KLRC1 Human Recombinant is produced in Sf9 Baculovirus cells as a glycosylated polypeptide chain containing 149 amino acids (residues 94–233) with a molecular mass of 17.1 kDa. The protein includes a C-terminal 9-amino-acid histidine tag and is stabilized in phosphate-buffered saline (pH 7.4) with 10% glycerol .
Parameter | Details |
---|---|
Molecular Weight | 17.1 kDa |
Purity | >90% (SDS-PAGE) |
Expression System | Sf9 Baculovirus cells |
Post-Translational Modifications | Glycosylated, Type II membrane orientation |
KLRC1 forms heterodimers with KLRD1 (CD94), creating the CD94/NKG2A receptor complex critical for HLA-E recognition . Alternative splicing generates isoforms, including NKG2-A and NKG2-B, which differ in cytoplasmic domains and signaling outcomes .
KLRC1 regulates immune responses through:
Inhibition of Cytotoxicity: Binds HLA-E loaded with self-peptides, preventing NK/T-cell-mediated destruction of healthy cells .
Tumor Immune Evasion: In HLA-E-rich tumor microenvironments, KLRC1 suppresses antitumor activity, contributing to T/NK cell exhaustion .
Viral Immune Evasion: Recognizes HLA-E complexed with viral peptides (e.g., human cytomegalovirus UL40), enabling viral persistence .
CD16 Signaling in NK Cells: Modulates cytotoxicity and cytokine production .
TGF-β1 Secretion: Triggers regulatory T-cell responses in mucosal tissues .
KLRC1 expression is dysregulated across malignancies:
In LUAD and lung squamous cell carcinoma (LUSC), KLRC1 expression negatively correlates with tumor purity but positively associates with CD8+ T-cell infiltration, suggesting a dual role in immune regulation .
KLRC1+ NK cells are expanded in cytomegalovirus (CMV)-positive individuals, exhibiting adaptive features (e.g., CD57+, KIR+) and reduced FcεR1γ expression .
CRISPR Knockout: KLRC1 knockout enhances NK cell cytotoxicity against HLA-E+ tumors, overcoming immune inhibition .
Clinical Trials: Anti-NKG2A antibodies (e.g., monalizumab) combined with PD-1/PD-L1 inhibitors show promise in restoring antitumor immunity .
Lung Cancer: High KLRC1 correlates with prolonged survival in LUSC but not LUAD .
Colorectal Cancer: Reduced KLRC1 associates with advanced disease stages .
KLRC1 interacts with:
Partner | Function | Interaction Score |
---|---|---|
KLRD1 | Forms CD94/NKG2A inhibitory receptor | 0.999 |
HLA-E | Presents self/viral peptides for immune recognition | 0.988 |
TGFB1 | Induces regulatory T-cell differentiation | Experimental |
NKG2-A/NKG2-B type II integral membrane protein isoform NKG2-A, KLRC1, Killer Cell Lectin-Like Receptor Subfamily C, Member 1, KLRC1, CD159A, NKG2, NKG2A, CD159 antigen-like family member A, NK cell receptor A, NKG2-A/B-activating NK receptor.
Sf9, Baculovirus cells.
ADPPSTLIQR HNNSSLNTRT QKARHCGHCP EEWITYSNSC YYIGKERRTW EESLLACTSK NSSLLSIDNE EEMKFLSIIS PSSWIGVFRN SSHHPWVTMN GLAFKHEIKD SDNAELNCAV LQVNRLKSAQ CGSSIIYHCK HKLHHHHHH
KLRC1 (killer cell lectin-like receptor subfamily C, member 1), also known as NKG2A or CD159A, is a type II transmembrane protein primarily expressed in Natural Killer (NK) cells. It belongs to the killer cell lectin-like receptor family characterized by a C-type lectin domain. KLRC1 forms a heterodimeric complex with KLRD1/CD94 and functions as an inhibitory receptor that recognizes MHC class I HLA-E molecules on target cells. This interaction is critical for NK cell-mediated immune surveillance, as it prevents NK cells from killing healthy cells expressing normal levels of MHC class I molecules .
Human KLRC1 is a transmembrane glycoprotein with a C-type lectin domain. It has a type II membrane orientation (N-terminus on the cytoplasmic side) and contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) in its cytoplasmic domain. The gene encoding KLRC1 is located on chromosome 12p13 within the NK gene complex, adjacent to other KLR family genes. The protein forms a complex with KLRD1/CD94 and recognizes MHC class I HLA-E molecules. Four alternatively spliced transcript variants encoding two distinct isoforms have been identified .
KLRC1 functions through inhibitory signaling pathways. When the KLRC1/CD94 heterodimer engages with HLA-E molecules on target cells, the ITIMs in KLRC1's cytoplasmic domain become phosphorylated. This phosphorylation creates binding sites for tyrosine phosphatases SHP-1 and SHP-2, which then dephosphorylate and inactivate components of NK cell activation pathways. Similar to other inhibitory NK cell receptors like KLRL1, this mechanism prevents NK-mediated cytotoxicity against cells expressing appropriate MHC class I molecules, ensuring that healthy self cells are protected from NK cell attack .
For comprehensive analysis of KLRC1 expression at the single-cell level, several complementary approaches are recommended:
Single-cell RNA sequencing (scRNA-seq): This technique has successfully delineated NK cell subsets based on KLRC1 expression. When analyzing scRNA-seq data, set appropriate detection thresholds for KLRC1 (e.g., log2 normalized counts above 0) .
Flow cytometry: Using anti-KLRC1 (anti-NKG2A) antibodies combined with other NK cell markers like CD56, CD16, and CD57 allows for detailed subset characterization.
RT-PCR with KLRC1-specific primers: For analysis of KLRC1 expression in sorted cell populations or tissues, primers such as 5′-ACGAATTCATGTCTGAAGAAGTTACTTA-3′ (sense) and 5′-TCAAGCTTGCCTCCCTAAAATATGTAG-3′ (antisense) have been validated .
Fluorescence confocal microscopy: For visualizing KLRC1 expression and subcellular localization, GFP-fused KLRC1 expression vectors can be particularly useful .
During NK cell maturation, there is a clear developmental relationship between KLRC1 and inhibitory KIR expression:
NK Cell Subset | KLRC1 (NKG2A) Expression | KIR Expression | Developmental Stage |
---|---|---|---|
CD56bright | High | Very low (<5%) | Immature/cytokine producer |
CD56dim GZMK+ | Intermediate | Low (16%) | Intermediate maturation |
CD56dim GZMK- (general) | Lower | Higher | More mature |
CD57+-like subset | Low | High (35%) | Terminal differentiation |
KLRC2+ (NKG2C+) subset | Very low | Very high (56%) | Terminal differentiation |
This inverse relationship, where KLRC1 expression decreases as KIR expression increases during NK cell maturation, suggests a developmental switch from KLRC1-mediated to KIR-mediated inhibition as NK cells mature .
To effectively study KLRC1 function in NK cells, researchers should consider these methodological approaches:
Functional assays:
Receptor modulation:
Antibody blocking of KLRC1-HLA-E interactions
siRNA/shRNA knockdown of KLRC1 expression
CRISPR-Cas9 gene editing for KLRC1 knockout
Signaling studies:
Immunoprecipitation to detect KLRC1 association with SHP-1/SHP-2 phosphatases
Phosphorylation analysis of ITIM motifs
Western blotting to assess downstream signaling events
Expression systems:
Distinguishing KLRC1 from other KLR family members requires careful attention to specificity:
Genetic analysis:
Use PCR primers targeting unique regions of KLRC1
Sequence verification to confirm identity
Protein analysis:
Western blotting with antibodies specifically validated against KLRC1
Mass spectrometry for precise identification
Co-immunoprecipitation with CD94, as KLRC1 forms heterodimers with CD94
Functional differentiation:
KLRC1 uniquely recognizes HLA-E, while other KLR members have different ligand specificities
KLRC1 contains ITIMs and associates with SHP-1/SHP-2, while activating KLRs like KLRC2 (NKG2C) associate with activating adaptor proteins
KLRC1 inhibits NK cell cytotoxicity, in contrast to activating members of the family
For comprehensive analysis of KLRC1 distribution:
Tissue expression profiling:
Immune cell subtype analysis:
Tissue localization:
Developmental analysis:
Ontogeny studies examining KLRC1 expression during immune cell development
Analysis of KLRC1 expression during immune responses
The complementary roles of KLRC1 and KIRs create a nuanced NK cell education system:
Differential ligand recognition:
KLRC1/CD94 recognizes the non-classical MHC-I molecule HLA-E
KIRs recognize classical MHC-I molecules (HLA-A, -B, -C)
This dual recognition system ensures comprehensive monitoring of MHC-I expression patterns
Developmental regulation:
Calibration of NK cell responsiveness:
The sequential acquisition of these receptors during development likely calibrates NK cell activation thresholds
The inhibitory strengths of KLRC1 and various KIR combinations may differ, creating varied response sensitivities
This diversity enables nuanced responses to different threats
Functional specialization:
KLRC1-dominant NK cells may be specialized for certain functions (e.g., cytokine production)
KIR-dominant NK cells may be optimized for cytotoxicity against transformed cells
KLRC1 heterogeneity significantly contributes to NK cell functional diversity:
Single-cell transcriptomic analysis reveals that KLRC1 expression helps define distinct NK cell subsets with potentially specialized functions. The CD56dim GZMK- NK cell population can be further divided into multiple clusters with varying KLRC1 expression patterns .
KLRC1 expression correlates with specific functional profiles:
The inverse relationship between KLRC1 and KIR expression creates diverse inhibitory receptor combinations:
NK cells with high KLRC1/low KIR likely have different activation thresholds than cells with low KLRC1/high KIR
This diversity creates a spectrum of NK cell responsiveness rather than a binary educated/uneducated state
KLRC1 heterogeneity may contribute to tissue-specific NK cell adaptations, with different expression patterns between blood and tissue-resident NK cells.
While classical immunological memory is primarily associated with adaptive immunity, emerging evidence suggests KLRC1 may play a role in NK cell memory-like properties:
Modulation during viral challenges:
KLRC1 expression can be downregulated during certain viral infections
This downregulation may contribute to enhanced NK cell responsiveness to subsequent challenges
Relationship with adaptive features:
The dynamic regulation of KLRC1 versus KLRC2 (NKG2C, an activating receptor) during viral infections
Expansion of KLRC1-low/KLRC2-high NK cells in some infections may represent memory-like adaptation
Epigenetic reprogramming:
Changes in KLRC1 expression during infection may be associated with epigenetic modifications
These modifications could contribute to sustained functional alterations characteristic of memory-like responses
Implications for vaccination strategies:
Understanding KLRC1 regulation during infection could inform approaches to harness NK cell memory-like properties for vaccines
Potential for modulating KLRC1 expression to enhance vaccine-induced innate immune responses
Several promising therapeutic approaches targeting KLRC1 are in development:
Monoclonal antibody blockade:
Anti-KLRC1 antibodies that block interaction with HLA-E
This enhances NK cell activity against tumor cells that express HLA-E as an immune evasion mechanism
Potential for combination with other checkpoint inhibitors
Bispecific antibodies:
Antibodies that simultaneously engage KLRC1 and tumor antigens
These redirect KLRC1+ NK cells to tumor cells while potentially blocking inhibitory signaling
Adoptive cell therapy modifications:
Engineering NK cells with reduced or absent KLRC1 expression
CRISPR-Cas9 modification of KLRC1 in CAR-NK cells
Enhancing NK cell potency against tumors that overexpress HLA-E
Peptide-based approaches:
Peptides that interfere with KLRC1-HLA-E interactions
Potential for higher specificity and reduced side effects
These approaches aim to overcome tumor immune evasion mechanisms that exploit the KLRC1-HLA-E inhibitory axis.
KLRC1 expression patterns have potential as biomarkers in several clinical contexts:
Cancer immunotherapy response prediction:
KLRC1 expression levels on tumor-infiltrating NK cells may predict response to immunotherapies
The ratio of KLRC1+ to KLRC1- NK cells could indicate the degree of NK cell inhibition within the tumor microenvironment
Viral infection monitoring:
Changes in KLRC1 expression during viral infections may correlate with disease progression
Could help identify patients at risk for chronic viral persistence
Transplantation medicine:
KLRC1 phenotyping might help predict graft rejection risk
Could identify patients who would benefit from more intensive immunosuppression
Inflammation assessment:
KLRC1 expression patterns may serve as biomarkers for inflammation severity and type
Different inflammatory conditions may show characteristic KLRC1 expression signatures
Implementation would require standardized assays for KLRC1 detection, including flow cytometry panels or gene expression profiles optimized for clinical use.
Despite promising advances, several challenges remain in translating KLRC1 research into clinical applications:
Heterogeneity and compensation:
The heterogeneity of KLRC1 expression across NK cell subsets complicates therapeutic targeting
Compensatory inhibitory mechanisms may limit efficacy of KLRC1-targeted therapies
Off-target effects:
KLRC1 is expressed on certain T cell subsets in addition to NK cells
Effects of KLRC1 modulation on these cells must be carefully characterized
Monitoring challenges:
Need for standardized, clinically validated assays to monitor KLRC1 expression
Development of surrogate markers for KLRC1 pathway activity
Patient stratification:
Identifying which patients will benefit most from KLRC1-targeted approaches
Developing predictive biomarkers for response to KLRC1-modulating therapies
Combinatorial approaches:
Determining optimal combinations with other immunotherapies
Understanding potential synergies or antagonisms with existing treatments
Addressing these challenges will be crucial for successfully translating KLRC1 research into effective clinical applications.
Killer Cell Lectin-Like Receptor Subfamily C, Member 1 (KLRC1), also known as NKG2A, is a protein encoded by the KLRC1 gene in humans. This protein is a member of the killer cell lectin-like receptor family, which plays a crucial role in the immune system, particularly in the function of natural killer (NK) cells.
KLRC1 is a type II transmembrane protein characterized by the presence of a C-type lectin domain. It forms a heterodimeric complex with another protein, CD94 (KLRD1), which is essential for its function. This complex is involved in the recognition of MHC class I molecules, specifically HLA-E, on target cells. The interaction between KLRC1/CD94 and HLA-E inhibits the cytotoxic activity of NK cells, thereby preventing the destruction of healthy cells .
KLRC1 is predominantly expressed in NK cells and some subsets of T cells. Its expression is regulated by various cytokines and other signaling molecules that modulate the immune response. The gene encoding KLRC1 is located on chromosome 12, within a cluster of other killer cell lectin-like receptor genes .
The primary function of KLRC1 is to regulate the activity of NK cells. By interacting with HLA-E, KLRC1/CD94 inhibits the activation of NK cells, thus preventing the lysis of normal, healthy cells. This inhibitory mechanism is crucial for maintaining self-tolerance and preventing autoimmune reactions. Additionally, KLRC1 plays a role in the immune response to viral infections and tumors by modulating the activity of NK cells .
Alterations in the expression or function of KLRC1 have been associated with various diseases, including cancer and viral infections. For instance, downregulation of KLRC1 expression has been observed in certain types of cancer, which may contribute to the evasion of immune surveillance by tumor cells. Conversely, upregulation of KLRC1 expression can enhance the inhibitory signals in NK cells, potentially leading to impaired immune responses .
Given its role in regulating NK cell activity, KLRC1 is a potential target for therapeutic interventions aimed at modulating the immune response. Strategies to inhibit KLRC1/CD94 interactions could enhance the cytotoxic activity of NK cells against tumor cells and virally infected cells. Conversely, enhancing KLRC1 activity could be beneficial in preventing autoimmune diseases and reducing tissue damage during inflammatory responses .