KLRK1 Human

Killer Cell lectin-Like Receptor Subfamily K, Member 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Immune Function and Mechanisms

KLRK1 activates cytotoxic responses in:

  • NK cells

  • γδ T cells

  • CD8+ T cells

Key Pathways

MechanismOutcome
Ligand binding (e.g., MICA/B)Triggers perforin/granzyme release
DAP10 signalingEnhances cytokine production (IFN-γ, TNF-α)
Synergy with TCRAmplifies antigen-specific T cell responses

Prognostic Value

Lung Adenocarcinoma

  • High expression linked to:

    • Better 5-year survival (p = 0.003)

    • Early TNM stages (p < 0.001)

Immune Evasion Mechanisms

  • Regulatory T cells suppress KLRK1+ CD8+ T cell cytotoxicity .

  • Tumors expressing soluble NKG2D ligands (e.g., sMICB) show worse prognoses .

Therapeutic Implications

Immunotherapy Targets

  • Adoptive Cell Therapy: KLRK1+ CD8+ T cells exhibit superior tumor-killing capacity in vivo .

  • Checkpoint Combinations: Anti-PD-1 synergizes with KLRK1 activation in preclinical models .

Pharmacological Modulation

ApproachEffect
EZH2 inhibitionUpregulates NKG2D ligands on HCC cells
IL-2 conditioningEnhances KLRK1+ T cell cytotoxicity

Expression and Regulation

Tissue Distribution

  • Immune cells: NK cells (highest), CD8+ T cells, γδ T cells .

  • Malignant tissues: Overexpressed in 67% of HNSCC and 58% of lung adenocarcinomas .

Regulatory Factors

  • HPV infection increases KLRK1 expression in HNSCC (p < 0.05) .

  • Hypoxia downregulates NKG2D ligands in solid tumors .

Research Frontiers

  1. Biomarker potential for immune checkpoint inhibitor response .

  2. Engineering KLRK1 CAR-T cells for solid tumors .

  3. Small-molecule enhancers of NKG2D ligand expression .

Product Specs

Introduction
Killer Cell lectin-Like Receptor Subfamily K, Member 1 (KLRK1) is an activating receptor of significant recent interest. KLRK1 interacts with MICA and MICB, cell-surface molecules distantly related to MHC class I proteins. MICA and MICB expression is upregulated in response to cellular stress, such as during tumorigenesis and viral infection. Consequently, the KLRK1-MICA/B interaction plays a crucial role in the immune response to various pathologies.
Description
Recombinant human KLRK1, expressed in E. coli, is a single, non-glycosylated polypeptide chain containing 168 amino acids (73-216). This 19.2 kDa protein is fused to a 24 amino acid His-tag at the N-terminus and purified by proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered clear solution.
Formulation
KLRK1 protein solution (0.5mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), 10% glycerol and 0.4M Urea.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid multiple freeze-thaw cycles.
Purity
Greater than 90.0% as determined by SDS-PAGE.
Synonyms
CD314, D12S2489E, KLR, NKG2-D, NKG2D, Killer cell lectin-like receptor subfamily K member 1, NK cell receptor D, NKG2-D-activating NK receptor, CD314.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMIWSAVF LNSLFNQEVQ IPLTESYCGP CPKNWICYKN NCYQFFDESK NWYESQASCM SQNASLLKVY SKEDQDLLKL VKSYHWMGLV HIPTNGSWQW EDGSILSPNL LTIIEMQKGD CALYASSFKG YIENCSTPNT YICMQRTV.

Q&A

What is KLRK1 and what protein does it encode?

KLRK1 (Killer cell lectin-like receptor K1) is a gene located on chromosome 12 in humans that encodes NKG2D, an activating receptor belonging to the NKG2 family of C-type lectin-like receptors . NKG2D forms a homodimeric structure and functions as a key activating receptor in natural killer (NK) cells and certain T cell populations . As a transmembrane protein, NKG2D recognizes induced-self proteins from the MIC and RAET1/ULBP families that appear on the surface of stressed, malignantly transformed, or infected cells .

What is the structural organization of the NKG2D receptor complex?

The human NKG2D receptor complex assembles into a hexameric structure with distinctive functional domains. NKG2D itself forms a homodimer whose ectodomains serve for ligand binding . Each NKG2D monomer associates with a DAP10 dimer through ionic interactions between a positively charged arginine in the transmembrane segment of NKG2D and negatively charged aspartic acids within both transmembrane regions of the DAP10 dimer . This structural arrangement is crucial for signal transduction, as DAP10 functions as an adaptor protein that recruits the p85 subunit of PI3K and the Grb2-Vav1 complex, which are responsible for subsequent downstream signaling events .

What cell types express NKG2D in humans versus mice?

Expression patterns of NKG2D show both similarities and differences between humans and mice:

SpeciesExpressing Cell Types
HumansNK cells, γδ T cells, CD8+ αβ T cells
MiceNK cells, NK1.1+ T cells, γδ T cells, activated CD8+ αβ T cells, activated macrophages

This differential expression pattern has important implications for translational research and interpretation of mouse model studies in the context of human disease .

What genomic techniques are most effective for studying KLRK1 polymorphisms?

For studying KLRK1 polymorphisms, TaqMan® allelic discrimination assays have proven effective in genotyping populations for KLRK1 region variants . When designing such studies, researchers should consider analyzing multiple loci to identify haplotype blocks, as demonstrated in studies that genotyped 82 reference cell lines and 388 newborn samples at 10 KLRK1 region variants . This approach allows for the identification of specific haplotypes that may correlate with functional differences in natural cytotoxic activity and disease susceptibility. To enhance statistical power, larger sample sizes are recommended, particularly when studying associations with complex traits like cancer predisposition .

How can researchers effectively analyze KLRK1 expression as a prognostic biomarker in cancer?

To analyze KLRK1 as a prognostic biomarker in cancer, researchers should employ a multi-faceted approach:

What experimental approaches best elucidate the role of KLRK1/NKG2D in cancer immunosurveillance?

Investigating KLRK1/NKG2D's role in cancer immunosurveillance requires multiple complementary approaches:

  • Cohort studies: Design longitudinal studies measuring natural cytotoxic activity levels and correlating them with subsequent cancer development, similar to the Japanese cohort study that identified KLRK1 polymorphisms as genetic markers of cancer predisposition .

  • Genetic association studies: Analyze KLRK1 polymorphisms across diverse populations to identify variants associated with altered cancer risk .

  • Functional studies: Evaluate the impact of KLRK1 expression on cancer cell biology through:

    • Proliferation assays (e.g., CCK-8 assay)

    • Migration assays (e.g., wound healing assay)

    • Invasion assays

  • Mechanistic investigations: Examine interactions between NKG2D and its ligands (MIC and RAET1/ULBP family proteins) on stressed or malignant cells to understand recognition and activation dynamics .

  • Single-cell analysis: Employ high-dimensional techniques like CITE-seq with weighted nearest neighbors (WNN) method to integrate both RNA and protein expression data for comprehensive characterization of NK cell populations expressing NKG2D .

These approaches collectively provide a comprehensive understanding of how KLRK1/NKG2D contributes to immune surveillance against cancer development and progression.

How should researchers interpret conflicting data regarding KLRK1 expression and function across different cancer types?

When encountering conflicting data about KLRK1 expression and function across cancer types, researchers should consider several methodological approaches:

  • Context-specific analysis: Recognize that KLRK1's role may be context-dependent. For example, in lung adenocarcinoma, high KLRK1 expression is associated with improved survival outcomes, but this relationship may differ in other cancer types .

  • Subgroup stratification: Perform comprehensive subgroup analyses based on histological subtypes, stage, gender, and age. Research has shown that KLRK1's prognostic value varies significantly across these subgroups, being particularly strong in lung adenocarcinoma, stage I/II disease, older patients, and males .

  • Microenvironment considerations: Evaluate the tumor microenvironment composition, as the presence and functionality of NK cells and T cells expressing NKG2D can significantly influence outcomes regardless of receptor expression levels.

  • Ligand expression analysis: Assess the expression of NKG2D ligands (MIC and RAET1/ULBP family proteins) alongside receptor expression, as the receptor-ligand interaction is ultimately what determines functional outcomes .

  • Genetic variation impact: Consider how KLRK1 polymorphisms might influence receptor function and expression patterns across different populations .

  • Technical variations: Account for methodological differences between studies, including sample preparation, detection methods, threshold definitions for "high" versus "low" expression, and statistical approaches.

By systematically addressing these factors, researchers can better interpret seemingly contradictory findings and develop a more nuanced understanding of KLRK1's role across different cancer contexts.

What approaches can identify novel KLRK1 haplotypes with functional significance?

To identify novel KLRK1 haplotypes with functional significance, researchers should implement a multi-step approach:

  • Comprehensive genotyping: Analyze multiple KLRK1 region variants (at least 10) using high-throughput methods such as TaqMan® allelic discrimination assays across diverse population samples .

  • Haplotype determination: Employ statistical algorithms to infer haplotypes from genotype data and identify distinct haplotype blocks within the KLRK1 region .

  • Natural selection analysis: Implement tests for detecting evidence of natural selection at KLRK1 loci, including sex-specific selection patterns that may influence heterozygosity frequencies .

  • Functional correlation: Correlate identified haplotypes with natural cytotoxic activity levels using ex vivo functional assays of NK cell activity .

  • Disease association studies: Examine associations between identified haplotypes and cancer susceptibility or other immune-related conditions in case-control or cohort studies .

  • Sex-stratified analysis: Given evidence of sex-specific selection patterns, analyze haplotype effects separately in males and females to detect potential sex-specific functional implications .

  • Maternal-fetal interaction studies: Investigate the role of KLRK1 haplotypes in maternal-fetal recognition, particularly given observations suggesting that heterozygosity at certain loci might be unfavorable for male fetuses .

This systematic approach can reveal functionally significant KLRK1 haplotypes with potential implications for cancer immunosurveillance and reproductive biology.

What single-cell analysis techniques best characterize NKG2D-expressing NK cell populations?

For optimal characterization of NKG2D-expressing NK cell populations, high-dimensional single-cell analysis techniques offer superior resolution:

  • CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing): This technique enables simultaneous profiling of both transcriptome (RNA) and surface protein expression in the same cell, providing comprehensive characterization of NK cell heterogeneity .

  • Weighted Nearest Neighbors (WNN) method: This analytical approach effectively integrates both RNA and protein expression data to identify distinct NK cell subsets with unique molecular signatures .

  • Comprehensive antibody panels: Utilize extensive antibody-derived tags (ADTs) covering multiple NK cell markers, including NKG2D/CD314, to thoroughly characterize surface protein expression .

  • Isolation strategy: Begin with isolation of non-proliferating NK cells at baseline before reclustering to elucidate foundational heterogeneity among blood NK cells .

  • Integrated multi-dataset analysis: Combine scRNA-seq data from multiple datasets while implementing proper batch correction to ensure findings are robust across different donor samples .

  • Signature scoring: Apply established gene signatures (e.g., the 13-gene signature including KLRC1, KLRC3, KLRD1, KLRF1) to validate NK cell populations and subpopulations .

This integrated approach has successfully identified three primary (NK1, NK2, and NK3) and six more refined NK cell subsets, revealing significant heterogeneity within NKG2D-expressing populations .

How should researchers optimize recombinant KLRK1 protein production for functional studies?

For optimal production of recombinant KLRK1 protein for functional studies, researchers should consider the following methodological guidelines:

  • Expression system selection: E. coli is an established system for producing non-glycosylated KLRK1 human recombinant protein, yielding a single polypeptide chain containing 168 amino acids (73-216 a.a.) with a molecular mass of 19.2kDa .

  • Fusion tag optimization: Consider implementing a 24 amino acid His-tag at the N-terminus to facilitate purification while maintaining protein functionality .

  • Purification methodology: Employ proprietary chromatographic techniques to achieve high purity levels (>90% as determined by SDS-PAGE) .

  • Buffer formulation: Optimize the storage buffer (e.g., 20mM Tris-HCl buffer (pH8.0), 10% glycerol, and 0.4M Urea) to maintain protein stability .

  • Storage conditions: Store at 4°C if the entire vial will be used within 2-4 weeks, or frozen at -20°C for longer periods .

  • Stability enhancement: For long-term storage, add a carrier protein (0.1% HSA or BSA) and avoid multiple freeze-thaw cycles .

  • Quality control: Confirm protein identity and purity through mass spectrometry and SDS-PAGE analysis .

  • Functional validation: Verify biological activity of the recombinant protein through binding assays with known ligands from the MIC and RAET1/ULBP families .

By following these guidelines, researchers can produce high-quality recombinant KLRK1 protein suitable for a range of functional studies, including receptor-ligand interaction assays, structural analyses, and immunological investigations.

What assays most accurately measure NKG2D-mediated NK cell activation in cancer models?

To accurately measure NKG2D-mediated NK cell activation in cancer models, researchers should employ multiple complementary assays:

  • Cytotoxicity assays: Use chromium release or flow cytometry-based assays to quantify NK cell-mediated killing of target cancer cells, with specific conditions to isolate NKG2D-dependent cytotoxicity (e.g., blocking antibodies against NKG2D or its ligands).

  • Cytokine production measurement: Quantify IFN-γ, TNF-α, and GM-CSF production by ELISA or intracellular cytokine staining following NKG2D engagement.

  • Degranulation assays: Measure CD107a (LAMP-1) surface expression as a marker of NK cell degranulation in response to NKG2D-mediated activation.

  • Calcium flux assays: Monitor intracellular calcium mobilization following NKG2D receptor engagement to assess early activation events.

  • Signaling pathway analysis: Evaluate phosphorylation of downstream signaling molecules (PI3K, Grb2-Vav1 complex) by western blotting or phospho-flow cytometry .

  • In vivo models: Develop orthotopic or xenograft tumor models with genetic manipulation of KLRK1 expression to assess the impact on tumor growth, metastasis, and immune infiltration .

  • Ex vivo assays: Measure natural cytotoxic activity levels of peripheral blood lymphocytes from patients with various KLRK1 genotypes to correlate genotype with functional outcomes .

These methodologies collectively provide a comprehensive assessment of NKG2D-mediated NK cell activation in cancer contexts, allowing researchers to understand both the extent and mechanisms of anti-tumor responses.

How might KLRK1/NKG2D polymorphisms impact cancer immunotherapy response?

KLRK1/NKG2D polymorphisms may significantly influence cancer immunotherapy response through several mechanisms:

  • Baseline NK cell functionality: Different KLRK1 haplotypes have been associated with varying levels of natural cytotoxic activity, which could predispose patients to differential responses to immunotherapies that rely on NK cell function .

  • Receptor-ligand interaction dynamics: Polymorphisms in the KLRK1 gene might alter the binding affinity or specificity of NKG2D for its ligands, potentially affecting recognition of stress-induced molecules on tumor cells .

  • Predictive biomarker potential: Genotyping patients for KLRK1 polymorphisms before immunotherapy could help predict response rates, as certain haplotypes might correlate with improved clinical outcomes .

  • Combination therapy design: Understanding how KLRK1 variants influence NK cell function could inform the design of combination immunotherapies that specifically enhance NKG2D-mediated anti-tumor responses in patients with less favorable genotypes.

  • Sex-specific considerations: Given evidence of sex-specific natural selection at certain KLRK1 loci, immunotherapy approaches might need to be tailored differently for male and female patients carrying particular polymorphisms .

  • Population-specific strategies: The frequency of KLRK1 haplotypes varies across different populations, suggesting that immunotherapy strategies might benefit from population-specific genetic considerations .

Research into these aspects could ultimately lead to personalized immunotherapy approaches based on KLRK1 genotyping, potentially improving response rates and clinical outcomes.

What experimental designs best investigate the relationship between KLRK1 expression and cancer prognosis?

To rigorously investigate the relationship between KLRK1 expression and cancer prognosis, researchers should implement the following experimental design elements:

This multi-faceted approach has successfully identified KLRK1 as an independent prognostic factor in lung adenocarcinoma, where high expression indicates better survival outcomes .

How can single-cell analysis of NK cell heterogeneity advance our understanding of KLRK1 functionality?

Single-cell analysis of NK cell heterogeneity has transformative potential for understanding KLRK1 functionality through several key advances:

  • Identification of distinct NK cell subpopulations: High-dimensional single-cell analysis has revealed three primary NK cell subsets (NK1, NK2, and NK3) that can be further divided into six refined subpopulations, each with potentially unique KLRK1/NKG2D expression patterns and functionality .

  • Integration of transcriptional and proteomic signatures: The weighted nearest neighbors (WNN) method enables effective integration of both RNA and protein expression data, providing a comprehensive view of NK cell states and their relationship to KLRK1 function .

  • Correlation with clinical outcomes: Single-cell profiling of NK cells from patients with different disease outcomes can reveal how specific NK subpopulations expressing KLRK1 correlate with clinical responses, potentially identifying cellular biomarkers of treatment efficacy.

  • Dynamic receptor expression analysis: Single-cell approaches can track changes in KLRK1 expression within NK cell populations during disease progression or treatment, revealing adaptive responses that may not be apparent in bulk analyses.

  • Identification of co-expression patterns: By examining the co-expression of KLRK1 with other receptors and functional molecules at the single-cell level, researchers can uncover receptor complexes and signaling networks that modulate NKG2D function .

  • Cell lineage and developmental insights: Single-cell RNA-seq data can reveal developmental trajectories of NKG2D-expressing NK cells, providing insights into the ontogeny of functional NK cell subsets .

  • Donor-specific NK cell signatures: Single-cell profiling has demonstrated that NK cell signatures, particularly within adaptive NK cells in HCMV+ individuals, show considerable heterogeneity both within and between individuals, reflecting clonal rather than functionally distinct populations .

These insights from single-cell analysis are crucial for developing more precise immunotherapeutic approaches targeting the KLRK1/NKG2D axis.

What emerging technologies might revolutionize KLRK1/NKG2D research in the next decade?

Several emerging technologies are poised to revolutionize KLRK1/NKG2D research in the coming decade:

  • Spatial transcriptomics and proteomics: These technologies will enable researchers to analyze KLRK1 expression and NKG2D-expressing cells within their native tissue context, revealing spatial relationships between NK cells and their target cells in tumors and infected tissues.

  • CRISPR-based genetic screens: High-throughput CRISPR screens will facilitate the identification of genes that modulate KLRK1 expression and NKG2D function, potentially uncovering novel regulatory mechanisms and therapeutic targets.

  • Organoid and microfluidic systems: Advanced 3D culture systems will allow more physiologically relevant studies of NKG2D-mediated NK cell interactions with target cells under controlled conditions that better recapitulate in vivo environments.

  • Single-cell multi-omics integration: Next-generation single-cell technologies that simultaneously profile the genome, epigenome, transcriptome, and proteome of individual cells will provide unprecedented insights into the molecular mechanisms governing KLRK1 expression and function .

  • Live-cell imaging techniques: Super-resolution microscopy and intravital imaging advancements will enable real-time visualization of NKG2D-mediated immunological synapse formation and signaling events in living cells and tissues.

  • Artificial intelligence and machine learning: These computational approaches will accelerate the analysis of complex datasets, facilitating the identification of subtle patterns in KLRK1 expression and NKG2D function across different contexts.

  • Precision genome editing: Advanced CRISPR-based technologies will enable precise modification of KLRK1 polymorphisms to directly test their functional significance in primary human cells.

  • Therapeutic antibody engineering: Novel antibody technologies will facilitate the development of agents that can selectively enhance or inhibit NKG2D function for therapeutic applications in cancer and infectious diseases.

These technological advances will collectively transform our understanding of KLRK1/NKG2D biology and accelerate the translation of research findings into clinical applications.

Product Science Overview

Introduction

Killer Cell Lectin-Like Receptor Subfamily K, Member 1 (KLRK1), also known as NKG2D, is a protein encoded by the KLRK1 gene. This protein is a member of the C-type lectin family and plays a crucial role in the immune system, particularly in the function of natural killer (NK) cells and certain T cells .

Gene and Protein Structure

The KLRK1 gene is located on chromosome 12p13.2 and encodes a type II transmembrane protein characterized by an extracellular C terminus and a C-type lectin domain . The protein is expressed as a major 1.8-kb and a minor 3.2-kb transcript in NK cell lines and some T-cell lines .

Function

KLRK1, or NKG2D, is an activating receptor that triggers NK cells and costimulates CD8-positive T cells. It is normally absent from almost all CD4-positive T cells but is present on most gamma/delta T cells . The receptor binds to a diverse family of ligands, including MHC class I chain-related A and B proteins and UL-16 binding proteins . These ligand-receptor interactions result in the activation of NK and T cells, promoting cytolytic responses against tumor cells and virus-infected cells .

Clinical Significance

The surface expression of KLRK1 ligands is crucial for the recognition of stressed cells by the immune system. This makes KLRK1 and its ligands therapeutic targets for the treatment of immune diseases and cancers . The receptor’s ability to activate immune responses against tumor cells highlights its potential in cancer immunotherapy .

Research and Applications

Research on KLRK1 has focused on its role in immune regulation and its potential therapeutic applications. Studies have shown that NKG2D is expressed on gamma/delta T cells, CD8-positive T cells, and NK cells, and its interaction with ligands like MICA can activate cytolytic responses against tumor cells . This receptor-ligand interaction is a promising target for developing new cancer treatments.

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.