KIR2DS5 antibodies are immunological tools designed to detect and study the KIR2DS5 receptor, an activating member of the killer cell immunoglobulin-like receptor (KIR) family expressed on natural killer (NK) cells and certain T lymphocytes . These antibodies enable researchers to investigate KIR2DS5's role in immune responses, including its interactions with ligands and implications in diseases such as viral infections, autoimmune disorders, and cancer .
Recent advances have produced monoclonal antibodies (mAbs) with high specificity for KIR2DS5:
These antibodies are critical for distinguishing KIR2DS5 from structurally similar KIRs (e.g., KIR2DL1) and for quantifying receptor expression levels in clinical samples .
Functional Studies: Antibodies like 5E11A6 stimulate KIR2DS5-positive NK cells to secrete IFN-γ and chemokines (MIP-1α/β), confirming the receptor’s activating potential .
Disease Associations:
COVID-19: KIR2DS5 antibodies identified elevated KIR2DS5+ NK cells in severe COVID-19 patients, correlating with hyperinflammatory responses .
Alcoholic Cirrhosis: KIR2DS5+ NK cells are linked to accelerated liver fibrosis in non-viral cases, suggesting a pathogenic role .
Preeclampsia: Allele-specific antibodies revealed KIR2DS5*006’s protective effect in Ugandan cohorts by countering inhibitory KIR2DL1 interactions .
Allele | Surface Expression | Glycosylation Pattern | Functional Outcome |
---|---|---|---|
*002 | Low | Immature N-linked glycans | No HLA-C binding |
*005 | High | Mature glycans | Binds HLA-C2 |
*006 | Moderate | Mature glycans | Protective in preeclampsia |
Autoimmunity: KIR2DS5 antibodies help identify patients with KIR2DS5/HLA-C mismatches, a risk factor in rheumatoid arthritis and ankylosing spondylitis .
Transplantation: Monitoring KIR2DS5+ NK cells may predict graft rejection, as activating KIRs promote alloreactivity .
Cancer Immunotherapy: Antibodies blocking KIR2DS5 could mitigate NK cell exhaustion in tumors, while agonists might enhance antiviral responses .
KIR2DS5 is an activating killer immunoglobulin-like receptor expressed on natural killer (NK) cells. It belongs to the KIR family of receptors that regulate NK cell cytotoxicity through recognition of altered MHC-I expression on infected or transformed cells. KIR2DS5 is particularly significant because it has been associated with improved clinical outcomes in various diseases, including COVID-19, where KIR2DS5-positive patients demonstrated significantly shorter recovery times compared to KIR2DS5-negative patients (mean 21.6 ± 2.8 days vs. 44.6 ± 2.2 days) . The receptor triggers both NK cell cytotoxicity and interferon γ (IFN-γ) release, potentially explaining its protective effects in viral infections . This makes it a valuable target for immunological research focused on host defense mechanisms and potential therapeutic approaches.
KIR2DS5 is polymorphic, with several identified alleles that differ in their expression levels and glycosylation patterns. The most frequently observed allele is KIR2DS5002, but researchers have identified multiple variants (KIR2DS5002 through KIR2DS5009) . These allelic products show consistent variation in expression levels when transfected into cell lines, with KIR2DS5003 exhibiting the highest level of expression, while KIR2DS5002 and KIR2DS5009 show the lowest . The intermediate expression levels are seen with KIR2DS5004 through KIR2DS5008 . These differences in expression are consistent across different cell lines and detection methods, indicating intrinsic properties of the polypeptide sequences rather than cell type-specific effects.
Research has demonstrated that KIR2DS5 positivity correlates with improved clinical outcomes in several conditions. In COVID-19 specifically:
KIR2DS5-positive patients with severe ARDS showed significantly shorter time to recovery compared to KIR2DS5-negative patients (21.6 ± 2.8 days vs. 44.6 ± 2.2 days)
By day 28 after admission, 60% of KIR2DS5-positive patients had recovered compared to only 9% of KIR2DS5-negative patients
In patients with moderate COVID-19, KIR2DS5 positivity was associated with freedom from transfer to ICU (0% vs 22%, p-value = 0.04)
KIR2DS5-positive patients required less oxygen supplementation than KIR2DS5-negative patients (22% vs. 46%), though this did not reach statistical significance (p-value = 0.08)
Beyond COVID-19, KIR2DS5 has shown protective effects in acute rejection of kidney grafts, malaria, HIV infection, and hepatitis C-induced hepatocellular carcinoma . The enhanced activation of NK cells through KIR2DS5 may explain these observations, as NK cell exhaustion has been associated with poorer outcomes in various infectious diseases.
Several antibodies have been developed for KIR2DS5 detection in research applications:
F6075: A rabbit polyclonal antisera raised against the peptide HEGFRRKPSLLA, corresponding to amino acids 1-12 of KIR2DS5
5E11A6: A mouse monoclonal antibody produced by immunizing BALB/c mice with a KIR2DS5*002-Fc fusion protein isolated from a baculovirus system
Both antibodies have been tested for specificity using flow cytometry with a panel of NKL cell lines expressing single KIR allotypes (KIR2DS1, 2DS2, 2DS4, 2DS5, 2DL1, 2DL2, 2DL3, 2DL5A, and 3DL1) . This comprehensive validation ensures minimal cross-reactivity with other KIR family members, making these antibodies suitable for selective detection of KIR2DS5 in research applications.
To validate KIR2DS5 antibody specificity in experimental systems, researchers should:
Use cell line panels: Test antibodies against a panel of cell lines expressing different KIR family members, similar to the validation approach used for F6075 and 5E11A6
Include appropriate controls:
Perform cross-validation with multiple detection methods:
Flow cytometry for surface expression
Western blotting for protein size and expression levels
Immunoprecipitation followed by mass spectrometry for definitive identification
Use isotype-matched controls for monoclonal antibodies to rule out non-specific binding (e.g., mouse IgG1 kappa for 5E11A6)
This systematic validation approach ensures that observed signals are specific to KIR2DS5 and not due to cross-reactivity with structurally similar KIR family members.
For detecting KIR2DS5 expression in primary human NK cells, researchers should consider a multi-method approach:
Flow Cytometry:
RT-PCR for detecting KIR2DS5 mRNA:
Design primers specific to KIR2DS5, avoiding regions with high homology to other KIR family members
Consider including sequence verification of amplicons due to high sequence similarity between KIR genes
Single-cell RNA sequencing:
Provides comprehensive expression profiles and can identify KIR2DS5+ subpopulations
Enables correlation of KIR2DS5 expression with other NK cell receptors and functional states
For all methods, comparison with KIR genotyped reference samples is recommended to establish reliable detection thresholds and account for allelic variation in expression levels.
Distinguishing between KIR2DS5 alleles requires specialized approaches due to their high sequence similarity:
DNA-based methods:
Sequence-specific primer PCR (SSP-PCR) with primers designed to target polymorphic regions
Next-generation sequencing of the KIR locus for comprehensive allele determination
Restriction fragment length polymorphism (RFLP) analysis based on differential cutting sites
Protein-based methods:
Expression pattern analysis:
For the most accurate results, researchers should combine DNA-based genotyping with protein expression analysis to confirm the presence and functionality of specific alleles.
To study the functional consequences of KIR2DS5 activation, consider these methodological approaches:
NK cell cytotoxicity assays:
Cytokine production assessment:
Signaling pathway analysis:
Western blotting for phosphorylation of downstream signaling molecules
Live cell imaging with fluorescent reporters to visualize calcium flux or other rapid signaling events
RNA-seq to characterize transcriptional responses to KIR2DS5 activation
Stable cell line models:
These approaches can systematically characterize the signaling and functional consequences of KIR2DS5 engagement in various experimental contexts.
Detection of low-expression KIR2DS5 alleles like KIR2DS5002 and KIR2DS5009 presents technical challenges that can be addressed through several strategies:
Signal amplification techniques:
Use biotin-streptavidin systems to amplify antibody signals
Employ tyramide signal amplification for immunohistochemistry and flow cytometry
Consider fluorescent nanoparticle-conjugated antibodies for enhanced sensitivity
Optimized staining protocols:
Increase antibody concentration (titrate to determine optimal concentration)
Extend incubation times to allow for complete binding
Reduce background with careful blocking and washing steps
Enrichment approaches:
Magnetic or flow-based cell sorting to enrich for KIR2DS5+ populations
Use RNA-binding protein immunoprecipitation to concentrate KIR2DS5 mRNA for detection
Culture cells with cytokines (IL-2, IL-15) that may upregulate KIR expression
Alternative detection methods:
Digital PCR for sensitive detection of KIR2DS5 transcripts
Proximity ligation assay for protein detection with single-molecule sensitivity
Mass cytometry (CyTOF) for high-dimensional analysis with enhanced signal-to-noise ratio
Combining these approaches can significantly improve detection of low-abundance KIR2DS5 allelic products in various experimental systems.
KIR2DS5 shows variable glycosylation patterns that affect protein detection and potentially function. Critical factors and analysis methods include:
Key determinants of glycosylation:
Analysis methods:
Methodological considerations:
Functional implications:
Monitor the ratio between mature (fully glycosylated) and immature forms across different alleles
Correlate glycosylation patterns with surface expression levels and functional activity
Use site-directed mutagenesis to modify potential glycosylation sites and assess functional consequences
Understanding these glycosylation patterns is crucial for accurate detection and functional characterization of KIR2DS5 variants.
Computational approaches offer powerful tools for predicting KIR2DS5 interactions and functions:
Homology modeling:
Molecular docking:
Predict potential binding partners by docking KIR2DS5 models with MHC-I molecules and other candidates
Calculate binding energies to rank likely interaction partners
Simulate the effects of allelic variations on binding properties
Sequence-based predictions:
Machine learning algorithms trained on known KIR-ligand pairs
Conservation analysis across species to identify functionally important residues
Coevolution analysis to detect correlated mutations suggesting interaction interfaces
Systems biology approaches:
Network analysis integrating expression data, protein interactions, and clinical correlations
Prediction of signaling pathways activated by different KIR2DS5 alleles
Population-level modeling of KIR2DS5 distribution and disease associations
These computational approaches can generate testable hypotheses about KIR2DS5 function and guide experimental designs for binding partner identification.
KIR2DS5 polymorphisms have significant implications for personalized medicine:
Disease risk stratification:
Treatment response prediction:
KIR2DS5 status could potentially predict response to immunotherapies that depend on NK cell function
Patient stratification based on KIR genotype might optimize treatment selection
Monitoring KIR2DS5+ NK cell populations during treatment could serve as a biomarker for response
Therapeutic targeting opportunities:
Development of agonistic antibodies to stimulate KIR2DS5 signaling in KIR2DS5-positive individuals
Allele-specific approaches might be needed due to expression level differences
Combined therapies targeting multiple NK receptors based on individual KIR profiles
Clinical implementation considerations:
KIR genotyping assays would need standardization for clinical use
Interpretation guidelines accounting for ethnic variations in KIR distribution
Integration with other genetic and clinical markers for improved predictive power
This emerging field connects basic KIR2DS5 biology with potential clinical applications, offering new pathways for personalized immunotherapy approaches.
KIR2DS5 functions within a complex network of activating and inhibitory receptors on NK cells:
Receptor co-expression patterns:
KIR2DS5 expression varies among NK cell subsets and may correlate with specific functional states
Co-expression with inhibitory KIRs may create balanced regulation of NK cell activation
NK cell education may be influenced by the specific combination of KIRs expressed
Signal integration mechanisms:
Methodological approaches to study receptor interactions:
Multi-parameter flow cytometry to correlate receptor expression with functional readouts
CRISPR-based genetic modification to create NK cells with defined receptor combinations
Single-cell analysis to identify receptor expression patterns associated with enhanced function
Temporal dynamics:
Sequential engagement of different receptors may influence the activation pathway
Receptor clustering and synapse formation patterns include KIR2DS5 in conjunction with other molecules
Receptor expression changes during NK cell activation may alter the contribution of KIR2DS5 over time
Understanding these complex interactions is essential for accurately predicting how KIR2DS5 variants influence immune responses in different contexts.
Standardizing KIR2DS5 expression data across different platforms requires careful normalization approaches:
Flow cytometry data normalization:
Western blot quantification:
RT-PCR and transcriptomic data:
Select appropriate reference genes with stable expression across conditions
Use absolute quantification with standard curves when possible
Apply batch correction methods for large-scale datasets
Cross-platform integration:
Develop conversion factors between different measurement methods
Use rank-based metrics rather than absolute values when combining diverse data types
Apply statistical methods that account for platform-specific technical variability
This standardized approach enables reliable comparison of KIR2DS5 expression data from different studies and experimental systems.
When analyzing KIR2DS5 genotype-outcome relationships, these statistical approaches are recommended:
Primary analysis methods:
Multivariate approaches:
Cox proportional hazards models to adjust for confounding factors
Logistic regression for binary outcomes with adjustment for demographics and comorbidities
Propensity score matching to balance KIR2DS5+ and KIR2DS5- groups
Sample size considerations:
Validation strategies:
These statistical approaches help establish robust associations between KIR2DS5 genotype and clinical outcomes while accounting for potential confounding factors.