NKp44 Antibody

Natural killer cell p44-related protein, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to NKp44 Antibody

NKp44 (Natural Cytotoxicity Receptor 2, CD336) is a transmembrane glycoprotein belonging to the Natural Cytotoxicity Receptor (NCR) family, which includes NKp30 and NKp46. NKp44 antibodies are tools used to study or modulate the function of this receptor, which is critical for natural killer (NK) cell activation and cytotoxic responses. NKp44 is expressed on activated human NK cells, decidual NK cells, and certain innate lymphoid cells (ILCs), but not on resting NK cells or other leukocytes .

Structure and Function of NKp44

NKp44 is a 44-kDa type I transmembrane protein with:

  • Extracellular domain: A single V-type immunoglobulin-like domain containing a positively charged groove for ligand recognition .

  • Cytoplasmic tail: Contains an immunoreceptor tyrosine-based inhibitory motif (ITIM), but signaling occurs via association with the ITAM-bearing adaptor protein DAP12 .

Key Functions:

FunctionMechanismExample Ligands
Tumor cell lysisRecognition of surface heparan sulfate proteoglycans or viral hemagglutininsInfluenza hemagglutinin, Sendai virus, PCNA
Cytokine releaseCross-linking induces IFN-γ and TNF-α secretionAntibody-mediated activation
Antiviral defenseBinding to viral envelope proteins (e.g., HIV gp41, Dengue virus)Enhanced killing of infected cells

Applications of NKp44 Antibodies in Research

NKp44 antibodies are used to:

Block NKp44 Signaling

  • Tumor cell lysis inhibition: Blocking NKp44 alone partially reduces lytic activity, but combined inhibition of NKp30/NKp46/NKp44 achieves near-complete suppression .

  • Immune checkpoint modulation: The mAb 14-25-9 blocks NKp44-PCNA interactions, enhancing NK cell antitumor responses in PDX models .

Activate NKp44

  • Redirected killing assays: Antibody cross-linking triggers cytotoxic granule release and cytokine production (e.g., IFN-γ) .

  • IFN-γ secretion: Dose-dependent induction in IL-2-activated NK cells (ED₅₀: 0.05–0.2 µg/mL) .

Diagnostic and Analytical Tools

  • Flow cytometry: Detection of NKp44 on activated NK subsets (CD56<sup>dim</sup>CD16<sup>+</sup>, CD56<sup>bright</sup>CD16<sup>−</sup>) .

  • Immunohistochemistry: Localization of NKp44-expressing cells in tissues (e.g., tonsil, decidua) .

Species-Specific Expression

SpeciesNKp44 ExpressionKey Findings
HumanActivated NK cells onlyRestricted to IL-2-stimulated NK cells
PigConstitutively expressed on resting NK cellsUpregulated by IL-2/IL-15; detected in blood, lymph nodes, and non-lymphatic organs

Viral Immune Evasion

  • HIV gp41: Induces NKp44 ligand expression on CD4<sup>+</sup> T cells, promoting NKp44-mediated T cell decline .

  • HIV-3S peptide: Downregulates NKp44 ligand (NKp44L) on astrocytes, protecting them from NK cytotoxicity .

Tumor Microenvironment Interactions

  • NKp44-PCNA axis: PCNA on tumor cells binds NKp44, but mAb 14-25-9 disrupts this interaction, boosting NK cell antitumor activity .

Clinical Relevance and Therapeutic Potential

Therapeutic StrategyExampleOutcome
Immune checkpoint blockademAb 14-25-9 (anti-PCNA)Enhanced IFN-γ secretion, tumor growth inhibition in PDX models
NK cell adoptive therapyNK92-NKp44-1 cell lineIncreased degranulation and cytotoxicity against solid tumors
Combinatorial immunotherapyNKp44 + NKp30/NKp46 blockadeSynergistic tumor lysis

Table 1: NKp44 Antibodies and Their Functions

Antibody CloneTargetApplicationKey FindingsReferences
Z231NKp44NeutralizationPartial inhibition of tumor lysis
44.189 (Thermo Fisher)NKp44Flow cytometryDetects activated NK cells in peripheral blood
14-25-9NKp44-PCNAImmune checkpoint blockadeEnhanced NK antitumor activity in PDX models
MAB22491 (R&D Systems)NKp44IFN-γ secretion assaysDose-dependent cytokine induction

Table 2: Effects of NKp44 Blockade on Tumor Cell Lysis

Experimental ConditionNK Cell ActivityInhibition (%)References
NKp44 alone blockedPartial reduction20–40%
NKp30 + NKp44 + NKp46 blockedNear-complete suppression>90%

Product Specs

Introduction
NCR2 (NKp44) is a member of the natural cytotoxicity receptor (NCR) family found on activated human NK cells. It features a single extracellular Ig-like V domain and a transmembrane region containing a charged lysine residue, potentially involved in interaction with KARAP/DAP12 molecules. The gene encoding NKp44 is located on human chromosome 6p21.1.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The antibody is supplied at a concentration of 1mg/ml in a phosphate-buffered saline (PBS) solution with a pH of 7.4. The solution also contains 10% glycerol and 0.02% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), keep at 4°C. For extended storage, store at -20°C. Repeated freezing and thawing should be avoided.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This NKp44 antibody has been validated for ELISA and Western blot analysis to ensure its specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. A starting dilution of 1:1000 is recommended.
Synonyms
Natural cytotoxicity triggering receptor 2, Natural killer cell p44-related protein, NK cell-activating receptor, Lymphocyte antigen 95 homolog, NK-p44, NKp44, CD336, NCR2, LY95, dJ149M18.1.
Purification Method
NKp44 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT1G6AT.
Immunogen
Anti-human NKp44 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human NKp44 protein 19-130 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and k light chain.

Q&A

What is NKp44 and how does it function in the immune system?

NKp44 is a 44-kD surface molecule belonging to the Natural Cytotoxicity Receptor family that functions as an activating receptor primarily expressed on activated Natural Killer cells. Unlike other activation markers, NKp44 is specifically expressed by activated NK cells but not by activated T lymphocytes or T cell clones, making it the first identified marker specific for activated human NK cells . It plays a crucial role in non-MHC-restricted tumor cell lysis and is involved in the recognition of various transformed cells while reportedly not reacting with normal tissues .

Methodologically, NKp44 function can be studied using anti-NKp44 monoclonal antibodies in redirected killing assays, where antibody-mediated cross-linking results in strong activation of target cell lysis . Masking experiments with anti-NKp44 antibodies can also determine the contribution of this receptor to NK cell-mediated killing of specific target cells. When designing such experiments, researchers should include appropriate controls such as isotype-matched antibodies and assess NKp44 expression by flow cytometry before functional assays.

How is NKp44 expression regulated in NK cells?

NKp44 expression follows a tightly regulated pattern distinct from other activation markers. It is absent in freshly isolated peripheral blood lymphocytes but becomes progressively expressed by all NK cells after culture in IL-2 . This unique expression pattern makes NKp44 a reliable marker for activated NK cells, distinguishing it from other activation markers like CD69 or VLA.2 that can be expressed by both activated NK and T cells .

For researchers investigating NKp44 regulation, time-course experiments monitoring expression during NK cell activation are recommended using flow cytometry with anti-NKp44 antibodies. These experiments should include multiple time points after IL-2 stimulation (typically 24, 48, 72, and 96 hours) to capture the dynamic expression pattern. Additionally, comparing NKp44 expression in response to different cytokines (IL-2, IL-15, IL-21) can provide insights into the specific regulatory mechanisms controlling this receptor.

What are the known ligands for NKp44?

NKp44 interacts with a large and heterogeneous panel of ligands (collectively termed NKp44L) that include:

  • Surface-expressed glycoproteins and proteoglycans

  • Nuclear proteins that can be exposed outside the cell

  • Molecules released in the extracellular space or carried in extracellular vesicles

  • Soluble plasma glycoproteins, including secreted growth factors

  • Extracellular matrix (ECM)-derived glycoproteins

These ligands can be induced upon tumor transformation or viral infection but may also be expressed in normal cells and tissues . The complexity of NKp44L complicates their study, requiring multiple complementary approaches such as mass spectrometry, surface plasmon resonance, and cell-based binding assays to identify and characterize them fully.

When designing experiments to investigate NKp44-ligand interactions, researchers should consider both membrane-bound and soluble ligands, as well as proteins that might be released during cellular stress or death. Various tumor types may express different NKp44 ligands, necessitating comprehensive screening approaches to identify the relevant ligands in specific experimental contexts.

How does NKp44 signal upon engagement?

Upon engagement, NKp44 initiates signaling through its association with KARAP/DAP12, which becomes tyrosine phosphorylated upon NK cell stimulation . This differs from other NCRs like NKp46, which associates with CD3ζ for signal transduction . The NKp44-DAP12 interaction triggers a signaling cascade involving tyrosine kinases that ultimately leads to cytotoxic activity and cytokine production.

To study NKp44 signaling, researchers can employ:

  • Western blotting with phospho-specific antibodies to detect activation of downstream signaling molecules

  • Immunoprecipitation of NKp44 complexes followed by analysis of associated proteins

  • Inhibitor studies to determine requirements for specific kinases

  • CRISPR/Cas9-mediated knockout of pathway components

When designing such experiments, it's essential to include appropriate controls, such as isotype control antibodies and unstimulated NK cells. Time-course analyses (30 seconds to 30 minutes post-stimulation) can capture the dynamic nature of signaling events, while comparison with signaling through other NK receptors can identify NKp44-specific pathways.

What methods can be used to assess NKp44 expression in experimental settings?

Multiple complementary methods can accurately assess NKp44 expression:

  • Flow cytometry:

    • Gold standard for quantifying expression on a per-cell basis

    • Use fluorochrome-conjugated anti-NKp44 antibodies

    • Co-stain with markers for NK cells (CD56, CD16) and activation status (CD69)

    • Include isotype controls and FcR blocking to prevent non-specific binding

  • Quantitative PCR:

    • Measures NKp44 mRNA expression

    • Useful for kinetic studies and correlation with protein expression

    • Requires appropriate housekeeping genes for normalization

  • Western blotting:

    • Detects total NKp44 protein levels

    • Can assess post-translational modifications

    • Less quantitative than flow cytometry for surface expression

  • Immunohistochemistry/Immunofluorescence:

    • Visualizes NKp44+ cells in tissue context

    • Can reveal spatial relationships with other cell types

    • Requires optimization of fixation and antigen retrieval methods

Each method has specific advantages and limitations. Flow cytometry provides the most quantitative assessment of surface expression, while mRNA analysis can detect early changes in expression. For comprehensive analysis, combining multiple methods is recommended to distinguish between transcriptional regulation, protein synthesis, and surface localization of NKp44.

How can NKp44-based chimeric antigen receptors be optimally designed?

Optimal design of NKp44-based CARs requires careful consideration of multiple structural elements. Based on research findings, the most effective NKp44-CAR constructs incorporate:

  • Antigen recognition domain: Extracellular immunoglobulin-like domain of NKp44

  • Hinge region: Native NKp44 hinge performs better than CD8α hinge

  • Transmembrane domain: CD8α transmembrane domain

  • Co-stimulatory domain: 4-1BB signaling domain performs better than CD28

  • Signaling domain: CD3ζ intracellular domain

Table 1: Comparison of NKp44-CAR Construct Designs and Their Performance

CAR DesignExtracellular DomainHingeTransmembraneCo-stimulatory DomainSignaling DomainRelative Performance
1G CARNKp44 Ig-likeNKp44NKp44NoneCD3ζModerate
1G CARNKp44 Ig-likeCD8αNKp44NoneCD3ζPoor
2G CARNKp44 Ig-likeNKp44CD8αCD28CD3ζGood
2G CARNKp44 Ig-likeNKp44CD8α4-1BBCD3ζExcellent

Notably, replacement of the extracellular hinge domain of NKp44 with that of CD8α results in diminished CAR function, highlighting the importance of the native NKp44 hinge for optimal performance . Among second-generation constructs, those incorporating the 4-1BB co-stimulatory domain show superior proliferation upon antigen exposure and better tumor control compared to constructs with the CD28 co-stimulatory domain .

What tumor types have shown sensitivity to NKp44-CAR T cell therapy?

NKp44-CAR T cells demonstrate remarkable versatility in targeting multiple tumor types, offering a potential advantage over single-antigen targeted approaches. Research has demonstrated efficacy against:

Table 2: NKp44-CAR T Cell Efficacy Against Different Tumor Types

Tumor TypeTumor SubtypeDemonstrated ActivityReference
LeukemiaAcute Myeloid Leukemia (AML)Cytotoxicity, cytokine secretion
LeukemiaT-cell Acute Lymphoblastic Leukemia (T-ALL)Cytotoxicity, cytokine secretion
SarcomaSynovial SarcomaSignificant IFN-γ production, antitumor effects
SarcomaOsteosarcomaCytotoxicity
SarcomaEwing SarcomaCytotoxicity
SarcomaRhabdomyosarcomaCytotoxicity
CNS TumorsGlioblastomaCytotoxicity
PediatricNeuroblastomaCytotoxicity

This broad spectrum of activity suggests that NKp44-CAR T cells may represent a promising platform for developing "universal" CAR-T therapies capable of targeting multiple tumor types, potentially addressing the challenge of tumor heterogeneity and antigen escape that limits current single-antigen targeted approaches .

How does the efficacy of NKp44-CAR T cells compare with other CAR-T approaches?

NKp44-CAR T cells demonstrate distinct advantages compared to conventional single-antigen targeted CAR-T approaches:

  • Broader tumor recognition: NKp44-CAR T cells exhibit activity against multiple types of neoplastic cells including hematological malignancies and solid tumors, suggesting potential as a "universal" CAR platform .

  • Efficacy comparison:

    • Second-generation NKp44-CAR T cells with 4-1BB co-stimulatory domain show superior tumor control compared to first-generation CARs and second-generation CARs with CD28 co-stimulatory domain

    • NKp44-CAR T cells exhibit significantly better tumor control in long-term co-culture assays compared with activated NK cells and NK cells transduced with identical NKp44-CAR

  • Proliferative capacity:

    • T cells with second-generation NKp44-CAR containing 4-1BB co-stimulatory domain proliferate at significantly higher levels upon single antigen exposure

    • This enhanced proliferation contributes to improved persistence and sustained anti-tumor activity

When evaluating the efficacy of NKp44-CAR T cells, researchers should include appropriate controls and assess multiple parameters including cytokine production, cytotoxicity, proliferation, and long-term tumor control. The ability to target ligands induced upon malignant transformation across multiple tumor types represents a significant advantage over single-target approaches.

What functional assays are recommended for evaluating NKp44-CAR T cell performance?

A comprehensive assessment of NKp44-CAR T cell performance requires multiple complementary assays:

  • Cytotoxicity assays:

    • Short-term (4-6 hours): Standard chromium release assay or flow cytometry-based killing assays

    • Long-term (7-10 days): Colorimetric assays (e.g., Cell Counting Kit-8) to measure residual viable target cells

    • Real-time cell analysis: Continuous monitoring of target cell viability using impedance-based systems (e.g., iCELLigence)

  • Cytokine secretion assays:

    • Quantification of IFN-γ, TNF-α, and other cytokines in co-culture supernatants using ELISA or cytometric bead array

    • Intracellular cytokine staining to identify cytokine-producing cells by flow cytometry

    • Multiplex cytokine analysis for comprehensive cytokine profiling

  • Proliferation and persistence:

    • CFSE dilution assay to track cell division

    • Long-term co-culture with target cells to assess sustained activity

    • Phenotypic analysis of memory/exhaustion markers over time

  • In vivo models:

    • Xenograft models using immunodeficient mice

    • Bioluminescence imaging to track tumor growth kinetically

    • Analysis of CAR T cell persistence in peripheral blood and tumor tissue

These assays should be performed at multiple effector-to-target ratios (typically 4:1, 2:1, and 1:1) to establish dose-response relationships . When interpreting results, consider comparing the performance of different CAR constructs against the same targets and using non-transduced T cells and irrelevant CAR-T cells as controls.

How can researchers optimize NKp44-CAR T cell production for experimental studies?

Optimizing NKp44-CAR T cell production involves several critical considerations:

  • Vector selection and design:

    • Lentiviral vectors generally provide efficient and stable gene transfer

    • Include reporter genes (e.g., GFP) to track transduction efficiency

    • Optimize promoter selection for consistent CAR expression

  • T cell isolation and activation:

    • Use negative selection to obtain untouched T cells

    • Activate with anti-CD3/CD28 beads or soluble antibodies

    • Consider the timing of activation relative to transduction (typically 24-48 hours)

  • Transduction protocol:

    • Optimize viral titer and multiplicity of infection

    • Consider multiple transduction rounds for higher expression

    • Include polybrene or other enhancers of transduction efficiency

  • Expansion conditions:

    • Supplement media with IL-2 (100-300 IU/ml)

    • Consider adding other cytokines (IL-7, IL-15) for optimal expansion

    • Monitor cell density and replenish media regularly

  • Quality control assessments:

    • CAR expression by flow cytometry (>50% expression typically desired)

    • Viability (>85% viable cells)

    • Phenotypic analysis (CD4/CD8 ratio, memory/effector status)

    • Functional testing against positive control targets

For research applications, smaller-scale production in standard culture flasks or G-Rex vessels is typically sufficient, whereas preclinical validation may require bioreactor-based approaches. Cryopreservation protocols should be optimized to maintain CAR expression and functionality upon thawing.

What controls should be included when evaluating NKp44-antibody specificity?

Rigorous evaluation of NKp44-antibody specificity requires multiple control strategies:

  • Cell type controls:

    • Resting NK cells (NKp44-negative)

    • IL-2 activated NK cells (NKp44-positive)

    • T cells and other lymphocytes (NKp44-negative)

    • NKp44-transfected cell lines versus parental cells

  • Antibody controls:

    • Isotype-matched control antibodies at equivalent concentrations

    • Multiple anti-NKp44 clones targeting different epitopes

    • Blocking with unconjugated antibody before adding conjugated detection antibody

    • F(ab')2 fragments to exclude Fc receptor-mediated effects

  • Experimental controls:

    • Pre-incubation with FcR blocking reagents

    • Competitive binding with recombinant NKp44 protein

    • Secondary-only controls (for indirect detection methods)

    • Fluorescence-minus-one (FMO) controls for flow cytometry

  • Validation approaches:

    • Correlation with NKp44 mRNA expression

    • siRNA or CRISPR knockdown of NKp44

    • Western blot analysis alongside flow cytometry

Implement systematic titration of antibodies to determine optimal concentrations and assess potential cross-reactivity with other surface molecules. Document batch-to-batch variation in antibody performance, as this can significantly impact experimental results.

How can researchers distinguish NKp44-mediated effects from other NK activating pathways?

Distinguishing NKp44-specific effects from other NK activating pathways requires targeted experimental approaches:

  • Receptor-specific stimulation:

    • Use plate-bound or cross-linked anti-NKp44 antibodies for specific triggering

    • Compare with antibodies targeting other NK activating receptors (NKp46, NKG2D)

    • Utilize cell lines expressing defined NKp44 ligands but lacking ligands for other NK receptors

  • Blocking strategies:

    • Selective blocking with F(ab')2 fragments of anti-NKp44 antibodies

    • Combined blocking of multiple receptors to assess synergistic effects

    • Use of receptor-specific antagonists or competitive inhibitors

  • Genetic approaches:

    • siRNA or CRISPR-mediated knockdown of NKp44

    • Transfection with dominant-negative forms of signaling adapters

    • Expression of chimeric receptors containing NKp44 extracellular domain with distinct signaling domains

  • Signaling analysis:

    • Assessment of DAP12 phosphorylation (NKp44-specific)

    • Comparison with CD3ζ phosphorylation (NKp46-associated)

    • Differential inhibition of specific downstream pathways

What factors should be considered when using NKp44 antibodies for immunohistochemistry?

Successful application of NKp44 antibodies in immunohistochemistry (IHC) requires attention to several critical factors:

  • Tissue preparation and fixation:

    • Optimize fixation protocols to preserve NKp44 epitopes (mild formalin fixation typically works best)

    • Consider heat-induced epitope retrieval in citrate or EDTA buffer

    • Compare frozen versus FFPE sections for optimal staining

    • Process tissues promptly to minimize autolysis

  • Antibody selection and validation:

    • Choose clones specifically validated for IHC applications

    • Determine optimal antibody concentration through titration

    • Validate on tissues with known NKp44-positive cells (e.g., tonsil with activated NK cells)

    • Test multiple detection systems (e.g., HRP, AP) for optimal signal-to-noise ratio

  • Protocol optimization:

    • Include blocking steps to reduce background staining

    • Optimize antibody incubation time and temperature

    • Consider signal amplification for tissues with low expression

    • Implement stringent washing steps to reduce non-specific binding

  • Controls and interpretation:

    • Include positive control tissues in each staining run

    • Use isotype control antibodies on serial sections

    • Implement double staining with NK markers (CD56, CD16) for confirmation

    • Consider quantitative image analysis for objective assessment

Remember that NKp44 is expressed only on activated NK cells, so detection in tissues may be limited to sites of active immune responses. The specific microanatomical localization of NKp44-positive cells can provide valuable information about NK cell activation status in different tissue compartments.

How should researchers optimize co-culture conditions for NKp44-CAR T cell functional assays?

Optimization of co-culture conditions is critical for reliable assessment of NKp44-CAR T cell function:

  • Media and supplements:

    • Use complete RPMI-1640 or DMEM supplemented with 10% FBS

    • Add IL-2 (200 IU/mL) to maintain CAR T cell viability

    • Consider serum-free alternatives for specific applications

    • Ensure consistent media lot usage across experiments

  • Cell preparation:

    • Target cells should be in log-phase growth and >90% viable

    • Standardize target cell seeding density (typically 1-2 × 10^5 cells per well for 96-well format)

    • Allow adherent targets to attach for 12-24 hours before adding effectors

    • Ensure single-cell suspensions for non-adherent targets

  • Co-culture parameters:

    • Test multiple effector-to-target ratios (4:1, 2:1, 1:1)

    • Optimize co-culture duration for different readouts:

      • Cytokine measurement: 24 hours

      • Short-term cytotoxicity: 4-6 hours

      • Long-term tumor control: 7-10 days

    • Consider 3D culture systems for solid tumor models

  • Data collection:

    • Sample collection timing should be consistent across experiments

    • For impedance-based assays, establish appropriate sampling intervals

    • For endpoint assays, determine optimal termination time points

    • Include technical replicates (minimum triplicates)

When developing a new assay, perform preliminary time-course and dose-response experiments to identify optimal conditions. Document detailed protocols including cell preparation, media composition, and incubation conditions to ensure reproducibility across experiments.

What approaches can validate the specificity of NKp44-CAR T cell responses?

Validating the specificity of NKp44-CAR T cell responses requires multiple complementary approaches:

  • Target cell panel characterization:

    • Screen diverse cell lines for sensitivity to NKp44-CAR T cells

    • Include NKp44 ligand-positive and negative cell lines

    • Test normal cell counterparts to assess tumor specificity

    • Quantify NKp44 ligand expression levels on target cells

  • Molecular validation:

    • Knockdown/knockout of suspected NKp44 ligands in target cells

    • Overexpression of NKp44 ligands in resistant cells

    • Competitive inhibition with soluble NKp44 protein

    • Blocking with anti-NKp44 antibodies

  • CAR-specific controls:

    • Compare with non-transduced T cells from the same donor

    • Use T cells expressing irrelevant CARs (e.g., CD19-CAR)

    • Test CAR constructs with mutated NKp44 binding domains

    • Employ CAR constructs with non-functional signaling domains

  • Functional validation:

    • Correlate cytotoxicity with NKp44 ligand expression levels

    • Compare cytokine release profiles against different targets

    • Assess activation marker upregulation upon target recognition

    • Evaluate CAR downregulation after target encounter

These approaches should be implemented systematically to establish that observed responses are specifically mediated through the NKp44-CAR and its interaction with cognate ligands on target cells. This comprehensive validation is essential for distinguishing CAR-specific effects from non-specific T cell responses or allogeneic reactions.

How should researchers interpret variations in NKp44-CAR T cell performance across different donors?

Interpreting donor-to-donor variability in NKp44-CAR T cell performance requires systematic analysis:

  • Source of variability:

    • Intrinsic T cell factors (differentiation state, exhaustion profile)

    • Genetic factors (polymorphisms affecting T cell function)

    • Technical factors (transduction efficiency, expansion conditions)

    • Pre-existing immunity to potential NKp44 ligands

  • Analytical approaches:

    • Normalize data to CAR expression level when comparing across donors

    • Calculate fold-change relative to non-transduced T cells from the same donor

    • Use paired statistical tests when comparing different constructs with the same donor cells

    • Consider categorizing donors as high, medium, or low responders

  • Correlation analysis:

    • Correlate performance with donor T cell phenotype (CD4/CD8 ratio, memory/naïve distribution)

    • Assess relationship between transduction efficiency and functional outcomes

    • Investigate correlation between expansion kinetics and final product efficacy

    • Examine association between donor age/sex and CAR T cell function

  • Reporting standards:

    • Report donor number and characteristics (age range, sex distribution)

    • State clearly how many independent donors were tested

    • Present individual donor data points alongside means/medians

    • Specify donor selection criteria and exclusion justifications

When identifying optimal NKp44-CAR constructs, prioritize those that perform consistently across multiple donors rather than those showing exceptional performance with high variability. Consider implementing standardized functional assays with reference standards to facilitate cross-study comparisons.

What statistical methods are appropriate for analyzing NKp44-CAR efficacy data?

Appropriate statistical analysis of NKp44-CAR efficacy data depends on experimental design and data characteristics:

  • For comparing multiple CAR constructs:

    • One-way ANOVA with post-hoc tests (Tukey's or Bonferroni) for normally distributed data

    • Kruskal-Wallis with Dunn's post-hoc test for non-parametric data

    • Repeated measures ANOVA when testing multiple constructs with cells from the same donors

    • Mixed effects models to account for both fixed (CAR design) and random (donor) effects

  • For dose-response relationships:

    • Non-linear regression for E:T ratio titrations

    • Calculate EC50 values to compare potency

    • Area under the curve analysis for comprehensive assessment

    • Two-way ANOVA to compare dose-response curves between CAR constructs

  • For time-course experiments:

    • Repeated measures ANOVA or mixed effects models

    • Area under the curve analysis for cumulative effects

    • Growth curve modeling for tumor control studies

    • Survival analysis for time-to-event data

  • Sample size and power:

    • Conduct power analysis based on preliminary data

    • For donor variability studies, typically 6-10 donors minimum

    • For target cell screening, at least 3-5 biological replicates

    • Technical replicates (typically triplicates) for each condition

When reporting results, include precise p-values, appropriate error bars (SD for technical variation, SEM for biological variation), and clear indication of statistical tests used. Apply multiple testing corrections when making numerous comparisons to control false discovery rates.

How can researchers address discrepancies between in vitro and in vivo NKp44-CAR efficacy?

Addressing discrepancies between in vitro and in vivo NKp44-CAR efficacy requires systematic investigation:

  • Microenvironmental factors:

    • Test CAR function in the presence of immunosuppressive factors (TGF-β, IL-10, PGE2)

    • Evaluate performance in hypoxic conditions mimicking the tumor microenvironment

    • Assess the impact of tumor-associated stromal cells on CAR function

    • Consider extracellular matrix interactions absent in standard in vitro assays

  • Persistence and trafficking:

    • Evaluate CAR T cell persistence using serial sampling in vivo

    • Assess tumor infiltration capacity using immunohistochemistry or flow cytometry

    • Examine expression of relevant chemokine receptors and adhesion molecules

    • Monitor CAR expression stability over time in vivo

  • Target antigen considerations:

    • Verify NKp44 ligand expression in in vivo tumor samples

    • Assess heterogeneity of ligand expression across the tumor

    • Investigate potential antigen downregulation or shedding in vivo

    • Consider competition with endogenous NK cells for ligand binding

  • Experimental approaches:

    • Develop 3D culture models to better recapitulate the tumor microenvironment

    • Implement ex vivo analysis of CAR T cells recovered from tumors

    • Consider humanized mouse models to evaluate interactions with human immune components

    • Use patient-derived xenografts to better represent clinical tumors

When reporting discrepancies, clearly describe both the in vitro and in vivo experimental conditions, discuss potential mechanisms underlying the differences, and propose strategies to improve the predictive value of preclinical models for clinical translation.

How should researchers troubleshoot low or variable NKp44-CAR expression?

Troubleshooting low or variable NKp44-CAR expression requires systematic investigation of multiple factors:

  • Vector and construct issues:

    • Verify plasmid sequence integrity

    • Assess vector titer and quality

    • Test alternative promoters (e.g., EF1α vs. CMV)

    • Evaluate codon optimization of the NKp44 domain

    • Consider the impact of CAR size on packaging efficiency

  • Transduction protocol:

    • Optimize timing of T cell activation before transduction

    • Test different multiplicities of infection

    • Consider multiple transduction rounds

    • Evaluate alternative transduction enhancers

    • Assess impact of cell density during transduction

  • Cell-intrinsic factors:

    • Compare expression in CD4+ versus CD8+ T cell subsets

    • Evaluate impact of donor variability

    • Assess T cell activation status at time of transduction

    • Monitor potential CAR silencing during expansion

  • Detection methodology:

    • Use multiple detection antibodies targeting different CAR components

    • Compare direct (anti-NKp44) versus indirect (anti-Fc tag) detection

    • Implement protein-level confirmation (Western blot) alongside flow cytometry

    • Consider mRNA quantification to distinguish transcriptional from translational issues

  • Experimental approaches:

    • Implement cell sorting to isolate high-expressors

    • Consider inducible vector systems for controlled expression

    • Test bicistronic vectors with reporters to track transduced cells

    • Evaluate CAR protein stability through pulse-chase experiments

When optimization fails to achieve adequate expression, consider redesigning the CAR construct with alternative hinge/transmembrane domains, as the structural configuration significantly impacts surface expression of NKp44-based CARs .

What factors influence the reproducibility of NKp44-CAR T cell manufacturing?

Multiple factors affect the reproducibility of NKp44-CAR T cell manufacturing for research applications:

  • Starting material variability:

    • Donor-to-donor heterogeneity in T cell composition

    • Cryopreservation effects on T cell subsets

    • Prior activation status of T cells

    • Standardize isolation methods and pre-activation protocols

  • Transduction process:

    • Vector lot-to-lot variability

    • Consistency in activation reagents

    • Timing of transduction relative to activation

    • Standardize vector production and quality control testing

  • Culture conditions:

    • Media composition and lot variation

    • Cytokine concentrations and schedule

    • Cell density during expansion

    • Vessel type and gas exchange parameters

    • Implement controlled-rate expansion protocols

  • Quality control:

    • CAR expression level measurement standardization

    • Viability assessment methods

    • Functional testing protocols

    • Implement comprehensive release criteria with accepted ranges

  • Documentation practices:

    • Detailed standard operating procedures

    • Comprehensive batch records

    • Electronic data management systems

    • Training and competency assessment for personnel

To enhance reproducibility, implement automation where possible, establish quality control checkpoints throughout the manufacturing process, and use reference standards to calibrate assays across production runs. Regular proficiency testing and inter-laboratory standardization can further improve consistency for multi-center research collaborations.

Product Science Overview

Introduction

Natural Killer (NK) cells are a type of lymphocyte critical to the innate immune system. They play a significant role in the body’s defense against tumors and virally infected cells. One of the key receptors on NK cells is the NKp44 receptor, also known as Natural Killer Cell p44-Related Protein or CD336. This receptor is involved in the activation and regulation of NK cell functions.

NKp44 Receptor

NKp44 is a transmembrane glycoprotein characterized by a single extracellular V-type Ig-like domain and a cytoplasmic tail containing an Immunoreceptor Tyrosine-based Inhibitory Motif (ITIM) but no known activating signaling motifs . It is encoded by the NCR2 gene and is expressed on activated NK cells. The receptor is involved in recognizing and binding to specific ligands on target cells, leading to the activation of NK cell cytotoxicity and cytokine production.

Mouse Anti-Human NKp44 Antibody

The Mouse Anti-Human NKp44 antibody is a monoclonal antibody developed to specifically detect human NKp44. This antibody is used in various research applications, including flow cytometry, immunohistochemistry, and functional assays . It is produced by immunizing mice with human NKp44 protein and then isolating the specific antibody-producing hybridoma cells.

Applications and Research

The Mouse Anti-Human NKp44 antibody is widely used in research to study NK cell biology and the role of NKp44 in immune responses. It has been shown to induce IFN-gamma secretion in human NK cells activated with recombinant human IL-2 in a dose-dependent manner . This makes it a valuable tool for investigating the mechanisms of NK cell activation and the potential therapeutic applications of targeting NKp44 in cancer and infectious 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.