamines, galactose, and L-glutamine can be strategically incorporated to enhance protein yield and functionality .

What are the critical differences between CD33 isoforms, and how does this affect experimental design?

Research working with CD33 must account for two main isoforms with distinct functions:

  • Long isoform (hCD33M): The full-length protein that contains the sialic acid-binding domain and represses phagocytosis

  • Short isoform (hCD33m): Lacks the sialic acid-binding domain but enhances phagocytosis and is associated with AD protection

When designing experiments, researchers should consider:

  • The specific isoform being expressed in the Sf9 system and whether it matches research objectives

  • Using appropriate antibodies that can distinguish between isoforms

  • Validating expression through multiple methods (Western blot, flow cytometry) as CD33 can display multiple bands (50-70kDa) on SDS-PAGE due to glycosylation patterns

  • Whether to co-express both isoforms to study their competitive interactions, as hCD33m appears dominant over hCD33M in some cellular contexts

How can researchers optimize Sf9 cell culture conditions specifically for CD33 expression?

Optimizing CD33 expression in Sf9 cells requires methodical parameter tuning and experimental design approaches:

  • Initial parameter screening: Implement a Placket-Burman design to identify critical parameters from the following:

    • Physical parameters: incubation temperature (27-29°C), cell count at infection (1-2×10⁶ cells/mL)

    • Infection parameters: multiplicity of infection (MOI) (1-10), time of harvest post-infection

    • Media supplements: cholesterol, polyamines, galactose, pluronic-F68, glucose, L-glutamine, and ZnSO₄

  • Parameter optimization: Apply a Box-Behnken approach to precisely determine optimal values for significant parameters identified in screening

    • Feed percentage optimization (typically 10-30%)

    • Cell density fine-tuning

    • MOI refinement based on protein yield and quality

  • Quality assessment: Verify glycosylation patterns through:

    • Enzymatic deglycosylation assays

    • Lectin binding assays to characterize glycoform distribution

    • Mass spectrometry for precise molecular characterization

Researchers should monitor CD33 expression through Western blotting and ELISA during optimization, noting that the protein appears as multiple bands between 50-70kDa due to variable glycosylation patterns .

What signaling pathways are specifically affected by CD33 isoforms, and how can these be measured in experimental systems?

CD33 isoforms differentially influence several signaling cascades that researchers can quantify through specific methodological approaches:

  • STAT signaling pathways:

    • hCD33M influences STAT5 signaling through hexamer formation

    • hCD33m modulates STAT1 signaling, which affects immediate early gene networks

    Measurement methods: Phospho-specific Western blotting, flow cytometry with phospho-specific antibodies, or STAT-responsive luciferase reporter assays

  • PI3K pathway involvement:

    • CD33 exerts inhibitory functions through PI3K-mediated signaling

    • This pathway is crucial for CD33's repressive effects on monocyte activation

    Measurement methods: PI3K activity assays, AKT phosphorylation status, use of PI3K inhibitors (e.g., wortmannin) to parse pathway contributions

  • p38 MAPK signaling requirement:

    • p38 MAPK signaling is required for cytokine production (IL-1β) during CD33 manipulation

    • Inhibition of p38 MAPK abolishes inflammatory responses

    Measurement methods: p38 phosphorylation Western blots, cytokine ELISAs coupled with pathway inhibitors, kinase activity assays

For comprehensive pathway analysis, researchers should consider multiplexed approaches such as phospho-proteomics or CyTOF to capture signaling dynamics across pathways simultaneously.

How do glycosylation patterns of CD33 produced in Sf9 cells differ from mammalian-expressed CD33, and what are the functional implications?

The glycosylation of CD33 in Sf9 cells follows insect-specific patterns that differ from mammalian systems in important ways:

  • Structural differences:

    • Sf9-produced CD33 contains predominantly high-mannose and paucimannose N-glycans

    • Mammalian CD33 features complex N-glycans with terminal sialic acids

    • Sf9 cells lack the ability to produce complex sialylated glycans unless engineered

  • Functional implications:

    • Sialic acid binding is crucial for CD33's inhibitory function on monocyte activation

    • Altered glycosylation may affect CD33's ability to bind sialic acid ligands

    • The lack of terminal sialylation in Sf9-produced CD33 may impact self-recognition mechanisms

  • Methodological approaches to address glycosylation differences:

    • Humanized Sf9 cell lines with enhanced glycosylation capabilities

    • In vitro enzymatic modification of purified CD33 to add complex glycans

    • Comparative functional assays between Sf9-produced and mammalian-produced CD33

When interpreting binding or functional assays with Sf9-expressed CD33, researchers should account for these glycosylation differences, especially when studying interactions dependent on sialic acid recognition .

What critical controls should be included when studying CD33 isoform functions in relation to Alzheimer's disease?

Robust experimental design for CD33 studies in Alzheimer's disease contexts requires:

  • Genetic controls:

    • Inclusion of cells expressing CD33 variants associated with rs12459419T (protective allele) and rs12459419C (susceptibility allele)

    • CRISPR/Cas9-generated CD33 knockout controls to establish baseline responses

    • Rescue experiments with specific isoforms to confirm phenotype attribution

  • Functional validation controls:

    • Phagocytosis assays using standardized substrates (e.g., fluorescent beads, amyloid-β)

    • Cytokine production measurements with and without CD33 manipulation

    • Side-by-side comparison of hCD33m and hCD33M expression effects

  • Signaling pathway controls:

    • Selective inhibition of suspected downstream pathways (PI3K, p38 MAPK)

    • Phosphorylation state analysis of key signaling molecules

    • Combined inhibitor treatments to parse pathway interactions

  • Ligand interaction controls:

    • Neuraminidase treatment to remove sialic acids

    • Competition assays with sialyllactosamine vs. lactosamine

    • Red blood cell addition as a source of sialic acid ligands

These controls help distinguish between isoform-specific effects and experimental artifacts, particularly important when working with the heterologous Sf9 expression system.

How can researchers effectively distinguish between CD33 isoforms in experimental systems and clinical samples?

Distinguishing between CD33 isoforms requires specialized methodological approaches:

  • Molecular distinction techniques:

    • RT-PCR with isoform-specific primers spanning exon junctions

    • Digital droplet PCR for absolute quantification of isoform ratios

    • RNA-seq analysis with specific attention to exon 2 splicing events

  • Protein detection methods:

    • Western blotting with antibodies targeting domains specific to each isoform

    • Use of isoform-specific antibodies (such as anti-hCD33m antibodies)

    • Immunoprecipitation followed by mass spectrometry for complex samples

  • Functional discrimination approaches:

    • Phagocytosis assays (enhanced with hCD33m, repressed with hCD33M)

    • Analysis of immediate early gene activation (enhanced in hCD33m+ cells)

    • Subcellular localization analysis (hCD33m shows preferential intracellular localization)

  • Single-cell analysis techniques:

    • scRNA-seq to identify cell clusters with differential isoform expression

    • CyTOF with isoform-specific antibodies

    • In situ validation within tissue samples

These techniques allow researchers to not only quantify isoform distribution but also correlate isoform expression with functional outcomes and disease phenotypes.

What methods are available for specifically manipulating CD33 expression or function in experimental systems?

Researchers have several sophisticated options for CD33 manipulation:

  • Genetic manipulation approaches:

    • CRISPR/Cas9 gene editing to create complete knockouts or specific isoform expression

    • siRNA/shRNA for transient or stable knockdown (demonstrated efficacy in monocyte systems)

    • Overexpression systems with isoform-specific constructs

    • Splicing modulators to shift isoform ratios

  • Protein-level manipulation:

    • Anti-CD33 monoclonal antibodies (can induce proinflammatory cytokine production)

    • Sialic acid removal from cell surfaces using neuraminidase

    • Competitive ligands (sialyllactosamine vs. lactosamine)

    • Small molecule inhibitors of CD33-ligand interactions

  • Signaling pathway interventions:

    • PI3K inhibitors (enhance IL-1β response)

    • p38 MAPK inhibitors (abolish cytokine production)

    • Combined pathway manipulations to parse complex interactions

  • Environmental manipulations:

    • Modulation of sialic acid environments

    • Co-culture systems with varying cell types

    • Inflammatory stimulus challenges with LPS or other activators

Each approach offers unique advantages and should be selected based on the specific research question regarding CD33 function.

How can researchers accurately assess the functional consequences of CD33 isoform expression in microglia and other myeloid cells?

To comprehensively evaluate CD33 isoform functionality, researchers should employ multiple complementary assays:

  • Phagocytosis assessment:

    • Fluorescent bead uptake quantification by flow cytometry

    • Amyloid-β phagocytosis assays for AD relevance

    • Time-lapse microscopy to capture dynamic phagocytic processes

    • Quantitative image analysis of phagocytic cup formation

  • Inflammatory response profiling:

    • Multiplex cytokine assays (IL-1β, TNF-α, IL-8)

    • Transcriptional profiling of inflammatory gene networks

    • Inflammasome activation assessment

    • NF-κB pathway activation analysis

  • Cellular phenotype characterization:

    • Immediate early gene expression network analysis

    • Cell morphology and activation state quantification

    • Surface marker expression profiles

    • Migration and chemotaxis assays

  • Pathway activation verification:

    • Phospho-flow cytometry for STAT1, STAT5 activation

    • Immunoblotting for PI3K and p38 MAPK pathway components

    • Transcription factor nuclear localization

    • Proximity ligation assays to detect protein-protein interactions

This comprehensive functional assessment helps establish causal relationships between CD33 isoform expression and phenotypic outcomes in myeloid cells.

How can findings from CD33 studies in Sf9 systems be effectively translated to human disease contexts?

Translating findings from Sf9-expressed CD33 to human disease relevance requires careful methodological bridging:

  • Validation across expression systems:

    • Parallel studies with CD33 expressed in human cell lines

    • Primary human microglia or monocyte confirmatory experiments

    • iPSC-derived microglia models with CD33 genetic modifications

  • Genetic correlation validation:

    • Analysis of CD33 polymorphism effects (rs12459419) in patient cohorts

    • Correlation of isoform ratios with disease progression

    • Integration with genome-wide association study (GWAS) data

  • Functional validation in disease-relevant assays:

    • Amyloid-β clearance studies

    • Neuroinflammatory response characterization

    • Ex vivo studies with patient-derived samples

  • Therapeutic application exploration:

    • CD33-targeting antibody effects on isoform-specific functions

    • Small molecule modulation of CD33 splicing

    • Pathway intervention strategies based on isoform mechanisms

By systematically validating findings across these domains, researchers can establish the translational relevance of mechanistic insights gained from CD33 studies in Sf9 systems.

What are the current contradictions or unresolved questions in CD33 research that require further investigation?

Several critical knowledge gaps and conflicting findings warrant focused research:

  • Mechanistic uncertainties:

    • Whether hCD33m represents a loss-of-function or gain-of-function variant remains contested

    • The precise molecular mechanisms by which hCD33m enhances phagocytosis while hCD33M represses it

    • How CD33 isoform ratios are regulated in response to environmental cues

  • Contradictory findings:

    • Anti-CD33 antibodies can induce proinflammatory cytokines despite CD33's reported inhibitory role

    • The relationship between sialic acid binding and intracellular signaling remains incompletely understood

    • Different experimental systems show variable dependencies on PI3K vs. MAPK pathways

  • Technical challenges:

    • The lack of truly selective antibodies for specific isoforms

    • Limited availability of physiologically relevant microglia models

    • Difficulties in simultaneously measuring multiple signaling pathways

  • Translational uncertainties:

    • Whether CD33-targeting therapeutic approaches should aim to enhance or inhibit its function

    • How CD33's role in microglia differs from its role in other myeloid cells

    • The long-term consequences of modulating CD33 activity in the context of neuroinflammation

Addressing these questions will require integrated approaches combining genetic, biochemical, and cellular methodologies across multiple model systems.

What bioinformatic and computational methods are most effective for analyzing CD33 isoform expression and function?

Advanced computational approaches offer powerful tools for CD33 research:

  • Transcriptomic analysis strategies:

    • RNA-seq with splicing-aware algorithms to quantify exon 2 inclusion/exclusion

    • Single-cell transcriptomics to identify cell populations with distinct isoform distributions

    • Differential expression analysis focused on immediate early gene networks associated with CD33 isoforms

  • Network analysis methods:

    • Gene regulatory network reconstruction to identify transcription factors mediating CD33 isoform effects

    • Pathway enrichment analysis to contextualize CD33-dependent transcriptional changes

    • Protein-protein interaction network analysis to identify novel CD33 binding partners

  • Structural biology approaches:

    • Molecular dynamics simulations to predict isoform-specific conformational differences

    • Protein-ligand docking simulations for CD33-sialic acid interactions

    • Structure-based virtual screening for potential CD33 modulators

  • Integrative data analysis:

    • Multi-omics integration (transcriptomics, proteomics, metabolomics)

    • Machine learning approaches to identify patterns associated with CD33 function

    • Systems biology models of CD33-dependent myeloid cell activation states

These computational approaches complement experimental data and help generate new hypotheses about CD33 function in health and disease.

How can researchers accurately quantify and characterize different CD33 glycoforms and their functional implications?

Glycosylation characterization requires specialized analytical techniques:

  • Mass spectrometry approaches:

    • Glycopeptide analysis using LC-MS/MS with electron transfer dissociation

    • MALDI-TOF analysis of released N-glycans

    • Site-specific glycosylation mapping

    • Intact protein mass analysis to determine glycoform heterogeneity

  • Chromatographic methods:

    • Hydrophilic interaction chromatography for glycan separation

    • Lectin affinity chromatography to enrich specific glycoforms

    • Size exclusion chromatography to separate different glycoform populations

  • Functional correlation techniques:

    • Glycoform-specific binding assays using surface plasmon resonance

    • Cell-based assays comparing differentially glycosylated CD33 preparations

    • Neuraminidase treatment combined with functional readouts

  • Visualization methods:

    • Lectin staining with glycan-specific lectins

    • Metabolic labeling of glycans with bioorthogonal handles

    • Super-resolution microscopy to visualize CD33 glycoform distribution

Understanding glycosylation patterns is particularly critical when working with CD33, as its sialic acid-binding properties and inhibitory functions depend on proper glycan structures and recognition .

Product Science Overview

Structure and Expression

CD33 is a transmembrane protein predominantly expressed on the surface of myeloid cells, including monocytes, macrophages, and myeloid progenitor cells. The protein consists of an extracellular domain that binds to sialic acids, a single transmembrane region, and an intracellular domain that contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs). These ITIMs are essential for the inhibitory signaling functions of CD33.

Function

CD33 functions as an inhibitory receptor in the immune system. Upon binding to its ligands, which are typically sialic acid-containing glycoproteins, CD33 undergoes tyrosine phosphorylation. This phosphorylation recruits cytoplasmic phosphatases, such as SHP-1 and SHP-2, which dephosphorylate signaling molecules and inhibit cellular activation. This mechanism helps regulate immune responses and maintain immune homeostasis.

In addition to its role in immune regulation, CD33 has been implicated in the pathogenesis of acute myeloid leukemia (AML). CD33 is expressed on the surface of leukemic blasts in most AML patients, making it a target for therapeutic interventions. Antibody-drug conjugates targeting CD33, such as gemtuzumab ozogamicin, have been developed for the treatment of AML.

Recombinant CD33 (Human, Sf9)

Recombinant CD33 protein is produced using the baculovirus expression system in Sf9 insect cells. This system allows for the production of glycosylated proteins that closely resemble their native forms. The recombinant CD33 protein typically consists of the extracellular domain of CD33 fused to a tag, such as a His-tag, to facilitate purification and detection.

The recombinant CD33 protein produced in Sf9 cells is a single, glycosylated polypeptide chain with a molecular mass of approximately 54 kDa . It is purified using chromatographic techniques to achieve high purity and is often used in research applications to study the structure, function, and interactions of CD33.

Applications

Recombinant CD33 protein is widely used in various research applications, including:

  • Structural studies: Understanding the three-dimensional structure of CD33 and its interactions with ligands.
  • Functional assays: Investigating the signaling pathways and inhibitory functions of CD33.
  • Drug development: Screening and characterizing potential therapeutic agents targeting CD33 for the treatment of AML and other diseases.

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.