KLRB1 is a type II transmembrane protein with an extracellular C-type lectin domain, a transmembrane region, and a cytoplasmic tail. Its interaction with ligands like CLEC2D (LLT1) modulates immune cell cytotoxicity and cytokine secretion . The Sf9-baculovirus system is widely used for recombinant protein production due to its capacity for post-translational modifications.
Key characteristics of KLRB1 expressed in Sf9:
Construct Design: Soluble KLRB1 variants lacking transmembrane domains were tested, focusing on the extracellular lectin-like domain (residues 67–225) with varying N-terminal stalk lengths .
Expression Challenges: Initial trials in Sf9 cells showed lower yields compared to HEK293 systems, prompting optimization of expression vectors and truncation designs .
Parameter | Sf9 Insect Cells | HEK293 Mammalian Cells |
---|---|---|
Yield | Lower | Higher |
Glycosylation | Insect-specific | Non-complex (GnTI⁻ cells) |
Tags Tested | SUMO, TRX, GST, MsyB | None |
Purity | >85% (SDS-PAGE verified) | Homogeneous, crystallizable |
Functional Use | Structural studies | Immunological assays |
Construct Optimization: Truncated variants (e.g., residues 97–225) improved solubility but reduced yield in Sf9 .
Protein Properties: Sf9-derived KLRB1 retained ligand-binding capability, confirmed via CLEC2D interaction assays .
Functional Role: KLRB1 acts as an inhibitory receptor in NK cells and a co-stimulatory molecule in T cells .
KLRB1 is a therapeutic target due to its role in tumor immune evasion:
GBM and Other Cancers: KLRB1–CLEC2D interactions suppress cytotoxic T-cell activity in glioblastoma (GBM), renal, and lung cancers .
Therapeutic Strategies: CRISPR-Cas9 knockout of KLRB1 enhances T-cell cytotoxicity in tumors .
While Sf9 expression offers scalability, HEK293 systems currently outperform it in yield and homogeneity for KLRB1 . Future work may focus on:
Killer Cell Lectin Like Receptor B1, Natural Killer Cell Surface Protein P1A, Killer Cell Lectin-Like Receptor Subfamily B, Member 1, C-Type Lectin Domain Family 5 Member B, HNKR-P1A, NKR-P1A, CLEC5B, NKRP1A, Killer Cell Lectin-Like Receptor Subfamily B Member 1, CD161 Antigen, NKR-P1, CD161, NKR.
Sf9, Baculovirus cells.
ADPQKSSIEK CSVDIQQSRN KTTERPGLLN CPIYWQQLRE KCLLFSHTVN PWNNSLADCS TKESSLLLIR DKDELIHTQN LIRDKAILFW IGLNFSLSEK NWKWINGSFL NSNDLEIRGD AKENSCISIS QTSVYSEYCS TEIRWICQKE LTPVRNKVYP DSLEPKSCDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS LSPGKHHHHH H.
KLRB1 (Killer Cell Lectin-Like Receptor B1), also known as CD161, is a C-type lectin-like receptor primarily expressed on natural killer (NK) cells, T cells, and NKT cells. It functions as an inhibitory receptor on NK cells when binding to its cognate ligand CLEC2D (also known as LLT1), inhibiting cytotoxic function and cytokine secretion . On T and NKT cells, it serves as a co-stimulatory receptor promoting IFNγ secretion . Most notably, KLRB1 is expressed on Th17 and Tc17 lymphocytes where it presumably promotes targeting into CLEC2D-expressing immunologically privileged niches .
Methodologically, KLRB1 expression can be quantified using flow cytometry, quantitative PCR, and single-cell RNA sequencing. Studies have shown that KLRB1 is only expressed by small subpopulations of human blood T cells, but its expression can be induced in specific environments such as tumor microenvironments .
KLRB1 exhibits distinct functional profiles across different immune cell populations:
In NK cells:
Functions as an inhibitory receptor
Binding to CLEC2D inhibits cytotoxic function and cytokine secretion
Expression decreases after interferon-β treatment in MS patients
In T cells:
Serves as a co-stimulatory receptor promoting IFNγ secretion
CD4+ KLRB1+ T cells exhibit greater IL-17A, IL-21, IL-22, and IFN-γ secretion upon stimulation
Significantly higher proportions of CCR5+, CCR2+, CX3CR1+, CCR6+, and CXCR3+ cells are found among CD4+ KLRB1+ T cells compared to CD4+ KLRB1- T cells
Marks activated T cells, with CD25 and Ki-67 expression significantly greater in CD4+ KLRB1+ T cells than in CD4+ KLRB1- T cells
These functional differences suggest context-specific roles in immune regulation and highlight the importance of studying KLRB1 function in isolated cell populations using techniques such as cell sorting followed by functional assays.
Human KLRB1 is a C-type lectin-like receptor with an extracellular lectin-like domain connected to a transmembrane region via a stalk region. The natural form is expressed on cell surfaces, but soluble forms can be produced for research purposes .
Key structural features include:
C-type lectin-like domain responsible for ligand binding
N-terminal stalk region with variable length affecting expression efficiency
Potential glycosylation sites that impact function and stability
KLRB1 interacts with its cognate protein ligand CLEC2D (LLT1), and this interaction has significant immunological consequences. Binding of CLEC2D to the KLRB1 receptor inhibits the cytotoxic function of NK cells and cytokine secretion . The interaction appears to have different effects in T cells, potentially serving a co-stimulatory function.
The detailed structural basis of this interaction can be studied using purified recombinant proteins. Research has shown that soluble KLRB1 can be produced that is "homogeneous, deglycosylatable, crystallizable and monomeric in solution," as confirmed by size-exclusion chromatography, multi-angle light scattering, and analytical ultracentrifugation .
Sf9 insect cells offer several advantages for KLRB1 expression:
Advantages:
Can perform post-translational modifications including basic glycosylation
Higher expression levels than many mammalian systems
Better protein folding capacity than bacterial systems
Cost-effective compared to mammalian cell culture
Scalable in suspension culture
Challenges:
Insect cell glycosylation differs from human patterns
Requires optimization of construct design, particularly the length of the N-terminal stalk region
May produce heterogeneous protein populations
Variable expression levels between batches
Research has shown that expression of soluble KLRB1 in Sf9 cells requires careful optimization of constructs. Studies have tested libraries of expression constructs in pOPING vector containing the extracellular lectin-like domain with different lengths of the putative N-terminal stalk region . This approach helps identify the most productive variant for structural and functional studies.
Expression tags can significantly impact KLRB1 solubility, yield, and functionality when produced in heterologous systems:
Tag | Impact on Solubility | System Tested | Functional Considerations |
---|---|---|---|
SUMO | Improved solubility | E. coli | May affect native structure |
TRX | Improved solubility | E. coli | Redox-active tag that may assist folding |
GST | Improved solubility | E. coli | Large tag that may affect function |
MsyB | Improved solubility | E. coli | Novel tag with varying effects |
His-tag | Minimal interference | Multiple systems | Allows affinity purification |
Researchers have tested various protein tags (SUMO, TRX, GST, MsyB) on the expression of soluble KLRB1 in E. coli, but encountered challenges with protein folding and solubility . The choice of tag should consider:
Impact on protein folding and native structure
Ease of tag removal after purification
Potential interference with ligand binding
Effect on crystallization properties
For structural and functional studies, smaller tags with minimal impact on native structure are preferred, and tag removal may be necessary to ensure physiologically relevant results.
Optimal construct design for soluble KLRB1 expression requires careful consideration of domain boundaries and expression systems:
The most successful approach involved:
Using the extracellular lectin-like domain with optimized N-terminal stalk region length
Testing multiple constructs with varying boundaries in expression vector libraries
Evaluating expression in different systems (Sf9 and HEK293 cells)
The highest yield of soluble, functional KLRB1 was achieved by:
Stable expression in suspension-adapted HEK293S GnTI- cells
Utilizing pOPINGTTneo expression vector
Including properly defined boundaries of the extracellular domain
This approach produced protein that was "homogeneous, deglycosylatable, crystallizable and monomeric in solution," as confirmed by size-exclusion chromatography, multi-angle light scattering, and analytical ultracentrifugation .
For researchers seeking to express KLRB1, a systematic approach testing multiple constructs with precisely defined domain boundaries is recommended, with particular attention to the length of the N-terminal stalk region.
KLRB1 expression shows significant alterations in multiple autoimmune conditions:
In Multiple Sclerosis (MS):
Blood from MS patients showed higher KLRB1 expression compared to healthy controls (P<0.001)
KLRB1 expression decreased significantly (P<0.001) after interferon-β treatment
Expression mainly decreased in NK cells of interferon-β treated patients
A genome-wide association study identified a marginally significant association between the KLRB1 gene SNP rs4763655 and MS (P=0.046, odds ratio=1.06 (1.00-1.13))
In Primary Sjögren's Syndrome (pSS):
KLRB1 expression in CD4+ T cells was markedly elevated compared to healthy controls
Expression significantly correlated with clinical disease indicators
KLRB1+ CD4+ T cells exhibited greater IL-17A, IL-21, IL-22, and IFN-γ secretion upon stimulation
ROC curve analysis suggested potential utility as an auxiliary diagnostic marker for pSS
These disease-specific alterations in KLRB1 expression suggest it plays important roles in autoimmune pathogenesis, potentially by modulating T cell activation, cytokine production, or tissue targeting through chemokine receptor regulation.
KLRB1 has emerged as a significant factor in cancer immunology, with particular relevance to glioblastoma multiforme (GBM):
In GBM microenvironment:
KLRB1 was identified as one of the top gene products expressed in the tumor
Expression is induced in T cells within the GBM microenvironment
Functions as an inhibitory receptor for human T cells by binding to the CLEC2D ligand on tumor cells
Inactivation of the KLRB1 gene in primary human T cells greatly enhances their cytotoxic function within tumors in a humanized mouse model
Mechanistically, KLRB1-CLEC2D interaction appears to create an immunosuppressive axis in GBM:
Binding of CLEC2D to the KLRB1 receptor inhibits cytotoxic function
Suppresses cytokine secretion in tumor-infiltrating lymphocytes
May contribute to immune evasion by GBM cells
These findings have led to therapeutic development strategies focusing on blocking KLRB1-CLEC2D interaction or reducing KLRB1 expression in tumor-infiltrating lymphocytes as potential approaches for enhancing anti-tumor immunity .
Interferon-β (IFN-β) treatment in multiple sclerosis patients has a significant modulatory effect on KLRB1 expression:
Key findings:
KLRB1 expression decreased significantly (P<0.001) after IFN-β treatment in MS patients
The decrease in expression was predominantly observed in NK cells rather than T cells
This alteration in expression correlates with treatment response
This differential regulation suggests a potential mechanism of action for IFN-β in MS therapy:
Reduction of KLRB1 expression may alter NK cell function
Changed receptor profile could affect immune cell trafficking or activation
Modified cytokine production patterns in KLRB1-expressing cells
The cell-type specific effect (primarily in NK cells) highlights the complex immunomodulatory actions of IFN-β and suggests that monitoring KLRB1 expression could potentially serve as a biomarker for treatment response in MS patients.
Several therapeutic approaches targeting KLRB1 are being developed for cancer treatment, particularly for glioblastoma multiforme (GBM):
Blocking KLRB1-CLEC2D Interaction:
Reducing KLRB1 Expression:
Modified T Cell Approaches:
A patent application describes methods involving "administering an agent capable of blocking the interaction of KLRB1 with its ligand," which may include humanized or chimeric antibodies targeting either KLRB1 or its ligands .
Preliminary data from a humanized mouse model of GBM demonstrated that inactivation of the KLRB1 gene in primary human T cells greatly enhanced their cytotoxic function within tumors, providing proof-of-concept for this approach .
In contrast to cancer therapy where KLRB1 inhibition is pursued, autoimmune disease treatment may benefit from KLRB1 pathway activation:
For chronic inflammatory and autoimmune diseases:
Increasing expression of KLRB1
Increasing expression of genes encoding CD161 ligands
Activating or stimulating cell signaling through CD161
The divergent approaches for cancer versus autoimmune conditions reflect the complex, context-dependent roles of KLRB1 in immune regulation. While elevated KLRB1 expression is associated with diseases like multiple sclerosis and primary Sjögren's syndrome , its activation may potentially limit inflammatory responses in certain contexts.
Therapeutic development would require:
Precise understanding of KLRB1 function in specific autoimmune conditions
Cell-type targeted delivery of KLRB1-modulating agents
Careful monitoring of immune responses to avoid unintended consequences
This bidirectional therapeutic potential highlights the importance of thoroughly characterizing KLRB1 pathway function in different disease contexts.
Different approaches to KLRB1 modulation offer distinct advantages and limitations:
Approach | Advantages | Limitations | Technology Readiness |
---|---|---|---|
Antibody-based | - Established manufacturing - Tunable half-life - No genetic modification - Reversible | - Limited tissue penetration - Potential immunogenicity - Continuous dosing required | More advanced clinical development |
Genetic modification | - Durable effect - Cell-specific targeting - One-time treatment - Potential for precise editing | - Delivery challenges - Off-target effects - Irreversibility concerns - Regulatory hurdles | Emerging technology |
Genetic approaches may use CRISPR-Cas systems (Cas9, Cas12, Cas13, or Cas14) or other programmable nucleic acid modifying agents like zinc fingers, TALEs, or meganucleases . These techniques can be applied to modify T cells ex vivo before reinfusion, potentially in combination with other genetic modifications to enhance anti-tumor activity.
Antibody approaches can target either KLRB1 or its ligands (such as CLEC2D) and may include humanized or chimeric antibodies or antibody fragments . They offer the advantage of established development pathways but may face challenges in accessing certain anatomical locations like the central nervous system in GBM treatment.
Single-cell technologies offer powerful approaches to elucidate KLRB1 function in complex tissue environments:
Key methodological applications:
Single-cell RNA sequencing (scRNA-seq):
Single-cell protein analysis:
Mass cytometry (CyTOF) enables simultaneous detection of dozens of proteins
Multiparameter flow cytometry allows sorting and functional analysis of KLRB1+ subsets
Spatial transcriptomics:
Maps KLRB1-expressing cells within tissue microenvironments
Provides context for receptor-ligand interactions in situ
Single-cell multi-omics:
Combined analysis of transcriptome, proteome, and epigenome
Reveals regulatory mechanisms controlling KLRB1 expression
Research applications include:
Characterizing KLRB1+ cell heterogeneity in disease tissues
Tracking changes in KLRB1-expressing populations during disease progression
Monitoring treatment effects on KLRB1+ cells
Studies have already utilized publicly available single-cell RNA-Seq data from primary Sjögren's syndrome patient PBMCs to analyze KLRB1 expression in T cells, demonstrating the value of these approaches .
Investigating KLRB1 signaling pathways requires sophisticated experimental designs:
Receptor engagement approaches:
Plate-bound or soluble antibodies against KLRB1
Recombinant CLEC2D ligand stimulation
Co-culture with CLEC2D-expressing cells
Signaling readouts:
Phosphoprotein analysis by flow cytometry
Western blotting for pathway components
Calcium flux measurement
Transcriptional reporter assays
Genetic manipulation:
CRISPR-based knockout of KLRB1 or downstream components
Overexpression systems with wildtype or mutant receptors
Domain swapping to identify functional regions
Advanced imaging:
Live-cell imaging to visualize receptor clustering
FRET-based approaches to detect molecular interactions
Super-resolution microscopy to study receptor organization
Proteomics:
Immunoprecipitation coupled with mass spectrometry
Proximity labeling techniques to identify interacting partners
Phosphoproteomics to map signaling cascades
Comparative analysis between NK cells (where KLRB1 is inhibitory) and T cells (where it can be co-stimulatory) is particularly valuable for understanding context-dependent signaling mechanisms and identifying cell-specific signaling partners.
Structural biology provides crucial insights for developing KLRB1-targeted therapeutics:
Protein crystallography:
Cryo-electron microscopy:
Visualizes KLRB1-ligand complexes in different conformational states
Less dependent on crystallization, suitable for membrane-associated forms
NMR spectroscopy:
Maps binding interfaces through chemical shift perturbations
Studies dynamics of receptor-ligand interactions
Molecular dynamics simulations:
Models receptor-ligand interactions in silico
Predicts effects of mutations or small molecule binding
Structure-guided drug design:
Rational design of antibodies targeting specific epitopes
Development of small molecule modulators of KLRB1-CLEC2D interaction
Protein engineering to create optimized soluble decoys
These approaches benefit from high-quality recombinant KLRB1 protein. Research has successfully produced soluble KLRB1 that is "homogeneous, deglycosylatable, crystallizable and monomeric in solution," providing a foundation for structural studies .
Several critical questions remain regarding KLRB1's tissue-specific functions:
Nervous system immunology:
How does KLRB1 expression in T cells contribute to GBM pathogenesis?
Is KLRB1 involved in other neurological disorders beyond MS?
What role might KLRB1 play in neuroinflammation more broadly?
Exocrine gland immunity:
What mechanisms drive elevated KLRB1 expression in Sjögren's syndrome?
How do KLRB1+ T cells contribute to salivary and lacrimal gland pathology?
Could KLRB1 expression serve as a biomarker for disease progression?
Tissue microenvironment regulation:
How does the tumor microenvironment induce KLRB1 expression in T cells?
What factors regulate CLEC2D expression in different tissues?
How does KLRB1-CLEC2D interaction influence immune cell trafficking?
Therapeutic targeting considerations:
Which tissues would benefit most from KLRB1 pathway modulation?
How can tissue-specific delivery of KLRB1-targeting agents be achieved?
What biomarkers can predict response to KLRB1-targeted therapies?
Addressing these questions will require integrating advanced technologies like spatial transcriptomics, tissue-specific genetic models, and sophisticated organoid systems that recapitulate the complex cellular interactions in different tissue environments.
KLRB1 targeting shows promise as a component of combination immunotherapy approaches:
Checkpoint inhibitor combinations:
CAR-T cell therapy enhancement:
KLRB1 gene inactivation could improve CAR-T function in solid tumors
Potential for dual targeting of KLRB1 and tumor-specific antigens
May improve persistence and activity in immunosuppressive microenvironments
Adoptive cell therapy optimization:
Bispecific antibody approaches:
One arm targeting KLRB1 or CLEC2D
Second arm engaging effector cells or additional tumor antigens
Could provide targeted modulation of the pathway
Research indicates that "inactivation of the KLRB1 gene in primary human T cells greatly enhances their cytotoxic function within tumors" in a humanized mouse model of GBM, suggesting significant potential for combining KLRB1 targeting with other immunotherapy approaches .
Artificial intelligence approaches offer transformative potential for KLRB1 research:
Target identification and validation:
Mining multi-omics data to identify patient subsets likely to benefit from KLRB1 targeting
Predicting functional consequences of KLRB1 genetic variants
Identifying novel KLRB1-expressing cell populations in disease contexts
Therapeutic design:
Structure-based AI models to design optimal antibodies against KLRB1 or CLEC2D
Predicting off-target effects of genetic modification approaches
Optimizing CRISPR guide RNA design for KLRB1 targeting
Clinical translation:
Patient stratification algorithms incorporating KLRB1 expression data
Predicting response to KLRB1-targeted therapies
Monitoring treatment efficacy through image analysis of KLRB1+ cells
Mechanistic understanding:
Network analysis to place KLRB1 in broader signaling contexts
Predicting dynamic changes in KLRB1 expression under different conditions
Modeling cell-cell interactions mediated by KLRB1-CLEC2D binding
As research progresses, integrating AI approaches with experimental validation will likely accelerate both fundamental discoveries about KLRB1 biology and therapeutic development targeting this pathway in cancer and autoimmune diseases.
Killer Cell Lectin-Like Receptor Subfamily B, Member 1 (KLRB1), also known as CD161, is a protein encoded by the KLRB1 gene in humans. This protein plays a crucial role in the immune system, particularly in the function of natural killer (NK) cells and certain T cells. The recombinant form of this protein, produced in Sf9 insect cells, is used in various research and therapeutic applications.
The KLRB1 gene is located on chromosome 12p13.31 in humans . The protein encoded by this gene is a type II transmembrane protein, meaning it has an external C terminus. It contains an extracellular domain with several motifs characteristic of C-type lectins, a transmembrane domain, and a cytoplasmic domain . The recombinant form produced in Sf9 cells is a single, glycosylated polypeptide chain containing 401 amino acids and has a molecular mass of 45.7 kDa .
KLRB1 is primarily expressed on the surface of NK cells and certain T cells. It acts as an inhibitory receptor, playing a significant role in regulating the cytotoxic activity of NK cells. The protein binds to specific ligands, such as CLEC2D/LLT1, which leads to the inhibition of NK cell-mediated cytotoxicity and interferon-gamma secretion . This regulatory function is crucial for maintaining immune homeostasis and preventing excessive immune responses.
The recombinant form of KLRB1 is produced using the baculovirus expression system in Sf9 insect cells. This system is widely used for producing recombinant proteins due to its ability to perform post-translational modifications similar to those in mammalian cells. The recombinant KLRB1 produced in Sf9 cells is tagged with a 239 amino acid hIgG-His-tag at the C-terminus and purified using proprietary chromatographic techniques .
Recombinant KLRB1 has several applications in research and therapeutic settings. It is used to study the mechanisms of NK cell regulation and to develop therapies targeting immune-related diseases. Additionally, it serves as a valuable tool in immunological assays and the development of diagnostic tests.