KIR2DL3 Human

Killer Cell Immunoglobulin-Like Receptor, 2 Domains Long Cytoplasmic Tail 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function

KIR2DL3 is encoded by the KIR gene cluster on chromosome 19q13.4 within the Leukocyte Receptor Complex (LRC) . Key features include:

  • Domains: Two extracellular immunoglobulin (Ig) domains and a long cytoplasmic tail containing an immunoreceptor tyrosine-based inhibitory motif (ITIM) .

  • Ligand Specificity: Binds HLA-C1 allotypes via residues centered at position 80 (Asn80) .

  • Signal Transduction: Ligand binding triggers ITIM phosphorylation, recruiting phosphatases (e.g., SHP-1/SHP-2) to inhibit NK cell activation .

Table 1: Comparison of KIR2DL3 with Related Receptors

FeatureKIR2DL3KIR2DL2KIR2DL1
Ligand GroupHLA-C1HLA-C1HLA-C2
Cytoplasmic TailLong (ITIM-containing)Long (ITIM-containing)Long (ITIM-containing)
Inhibitory StrengthModerateStrongStrong
Allelic DiversityHigh (e.g., *005, *015)ModerateModerate
Data derived from .

Genetic Polymorphism and Allelic Variants

KIR2DL3 exhibits significant allelic diversity, influencing receptor-ligand interactions and clinical outcomes:

  • KIR2DL3*005: Fails to react with ECM41 monoclonal antibody due to structural variations in the D1 domain .

  • KIR2DL3*015: A novel allele identified in donors with discrepancies between genotypic and phenotypic KIR expression .

  • Population Genetics: The KIR2DL3-HLA-C1 combination is less frequent in malaria-endemic regions, likely due to selective pressure against cerebral malaria susceptibility .

Infectious Disease

  • Cerebral Malaria: A genome-wide association study found that individuals with both KIR2DL3 and HLA-C1 had a 3.14-fold higher risk of cerebral malaria (95% CI: 1.52–6.48; P = 0.00079) .

  • Viral Infections: KIR2DL3+ T cells exhibit reduced degranulation against cytomegalovirus (CMV)-infected autologous cells, suggesting immunomodulatory roles in viral persistence .

Transplantation and Cancer

  • Hematopoietic Stem Cell Transplantation (HSCT): Elevated KIR2DL3+ T cell frequencies post-transplant correlate with relapse risk, possibly due to inhibited anti-leukemic responses .

  • NK Cell Education: KIR2DL3-HLA-C1 interactions shape NK cell tolerance, affecting responses to malignancies .

Research Tools and Recombinant Proteins

  • Recombinant KIR2DL3:

    • E. coli-derived: 22.2 kDa non-glycosylated protein (amino acids 23–223) .

    • HEK293-derived: Biotinylated Fc-fusion protein (53 kDa, ≥90% purity) for ligand-binding assays .

Functional Studies and Mechanistic Insights

  • Ligand Binding Avidity: KIR2DL3 exhibits weaker inhibition of HLA-C2 compared to KIR2DL2, suggesting a higher specificity threshold for C1 .

  • Synergistic Polymorphisms: Residues 71 and 131 in the D0-D2 domains collectively enhance HLA-C1 binding, impacting NK cell education .

Evolutionary Perspectives

The KIR2DL3-HLA-C1 combination has undergone negative selection in malaria-endemic populations, likely due to its association with cerebral malaria pathogenesis . This highlights the interplay between host genetics and infectious disease susceptibility.

Future Research Directions

  • Allele-Specific Functional Studies: Characterize novel alleles (e.g., *015) in diverse populations.

  • Therapeutic Targeting: Explore blockade of KIR2DL3-HLA-C1 interactions to mitigate cerebral malaria or enhance anti-tumor immunity.

Product Specs

Introduction
Killer-cell immunoglobulin-like receptors (KIRs) are proteins found on Natural Killer (NK) cells, which are part of the immune system. KIRs regulate NK cell activity by interacting with MHC class I molecules present on all cells. This interaction helps NK cells identify infected or cancerous cells with low MHC class I levels. Most KIRs are inhibitory, meaning they suppress NK cell activity upon binding to MHC. A few KIRs can activate NK cells. KIR genes are located on chromosome 19q13.4 and exhibit high polymorphism, resulting in diverse KIR gene profiles among individuals. KIR proteins are classified by their extracellular immunoglobulin domains (2D or 3D) and cytoplasmic tail length (long (L) or short (S)). KIRs with long cytoplasmic tails contain an ITIM motif that inhibits NK cell activity upon ligand binding. In contrast, KIRs with short cytoplasmic tails lack the ITIM motif and instead activate NK cells through the TYRO protein tyrosine kinase binding protein. KIR2DL3 is an inhibitory KIR that recognizes MHC class I molecules (HLA-Cw1, -Cw3, -Cw7, and Cw8) and suppresses NK cell activity to prevent cell lysis.
Description
Recombinant KIR2DL3, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising amino acids 23-223. It has a molecular weight of 22.2 kDa. KIR2DL3 is purified using specialized chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The protein solution (1mg/ml) is prepared in 25mM Tris-HCl buffer at pH 7.5.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Repeated freezing and thawing should be avoided.
Purity
The purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Killer cell immunoglobulin-like receptor 2DL3, MHC class I NK cell receptor, Natural killer-associated transcript 2, NKAT-2, NKAT2a, NKAT2b, p58 natural killer cell receptor clone CL-6, p58 NK receptor, p58.2 MHC class-I-specific NK receptor, Killer inhibitory receptor cl 2-3, KIR-023GB, CD158 antigen-like family member B2, CD158b2 antigen, KIR2DL3, CD158B2, KIRCL23, NKAT2, p58, NKAT, GL183, CD158b, KIR-K7b, KIR-K7c, MGC129943.
Source
Escherichia Coli.
Amino Acid Sequence

MEGVHRKPSL LAHPGPLVKS EETVILQCWS DVRFQHFLLH REGKFKDTLH LIGEHHDGIS KANFSIGPMM QDLAGTYRCY GSVTHSPYQL SAPSDPLDIV ITGLYEKPSL SAQPGPTVLA GESVTLSCSS RSSYDMYHLS REGEAHERRF SAGPKVNGTF QADFPLGPAT HGGTYRCFGS FRDSPYEWSN SSDPLLVSVT GN.

Q&A

What is KIR2DL3 and what is its primary function in human immunity?

KIR2DL3 is an inhibitory killer-cell immunoglobulin-like receptor that regulates the activation of natural killer (NK) cells by interacting with human leukocyte antigen-C1 (HLA-C1) group molecules. It acts as a critical checkpoint in the NK cell activation pathway, providing inhibitory signals when engaged with its HLA-C1 ligands. This interaction maintains self-tolerance and prevents inappropriate NK cell activation against healthy cells. KIR2DL3 functions within a complex network of activating and inhibitory receptors that collectively determine NK cell responsiveness to various cellular targets, including virally infected or transformed cells .

How does KIR2DL3 differ structurally and functionally from its close relative KIR2DL2?

While KIR2DL2 and KIR2DL3 share approximately 94% sequence identity and segregate as alleles of a single locus, they exhibit significant structural and functional differences:

What is the genomic organization of KIR2DL3 and how is it inherited?

KIR2DL3 is encoded within the KIR gene complex located on chromosome 19q13.4. It segregates as an allele of a single locus with KIR2DL2, but these receptors are inherited on different KIR haplotypes. KIR2DL3 is typically found on KIR haplotype A, while KIR2DL2 is associated with haplotype B. This difference in haplotype inheritance has significant functional implications, as other KIR genes in linkage disequilibrium with either KIR2DL2 or KIR2DL3 can influence NK cell function. Notably, KIR2DS2 is in linkage disequilibrium with KIR2DL2 but not with KIR2DL3, creating distinct receptor combinations that affect NK cell responsiveness to various targets. The inheritance pattern of these receptors contributes to the diversity of NK cell responses across different individuals and populations .

What are the recommended methods for expression and purification of KIR2DL3 for structural studies?

For high-quality structural studies of KIR2DL3, the following methodology has proven effective:

  • Cloning: Clone the extracellular D1-D2 domains (residues 1-204) of KIR2DL3*001 into an expression vector such as pET-30(b) for bacterial expression .

  • Expression system: Use Escherichia coli BL21(DE3) strain for protein expression, which typically results in the formation of inclusion bodies .

  • Refolding protocol:

    • Extract inclusion bodies and solubilize them in denaturing conditions

    • Perform refolding by rapid dilution in a buffer containing 100 mM Tris-HCl pH 8.0, 400 mM L-arginine-HCl, 5 mM reduced glutathione, and 0.5 mM oxidized glutathione

    • Allow refolding to proceed for 72 hours at controlled temperature

  • Purification strategy:

    • Initial purification using diethylaminoethyl (DEAE) cellulose column chromatography

    • Size exclusion chromatography using a Superdex 200 16/60 column

    • Final polishing via anion exchange chromatography using a Hi-Trap Q HP 5 ml column

This approach yields properly folded, homogeneous KIR2DL3 protein suitable for crystallization trials and other structural studies. Protein quality should be assessed by SDS-PAGE, size exclusion profiles, and circular dichroism to ensure proper folding before proceeding to structural analyses.

How can researchers effectively study the interaction between KIR2DL3 and various HLA-C allotypes?

To comprehensively characterize KIR2DL3 interactions with HLA-C allotypes, researchers should employ a multi-faceted approach:

  • Surface Plasmon Resonance (SPR):

    • Express and purify KIR2DL3 and various HLA-C allotypes

    • Immobilize either the receptor or ligand on a sensor chip

    • Determine binding kinetics and affinity constants (ka, kd, KD)

    • Compare relative binding across multiple HLA-C allotypes

    • Evaluate how peptide substitutions affect binding

  • Crystallographic studies:

    • Co-crystallize KIR2DL3 with HLA-C molecules loaded with specific peptides

    • Determine structures at high resolution (preferably <2.5 Å)

    • Analyze binding interfaces and key contact residues

    • Compare docking geometries across different complexes

  • Cellular functional assays:

    • Use primary NK cells expressing KIR2DL3 in degranulation assays (CD107a)

    • Co-culture with target cells expressing different HLA-C allotypes

    • Compare inhibitory capacities of different HLA-C molecules

    • Include appropriate controls (e.g., HLA-deficient cells like 721.221)

  • Mutagenesis studies:

    • Create point mutations at key interface residues

    • Express mutants in cellular systems

    • Assess impacts on binding and functional outcomes

    • Use these data to validate structural observations

By combining these complementary approaches, researchers can develop a comprehensive understanding of the specificity, affinity, and functional consequences of KIR2DL3 interactions with different HLA-C allotypes.

What flow cytometry panels and protocols are optimal for identifying and analyzing KIR2DL3-expressing NK cells?

For optimal identification and analysis of KIR2DL3-expressing NK cells, the following flow cytometry approach is recommended:

  • Antibody panel selection:

    • Anti-CD3 and anti-CD56 antibodies to identify NK cells (CD3-CD56+)

    • Anti-KIR2DL3-specific antibody (e.g., REA147 clone)

    • Anti-KIR2DL2/L3/S2 (e.g., GL183 clone)

    • Anti-KIR2DL3/S2 (e.g., 1F12 clone)

    • Anti-KIR2DL1/S1 to exclude these populations

    • Additional functional markers (e.g., CD107a for degranulation)

  • Gating strategy:

    • Exclude debris and doublets

    • Gate on CD3-CD56+ NK cells

    • Identify KIR2DL3+ cells as KIR2DL1-KIR2DL2-KIR2DL3+

    • Distinguish KIR2DL3+ from KIR2DL2+/S2- cells using antibody combinations (GL183+REA147+ vs. GL183+REA147-)

  • Functional assessment:

    • Co-culture NK cells with target cells (e.g., 721.221 cells or HLA-C transfectants)

    • Stain for CD107a to measure degranulation

    • Compare functional responses between different KIR-expressing populations

  • Controls and validation:

    • Include KIR-negative NK cell populations as controls

    • Use donors with known KIR genotypes as reference samples

    • Include HLA-I deficient target cells (e.g., 721.221) and HLA-C transfectants

    • Consider including NK cells from individuals homozygous for KIR2DL3 or KIR2DL2/S2 as controls

This comprehensive approach enables accurate identification of KIR2DL3-expressing NK cells and assessment of their functional responses to different stimuli.

How do structural differences between KIR2DL2 and KIR2DL3 impact their recognition of HLA-C molecules?

The structural differences between KIR2DL2 and KIR2DL3 significantly impact their recognition of HLA-C molecules in several ways:

  • Docking geometry differences:

    • Despite high sequence similarity (~94%), KIR2DL2 and KIR2DL3 exhibit distinct docking modes when binding to HLA-C*07:02

    • The twist differences between their D1 (13.3°) and D2 (10.4°) domains alter the contact interface with HLA-C molecules

    • These differences in receptor orientation influence the precise interactions at the binding interface

  • HLA-C allotype recognition patterns:

    • KIR2DL3 shows clearer discrimination between HLA-C1 and HLA-C2 allotypes

    • KIR2DL3 exhibits higher average relative avidity (82.2%) across HLA-C1 allomorphs compared to KIR2DL2 (68.3%)

    • Individual HLA-C allomorphs demonstrate preferences for binding either KIR2DL2 or KIR2DL3

    • For example, HLA-C08:01 and HLA-C12:03 bind considerably better to KIR2DL3, while HLA-C*07:02 is bound with higher avidity by KIR2DL2

  • Functional consequences:

    • The structural differences translate to distinct functional outcomes in NK cell responses

    • NK cells expressing KIR2DL3 show different patterns of inhibition when encountering various HLA-C1-expressing target cells compared to KIR2DL2-expressing NK cells

    • These differences may contribute to the associations of KIR2DL3 (but not KIR2DL2) with specific disease outcomes

Understanding these structure-function relationships is crucial for interpreting how genetic variation in these receptors impacts NK cell function and associated disease outcomes.

What is the current understanding of KIR2DL3's role in viral clearance and autoimmune diseases?

KIR2DL3 has emerged as a significant factor in both viral clearance and autoimmune disease progression:

  • Viral clearance:

    • KIR2DL3, in the presence of HLA-C1 alleles, has been associated with increased clearance of hepatitis C virus (HCV) infection

    • This association is not observed with KIR2DL2, despite their high sequence similarity

    • The mechanism likely involves differences in inhibitory signaling strength, with KIR2DL3-HLA-C1 interactions potentially providing weaker inhibition than KIR2DL2-HLA-C1, allowing for more effective NK cell responses against infected cells

  • Autoimmune diseases:

    • KIR2DL3 has been associated with the progression of ulcerative colitis in the presence of HLA-C1 alleles

    • This association suggests that the KIR2DL3-HLA-C1 interaction influences inflammatory responses in the intestinal mucosa

    • The specific mechanism may involve altered NK cell regulation of inflammatory processes

  • Mechanistic insights:

    • The distinct binding properties of KIR2DL3 to different HLA-C1 allotypes likely contribute to these disease associations

    • Polymorphism in both the receptor and its HLA-C ligands creates a complex network of interactions with varying inhibitory potential

    • The quality of inhibitory signaling, rather than simple presence/absence of the receptor, appears to be crucial

    • The genetic linkage of KIR2DL3 and KIR2DL2 with other KIR genes (e.g., KIR2DS2 is linked with KIR2DL2 but not KIR2DL3) may also contribute to these differential disease associations

Future research should focus on understanding how these receptor-ligand interactions specifically modulate NK cell responses in different disease contexts.

How do peptides presented by HLA-C molecules influence KIR2DL3 binding and function?

Peptides presented by HLA-C molecules significantly influence KIR2DL3 binding and function through multiple mechanisms:

  • Peptide positions influencing binding:

    • Positions 7 (P7) and 8 (P8) of the peptide presented by HLA-C are particularly critical for KIR2DL3 recognition

    • These residues directly contact the KIR receptor or influence the HLA conformation at the binding interface

    • Substitutions at these positions can dramatically alter binding affinity

  • Amino acid preferences:

    • Acidic residues (e.g., Glu) at P7 or P8 are generally detrimental to KIR2DL3 binding

    • Certain substitutions (e.g., P8F, P8V) may maintain or even enhance binding

    • These peptide preferences are similar between KIR2DL2 and KIR2DL3, at least in the context of HLA-C*07:02-RL9

  • Peptide-dependent functional outcomes:

    • Different peptides presented by the same HLA-C allotype can alter inhibitory signaling strength

    • This "peptide selectivity" creates a spectrum of NK cell inhibition depending on the specific peptide-HLA complex

    • This mechanism allows for fine-tuned recognition of altered self (e.g., virally infected cells presenting viral peptides)

  • Experimental approaches to study peptide effects:

    • Surface Plasmon Resonance (SPR) with peptide-loaded HLA-C molecules

    • Crystal structures of KIR2DL3 bound to HLA-C presenting different peptides

    • NK cell functional assays with target cells presenting defined peptides

Understanding peptide influences on KIR2DL3-HLA-C interactions is crucial for comprehending how NK cells discriminate between healthy and diseased cells, as changes in the peptide repertoire during infection or transformation may alter inhibitory signaling through KIR2DL3.

What strategies can overcome the challenges in distinguishing between KIR2DL2 and KIR2DL3 in functional studies?

Distinguishing between KIR2DL2 and KIR2DL3 in functional studies presents significant challenges due to their high sequence similarity and cross-reactivity of most available antibodies. Here are effective strategies to overcome these limitations:

  • Specialized antibody combinations:

    • Use a combination of antibodies: GL183 (recognizes KIR2DL2/L3/S2), REA147 (specific for KIR2DL3), and 1F12 (recognizes KIR2DL3/S2)

    • This panel allows discrimination between KIR2DL3+, KIR2DL2+KIR2DS2-, and KIR2DS2+ populations

  • Donor selection based on genotyping:

    • Perform KIR genotyping to identify donors who are homozygous for KIR2DL3 or KIR2DL2/S2

    • Use these genotyped donors as reference controls for antibody validation

    • Include heterozygous donors (KIR2DL2+/KIR2DL3+) for comparative studies

  • Functional readout optimization:

    • Utilize degranulation assays (CD107a surface expression) combined with KIR-specific staining

    • Compare responses to standard target cells (e.g., 721.221 and HLA-C transfectants)

    • Analyze functional data in the context of KIR expression patterns

  • Exclude confounding factors:

    • Account for the presence of KIR2DS2 (often in linkage disequilibrium with KIR2DL2)

    • Separate analyses for KIR2DL2+KIR2DS2- and KIR2DL2+KIR2DS2+ populations

    • Control for other inhibitory receptors that might influence functional outcomes

By implementing these approaches, researchers can reliably distinguish between NK cells expressing KIR2DL2 and KIR2DL3, enabling more accurate assessment of their specific functional properties.

What are the best approaches for analyzing the impact of KIR2DL3 polymorphisms on HLA-C binding?

To effectively analyze the impact of KIR2DL3 polymorphisms on HLA-C binding, researchers should employ a multi-dimensional approach:

  • Structural analysis and prediction:

    • Analyze existing crystal structures to identify key interface residues

    • Use computational modeling to predict the effects of specific polymorphisms

    • Identify polymorphic sites that might directly contact HLA-C or affect receptor conformation

  • Site-directed mutagenesis:

    • Generate a panel of KIR2DL3 mutants targeting polymorphic residues

    • Create both naturally occurring variants and designed mutations to test structural hypotheses

    • Express these constructs in appropriate expression systems (bacterial for structural studies, mammalian for functional assays)

  • Binding assays:

    • Employ Surface Plasmon Resonance (SPR) to quantitatively measure binding kinetics and affinity

    • Test binding to multiple HLA-C allotypes to assess how polymorphisms affect ligand specificity

    • Examine binding across different peptide-HLA complexes to evaluate peptide selectivity

  • Functional validation:

    • Transfect KIR2DL3 variants into NK cell lines or primary NK cells

    • Assess inhibitory function using degranulation or cytotoxicity assays

    • Compare results with binding data to establish structure-function relationships

  • Comparative analysis with KIR2DL2:

    • Include KIR2DL2 variants in analyses as relevant comparisons

    • Identify polymorphisms that drive functional differences between KIR2DL2 and KIR2DL3

    • Create chimeric receptors to pinpoint domains responsible for binding differences

This comprehensive approach enables researchers to understand how specific polymorphisms in KIR2DL3 influence HLA-C recognition and subsequent NK cell function, providing insights into the molecular basis of disease associations.

What crystallization conditions and techniques are most successful for structural studies of KIR2DL3-HLA-C complexes?

For successful structural studies of KIR2DL3-HLA-C complexes, the following crystallization conditions and techniques have proven effective:

  • Protein preparation:

    • Express and purify KIR2DL3 D1-D2 domains (residues 1-204) using the bacterial expression system

    • Produce HLA-C heavy chain and β2-microglobulin separately, then refold with synthetic peptide

    • Ensure high purity (>95% by SDS-PAGE) and monodispersity (by size exclusion chromatography)

    • Form the complex by mixing purified KIR2DL3 and peptide-loaded HLA-C at equimolar ratios

  • Crystallization screening:

    • Initial screening at protein concentrations of 5-10 mg/ml

    • Use commercial sparse matrix screens as starting points

    • Focus on conditions containing PEG 3350 or PEG 6000 as precipitants

    • Ideal temperature range: 4-20°C

    • Successful conditions typically include:

      • 16% PEG 3350, 0.2 M potassium sodium tartrate, 0.1 M Bis-Tris propane pH 7.5

  • Optimization strategies:

    • Fine-tune precipitant concentration, pH, and additive compositions

    • Employ microseeding techniques to improve crystal quality

    • Use additives like glycerol or low concentrations of detergents to reduce non-specific aggregation

    • Implement streak seeding from initial microcrystals

  • Data collection considerations:

    • Harvest crystals using appropriate cryoprotectant (e.g., mother liquor supplemented with 20% glycerol)

    • Collect high-resolution data (aim for resolution better than 2.5 Å)

    • Use synchrotron radiation sources for optimal data quality

    • Process data using appropriate software packages (e.g., XDS, MOSFLM)

  • Structure solution and refinement:

    • Solve structures by molecular replacement using existing KIR-HLA structures as search models

    • Apply non-crystallographic symmetry restraints if multiple copies exist in the asymmetric unit

    • Perform iterative cycles of manual building and refinement

    • Validate final structures using tools like MOLPROBITY

These approaches have successfully yielded high-quality crystals diffracting to 2.5 Å resolution or better, allowing detailed structural analysis of KIR2DL3-HLA-C interactions.

How should researchers interpret contradictory findings regarding KIR2DL3 associations with disease outcomes?

When faced with contradictory findings regarding KIR2DL3 associations with disease outcomes, researchers should consider several factors and implement the following analytical approaches:

  • Evaluate study design differences:

    • Compare sample sizes and statistical power across studies

    • Assess population demographics and genetic backgrounds

    • Examine disease classification criteria and phenotype definitions

    • Consider environmental factors that might differ between populations

  • Analyze KIR-HLA combinations holistically:

    • KIR2DL3 functions within a complex genetic network; evaluate it in context with:

      • Presence/absence of HLA-C1 alleles (the ligand for KIR2DL3)

      • Co-inherited KIR genes, especially those in linkage disequilibrium

      • HLA-C allotype variations that affect binding strength

    • Consider compound genotypes rather than individual receptor presence

  • Account for functional complexity:

    • Not all KIR2DL3-HLA-C1 interactions are functionally equivalent

    • Different HLA-C1 allotypes show variable binding to KIR2DL3

    • Peptide repertoires presented by HLA-C can significantly impact binding

    • Consider the functional implications of specific polymorphisms

  • Implement advanced statistical approaches:

    • Use multivariate analyses to account for confounding factors

    • Consider interaction terms between KIR and HLA genes

    • Apply haplotype analysis rather than single-gene association

    • Perform meta-analyses when multiple studies are available

  • Validate with functional studies:

    • Complement genetic association studies with functional experiments

    • Test NK cell responses from individuals with relevant genotypes

    • Investigate the specific mechanisms proposed to explain associations

    • Use appropriate cellular models that recapitulate disease conditions

By applying these approaches, researchers can better understand the true nature of KIR2DL3 associations with disease outcomes and reconcile apparently contradictory findings.

What statistical methods are most appropriate for analyzing KIR2DL3 binding data across multiple HLA-C allotypes?

When analyzing KIR2DL3 binding data across multiple HLA-C allotypes, the following statistical methods are most appropriate:

  • Descriptive statistics and visualization:

    • Calculate mean, median, standard deviation, and coefficient of variation for binding parameters

    • Create heat maps displaying relative binding across HLA-C allotypes

    • Use radar plots to visualize binding profiles across multiple parameters

    • Implement hierarchical clustering to identify patterns in binding preferences

  • Normalization approaches:

    • Express binding values as percentage of maximum binding observed

    • Use internal standards (e.g., a reference HLA-C allotype) for normalization

    • Apply Z-score normalization when comparing across different experimental batches

    • Consider log transformation for binding data that spans multiple orders of magnitude

  • Comparative statistical tests:

    • Use paired t-tests when comparing KIR2DL2 vs. KIR2DL3 binding to the same HLA-C allotype

    • Apply ANOVA with post-hoc tests for comparing binding across multiple allotypes

    • Implement non-parametric alternatives (Wilcoxon, Kruskal-Wallis) when data is not normally distributed

    • Use repeated measures designs when appropriate

  • Correlation and regression analyses:

    • Calculate correlation coefficients between binding parameters and functional outcomes

    • Implement multiple regression to identify determinants of binding strength

    • Use principal component analysis to reduce dimensionality in complex datasets

    • Consider mixed-effects models when analyzing data with nested structure

  • Specialized analyses for binding kinetics:

    • For SPR data, use appropriate models (1:1 binding, heterogeneous ligand, etc.)

    • Calculate and compare kinetic parameters (ka, kd) and equilibrium constants (KD)

    • Apply statistical tests specifically designed for comparing binding curves

    • Consider global fitting approaches when appropriate

These statistical methods, when properly applied, allow for robust analysis of KIR2DL3 binding data across multiple HLA-C allotypes, enabling meaningful comparisons and identification of biologically significant patterns.

How can researchers effectively integrate structural, binding, and functional data to build comprehensive models of KIR2DL3 biology?

To effectively integrate structural, binding, and functional data for comprehensive modeling of KIR2DL3 biology, researchers should implement the following systematic approach:

  • Data harmonization and integration framework:

    • Create standardized formats for different data types

    • Develop a common ontology for describing molecular interactions

    • Implement database solutions for organizing diverse experimental results

    • Establish clear provenance tracking for all data sources

  • Structure-function correlation analysis:

    • Map binding affinity data onto structural interfaces

    • Identify structural features that correlate with functional outcomes

    • Analyze how specific amino acid contacts influence binding kinetics

    • Examine how conformational changes relate to signaling events

  • Predictive computational modeling:

    • Develop molecular dynamics simulations informed by crystal structures

    • Implement machine learning approaches to predict binding from sequence data

    • Create systems biology models integrating receptor-ligand interactions with downstream signaling

    • Validate computational predictions with experimental data

  • Integrative visualization approaches:

    • Generate interactive visualizations linking structure to function

    • Create molecular movies illustrating binding mechanisms

    • Develop network diagrams showing relationships between different variables

    • Implement dashboards that allow exploration of integrated datasets

  • Validation through hypothesis testing:

    • Design experiments that specifically test predictions from integrated models

    • Create mutants that probe key structural features identified in the model

    • Measure multiple parameters simultaneously to capture system behavior

    • Iterate between model refinement and experimental validation

  • Consider biological context:

    • Integrate polymorphism data from population studies

    • Incorporate disease association information

    • Account for the impact of peptide repertoire on binding

    • Consider the broader context of NK cell receptor expression patterns

This integrative approach allows researchers to develop comprehensive models of KIR2DL3 biology that capture the complex relationships between receptor structure, binding properties, and functional outcomes, ultimately providing deeper insights into how these molecules contribute to human health and disease.

What novel methodologies are emerging for studying KIR2DL3-mediated NK cell regulation?

Several cutting-edge methodologies are revolutionizing the study of KIR2DL3-mediated NK cell regulation:

  • Single-cell technologies:

    • Single-cell RNA sequencing to profile gene expression in KIR2DL3+ NK cells

    • Mass cytometry (CyTOF) for high-dimensional phenotypic and functional profiling

    • Single-cell proteomics to measure signaling pathway activation

    • Integration of these datasets to create comprehensive cellular states

  • Advanced imaging approaches:

    • Super-resolution microscopy to visualize KIR2DL3 organization at immune synapses

    • Live-cell imaging to track receptor dynamics during NK-target interactions

    • Fluorescence resonance energy transfer (FRET) to measure receptor-ligand interactions

    • Lattice light-sheet microscopy for long-term 3D imaging of cellular interactions

  • Genome editing technologies:

    • CRISPR-Cas9 to create precise KIR2DL3 variants in primary NK cells

    • Base editing for introducing specific polymorphisms

    • Inducible gene expression systems to control receptor levels

    • Genetic screens to identify novel regulators of KIR2DL3 function

  • Proteomics and interactomics:

    • Proximity labeling to identify proteins associated with KIR2DL3

    • Phosphoproteomics to map signaling pathways downstream of KIR2DL3

    • Interaction proteomics to characterize KIR2DL3 binding partners

    • Temporal profiling of signaling events following receptor engagement

  • Organoid and physiological models:

    • NK-target interactions in 3D organoid cultures

    • Humanized mouse models expressing KIR2DL3 and HLA-C variants

    • Microfluidic systems to study NK cell migration and killing dynamics

    • Patient-derived models to study disease-relevant contexts

These emerging methodologies promise to provide unprecedented insights into how KIR2DL3 regulates NK cell function in health and disease, potentially leading to new therapeutic strategies targeting this receptor system.

What are the key unresolved questions regarding the role of KIR2DL3 in human immunity?

Despite significant advances in understanding KIR2DL3 biology, several critical questions remain unresolved:

  • Mechanistic basis of disease associations:

    • Why is KIR2DL3, but not KIR2DL2, associated with HCV clearance despite their high sequence similarity?

    • What specific aspects of KIR2DL3-HLA-C1 interactions contribute to ulcerative colitis progression?

    • How do genetic polymorphisms in both receptor and ligand translate to functional differences in disease contexts?

  • Education and licensing processes:

    • How does KIR2DL3 interaction with HLA-C1 during NK cell development shape functional responsiveness?

    • Do different HLA-C1 allotypes vary in their ability to educate KIR2DL3+ NK cells?

    • How stable is the educated state, and can it be reprogrammed during immune responses?

  • Peptide repertoire effects:

    • How does the cellular peptide repertoire change during infection or transformation?

    • Do these changes significantly alter KIR2DL3 recognition of HLA-C1?

    • Can specific pathogen-derived peptides modulate KIR2DL3 function?

  • Receptor cooperation and competition:

    • How does KIR2DL3 function integrate with signals from other inhibitory and activating receptors?

    • What is the significance of KIR2DL3 co-expression with other KIR family members?

    • How do KIR2DL3 and KIR2DS2 interact functionally when co-expressed?

  • Therapeutic targeting potential:

    • Can modulation of KIR2DL3-HLA-C1 interactions enhance anti-viral or anti-tumor immunity?

    • Would blocking KIR2DL3 have different effects than targeting pan-KIR molecules?

    • How can KIR2DL3 polymorphisms inform personalized immunotherapeutic approaches?

  • Evolutionary perspectives:

    • Why has polymorphism been maintained in both KIR2DL2/L3 and their HLA-C ligands?

    • What selective pressures have shaped the distribution of these alleles in human populations?

    • How do KIR2DL2/L3 differences contribute to population-level disease resistance?

Addressing these unresolved questions will require integrated approaches combining structural biology, genetics, cellular immunology, and systems biology.

How might KIR2DL3-targeted approaches be developed for immunotherapy applications?

Developing KIR2DL3-targeted approaches for immunotherapy presents both challenges and opportunities. Here's a framework for potential therapeutic development:

  • Blocking antibody approaches:

    • Develop highly specific monoclonal antibodies targeting KIR2DL3

    • Engineer antibodies with enhanced specificity to distinguish KIR2DL3 from KIR2DL2

    • Create F(ab')2 fragments to prevent Fc receptor engagement

    • Combine with existing checkpoint inhibitors for potential synergistic effects

  • Peptide-based modulators:

    • Design peptides that selectively bind HLA-C1 and either enhance or disrupt KIR2DL3 interaction

    • Leverage knowledge of peptide positions (P7, P8) critical for KIR2DL3 recognition

    • Create stabilized peptide analogs with improved pharmacokinetic properties

    • Develop peptide delivery systems targeting specific tissue microenvironments

  • Small molecule inhibitors:

    • Identify small molecule compounds that disrupt KIR2DL3-HLA-C1 interaction

    • Target specific structural features of KIR2DL3 not shared with KIR2DL2

    • Use structure-based drug design informed by crystal structures

    • Optimize compounds for specificity, potency, and drug-like properties

  • Cell therapy enhancements:

    • Engineer NK cells to express modified KIR2DL3 receptors with altered signaling properties

    • Silence or knockout KIR2DL3 in NK cells for adoptive transfer

    • Use CRISPR-based approaches for precise genetic modifications

    • Create chimeric receptors incorporating KIR2DL3 components with altered specificity

  • Biomarker development and patient stratification:

    • Develop assays to determine KIR and HLA genotypes for patient selection

    • Create functional tests to assess KIR2DL3-mediated inhibition in patient NK cells

    • Identify biomarkers that predict response to KIR2DL3-targeted therapies

    • Implement companion diagnostics for clinical trials

  • Therapeutic contexts:

    • Viral infections: Enhance viral clearance by releasing KIR2DL3-mediated inhibition

    • Cancer immunotherapy: Combine with tumor-targeting strategies to enhance NK killing

    • Autoimmune conditions: Potentially modulate KIR2DL3 function to reduce pathological inflammation

    • Transplantation: Engineer donor NK cells with modified KIR2DL3 function

These approaches must carefully consider the complex biology of KIR2DL3, including its polymorphism, specificity for HLA-C allotypes, and interplay with other receptors, to develop effective and safe immunotherapeutic strategies.

Product Science Overview

Structure and Function

KIR2DL3 is characterized by:

  • Two extracellular C2-type immunoglobulin-like domains: These domains are responsible for recognizing specific molecules on the surface of target cells.
  • A transmembrane domain: This region anchors the receptor to the cell membrane.
  • A long cytoplasmic tail with two immunoreceptor tyrosine-based inhibitory motifs (ITIMs): These motifs are crucial for transmitting inhibitory signals to the NK cell, preventing it from attacking normal, healthy cells .
Genetic and Molecular Background

The KIR2DL3 gene is located on chromosome 19q13.4 . It is part of a gene family that includes several other KIR genes, each with varying specificities and functions. The expression of KIR2DL3 and its interaction with human leukocyte antigen (HLA) molecules are essential for the regulation of NK cell activity. Specifically, KIR2DL3 interacts with HLA-C1 alleles, which helps in distinguishing between healthy cells and potential threats .

Clinical Significance

KIR2DL3 has been associated with various clinical conditions, including:

  • Graft-Versus-Host Disease (GVHD): The presence of certain KIR and HLA combinations can influence the outcome of hematopoietic stem cell transplantation .
  • Prostate Cancer: Variations in KIR2DL3 expression have been linked to the progression of prostate cancer .
  • HIV Resistance: Studies have shown that certain KIR2DL3 genotypes may confer resistance to HIV infection in exposed individuals .
Research and Applications

The recombinant form of KIR2DL3 is used in various research applications to study its role in immune regulation and its potential therapeutic implications. Understanding the interactions between KIR2DL3 and HLA molecules can lead to the development of new treatments for immune-related diseases and conditions.

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.