RAET1L expression is stress-induced and context-dependent:
HCMV-encoded UL16 sequesters RAET1L intracellularly, preventing surface exposure and immune recognition . This immune evasion mechanism highlights RAET1L’s role as a viral target.
RAET1L binds NKG2D, a receptor expressed on NK cells, CD8+ T cells, and γδ T cells, activating cytotoxic responses . Key interactions:
Binding Partner | Function |
---|---|
NKG2D | Strong binding induces NK-cell cytotoxicity and cytokine release |
UL16 (HCMV) | Retains RAET1L intracellularly, blocking surface presentation |
Ligand | NKG2D Binding Strength | UL16 Binding Strength |
---|---|---|
RAET1L | High | High |
ULBP5 | Moderate | Moderate |
ULBP2 | Low | Low |
Data adapted from . RAET1L’s strong dual binding to NKG2D and UL16 underscores its evolutionary pressure during viral infections.
RAET1L polymorphisms may influence alloreactive immune responses in hematologic malignancies treated with allogeneic stem cell transplantation, though specific mechanisms remain under investigation .
Recombinant RAET1L (e.g., PRO-2723, R&D Systems) is used in:
The protein RAET1L, which stands for Retinoic Acid Early Transcript 1L, functions as a ligand for the NKG2D receptor when the body is under stress. It anchors to cell membranes through a GPI anchor. When the human cytomegalovirus (HCMV) infects a cell, it increases the expression of RAET1L. However, HCMV also manipulates RAET1L's function. The virus uses its own protein, UL16, to trap RAET1L inside the infected cell, preventing it from reaching the cell surface and interacting with the immune system. Compared to other NKG2D ligands, RAET1L is found in a narrower range of cell lines and human tissues.
This product is a recombinant human RAET1L protein produced in Sf9 insect cells using a baculovirus expression system. It consists of a single polypeptide chain with 202 amino acids (specifically, amino acids 26 to 218) and has a molecular weight of 22.9 kDa. The protein is glycosylated, meaning it has sugar molecules attached. For purification and detection purposes, a 6-amino acid histidine tag (His-tag) has been added to the C-terminus of the protein. The protein is purified using proprietary chromatographic techniques.
The product appears as a clear and colorless solution that has been sterilized by filtration.
The RAET1L protein is supplied in a solution containing 1 mg of protein per milliliter (1mg/ml). The solution also contains 10% glycerol and Phosphate-Buffered Saline (PBS) with a pH of 7.4.
To ensure product stability, store the vial at 4°C if you plan to use it within 2-4 weeks. For longer-term storage, freezing the product at -20°C is recommended. If storing for an extended period, consider adding a carrier protein such as HSA or BSA (0.1%) to protect the protein. It is important to minimize the number of times the product is frozen and thawed to maintain protein integrity.
The purity of the RAET1L protein is greater than 95%, as determined by SDS-PAGE analysis.
The biological activity of this product is determined by its ability to bind to human KLRK1 in a functional ELISA assay (using product code pro-2474).
early transcript 1L, ULBP6, UL16-binding protein 6, RAET1L, retinoic acid early transcript 1L protein, retinoic acid early transcript 1L
Sf9, Baculovirus cells.
ADPRRDDPHS LCYDITVIPK FRPGPRWCAV QGQVDEKTFL HYDCGNKTVT PVSPLGKKLN VTMAWKAQNP VLREVVDILT EQLLDIQLEN YTPKEPLTLQ ARMSCEQKAE GHSSGSWQFS IDGQTFLLFD SEKRMWTTVH PGARKMKEKW ENDKDVAMSF HYISMGDCIG WLEDFLMGMD STLEPSAGAP LAMSSGHHHH HH
RAET1L (retinoic acid early transcript 1L), also called ULBP6 (cytomegalovirus glycoprotein UL16 binding protein 6), is an approximately 31 kDa glycophosphatidylinositol (GPI)-linked member of a family of human cell-surface proteins that serve as ligands for human NKG2D receptors . Its primary function involves activating cytolytic activity and cytokine production when engaged with effector cells expressing NKG2D, such as natural killer (NK) cells and CD8+ T cells . RAET1L plays a critical role in immune surveillance, particularly in the context of stress responses, viral infections, and cancer development. Structurally, human RAET1L mRNA encodes a 25 amino acid signal sequence, a 193 amino acid extracellular domain, and a 28 amino acid C-terminal propeptide that is removed during GPI linkage .
RAET1L shares significant amino acid sequence identity with other family members, particularly in the extracellular regions. It has 97% sequence identity with ULBP-2/RAET1H and 92% identity with ULBP-5/RAET1G, while sharing 35-60% amino acid identity with remaining family members . While most family proteins including RAET1L are GPI-anchored membrane proteins, ULBP-5/RAET1G and ULBP4/RAET1E express transmembrane forms, indicating structural diversity within the family . This family is distinct from rodent NKG2D ligands like Rae-1 α-ε, which are distantly related to MHC class I proteins, though the human ULBP and rodent Rae-1 family proteins do not share significant sequence identity . Understanding these relationships helps researchers contextualize RAET1L within the broader immune recognition system.
RAET1L mRNA shows a restricted expression pattern in normal human tissues, with notable expression in the trachea . In pathological contexts, RAET1L expression has been observed in certain HPV-positive cervical carcinoma and colorectal carcinoma cell lines . Importantly, RAET1L has a more restricted expression profile in cell lines and primary human tissues compared to other NKG2D ligands, suggesting tissue-specific regulation and function . RAET1L expression is also induced upon cytomegalovirus (CMV) infection in primary human fibroblasts, though the virus has evolved mechanisms to sequester RAET1L from the cell surface through the viral protein UL16 . This restricted expression pattern may indicate specialized roles for RAET1L in specific immunological contexts.
For comprehensive analysis of RAET1L expression, researchers should employ a multi-modal approach:
Transcriptional Analysis: RT-qPCR targeting RAET1L-specific sequences is the gold standard for quantifying mRNA expression levels. This should be complemented with RNA-seq for genome-wide expression analysis and identification of splice variants.
Protein Detection: Western blotting with validated anti-RAET1L antibodies can determine protein levels, while flow cytometry is essential for quantifying surface expression specifically, given RAET1L's membrane localization .
Subcellular Localization: Immunofluorescence microscopy with co-localization markers can determine whether RAET1L is properly expressed at the cell surface or retained intracellularly, particularly important when studying viral immune evasion mechanisms .
Gene Editing Approaches: CRISPR/Cas9 knockout cell lines, such as the RAET1L CRISPR Knockout 293T Cell Line, provide excellent negative controls and can be verified using abm's Screen ItTM CRISPR Cas9 Cleavage Detection Kit and Sanger sequencing .
For stress-induced expression studies, researchers should systematically apply stressors (heat shock, oxidative stress, DNA damage) followed by time-course expression analysis to capture the dynamics of RAET1L upregulation.
Production of high-quality recombinant RAET1L requires careful consideration of expression systems and purification strategies:
Expression Systems Comparison:
System | Advantages | Limitations | Yield | Glycosylation |
---|---|---|---|---|
E. coli | Cost-effective, rapid | Limited post-translational modifications | High | None |
Sf9 Baculovirus | Proper folding, some glycosylation | More complex than bacterial systems | Medium-high | Partial |
Mammalian cells | Native-like glycosylation patterns | Higher cost, slower production | Lower | Complete |
For optimal functional studies of RAET1L, insect cell expression (Sf9 Baculovirus) represents an excellent compromise, producing properly folded, glycosylated RAET1L with reasonable yields . The recombinant RAET1L should contain the extracellular domain (amino acids 26-218) fused to a C-terminal 6-His tag for purification purposes . After expression, purification via proprietary chromatographic techniques yields a single, glycosylated polypeptide chain with a molecular mass of approximately 22.9kDa .
For storage stability, the purified protein should be maintained in a solution containing 10% glycerol and Phosphate-Buffered Saline (pH 7.4) at 4°C if used within 2-4 weeks, or frozen at -20°C for longer periods . Addition of carrier proteins (0.1% HSA or BSA) is recommended for long-term storage, and multiple freeze-thaw cycles should be avoided .
To comprehensively investigate RAET1L's immunological functions, researchers should implement the following key assays:
NKG2D Binding Assays: ELISA-based binding assays using purified RAET1L and NKG2D can quantify binding affinity and kinetics . Surface plasmon resonance (SPR) provides real-time binding kinetics and affinity measurements between RAET1L and its binding partners.
NK Cell Activation Assays: Co-culture systems using RAET1L-expressing cells and NK cells, measuring NK cell degranulation (CD107a expression), cytokine production (IFN-γ, TNF-α), and target cell killing through chromium release or flow cytometry-based methods.
UL16 Evasion Studies: Dual-expression systems of RAET1L and HCMV UL16 to investigate intracellular retention mechanisms, using flow cytometry to quantify surface vs. intracellular protein localization .
T Cell Activation Assays: Similar to NK cell assays but using CD8+ T cells as effectors, measuring proliferation, cytokine production, and cytotoxicity against RAET1L-expressing targets.
In vivo Models: Humanized mouse models expressing human RAET1L and NKG2D can provide insights into systemic immune responses in cancer and infection contexts.
These assays should be performed with appropriate controls, including RAET1L knockout cells and blocking antibodies against NKG2D, to establish specificity of the observed effects.
Two common alleles of RAET1L were characterized, with the RAET1L02 allele showing profound clinical benefits. Patients possessing the RAET1L02 allele demonstrated significantly improved long-term outcomes:
These findings suggest that RAET1L genotyping could serve as a valuable prognostic marker for transplant outcome prediction . Mechanistically, these polymorphisms likely modulate the strength of NKG2D-mediated immune responses against residual malignant cells or against host tissues in graft-versus-host disease contexts.
For researchers investigating these polymorphisms, next-generation sequencing approaches focusing on the five key SNPs identified within the RAET1L gene should be prioritized. Furthermore, functional studies comparing the binding affinity of different RAET1L allelic variants to NKG2D and their effects on NK and T cell activation would provide mechanistic insights into the observed clinical differences.
HCMV has evolved sophisticated mechanisms to evade RAET1L-mediated immune recognition, primarily through the viral glycoprotein UL16. While HCMV infection induces RAET1L expression in primary human fibroblasts, the virus simultaneously counteracts this immune activation signal .
The primary evasion mechanism involves UL16 binding to RAET1L with high affinity, physically retaining it inside the cell and preventing its surface expression . This intracellular sequestration prevents recognition by NKG2D-expressing immune cells like NK and CD8+ T cells. RAET1L shows stronger binding to both NKG2D and UL16 compared to other family members like ULBP-5/RAET1G, suggesting it may be a primary target for HCMV immune evasion .
To effectively study these evasion mechanisms, researchers should:
Utilize fluorescently tagged RAET1L and UL16 proteins in live-cell imaging to track intracellular trafficking
Implement proximity ligation assays to confirm direct protein-protein interactions within cellular compartments
Develop UL16 mutants to map the critical binding interfaces with RAET1L
Compare immune activation in the presence of wild-type versus UL16-deficient HCMV strains
These approaches would provide insights into not only HCMV pathogenesis but also potential therapeutic strategies to restore immune surveillance in HCMV-infected cells.
While most RAET1 family members, including RAET1L/ULBP6, are GPI-anchored membrane proteins, ULBP-5/RAET1G and ULBP4/RAET1E express transmembrane forms . This structural difference has significant implications for their signaling capabilities and functional properties:
Membrane Localization: Transmembrane RAET1 proteins may localize to different membrane microdomains compared to GPI-anchored forms, potentially affecting their accessibility to NKG2D-expressing cells and interactions with other membrane proteins.
Shedding Dynamics: GPI-anchored proteins like RAET1L can be released from the cell surface through phospholipase action or proteolytic cleavage, creating soluble forms that may act as decoy receptors. In contrast, transmembrane forms require specific proteases for shedding, potentially resulting in different release kinetics and regulation .
Signal Transduction: Transmembrane RAET1 proteins may participate in bidirectional signaling, not only activating NKG2D+ immune cells but potentially transmitting reverse signals into the RAET1-expressing cell, a capability likely absent in GPI-anchored forms .
Protein Stability: The different membrane anchoring methods may affect protein half-life at the cell surface and susceptibility to internalization and degradation.
To investigate these differences, researchers should implement comparative studies using RAET1L (GPI-anchored) and RAET1G (transmembrane) in identical cellular contexts, examining their:
Lateral mobility using fluorescence recovery after photobleaching (FRAP)
Membrane microdomain localization through detergent resistance and co-localization studies
Differential responses to phospholipases versus proteases
Capacity to induce signaling events within their host cells
Such studies would provide valuable insights into the evolutionary significance of these distinct anchoring mechanisms within the RAET1 family.
CRISPR-Cas9 technology offers revolutionary approaches for investigating RAET1L biology across multiple dimensions:
Precise Genome Editing: CRISPR knockout cell lines, such as the RAET1L CRISPR Knockout 293T Cell Line, provide valuable negative controls for antibody validation and functional studies . Beyond simple knockouts, CRISPR-Cas9 can create precise point mutations to study how specific amino acid changes affect RAET1L function and interactions.
Regulatory Element Mapping: CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems can target the RAET1L promoter and enhancer regions to identify critical regulatory elements controlling its stress-induced expression. This approach would help decipher the transcriptional regulation pathways activated during viral infection or cellular stress.
High-Throughput Screening: CRISPR screens targeting genes involved in RAET1L processing, trafficking, and degradation can identify novel regulators of RAET1L surface expression, potentially revealing new therapeutic targets to enhance immune surveillance.
In vivo Modeling: CRISPR-engineered mouse models expressing humanized RAET1L can provide systems for testing the in vivo relevance of findings from cell culture studies, particularly regarding immune responses to cancer and infections.
Allelic Replacement: CRISPR-mediated homology-directed repair can replace one RAET1L allele with another (e.g., RAET1L01 with RAET1L02) to directly test the functional consequences of polymorphisms identified in transplantation studies .
These approaches should be combined with rigorous validation using complementary techniques such as RNA-seq, proteomics, and functional immunological assays to fully leverage CRISPR's potential for advancing RAET1L research.
RAET1L's role in activating anti-tumor immune responses through NKG2D makes it a promising target for cancer immunotherapy development:
Therapeutic Upregulation: Pharmacological approaches to selectively induce RAET1L expression on tumor cells could enhance their visibility to the immune system. Potential strategies include epigenetic modifiers, stress response pathway activators, or targeted gene delivery systems.
Chimeric Antigen Receptor (CAR) Development: The extracellular domain of NKG2D could be incorporated into CARs to target multiple NKG2D ligands including RAET1L on tumor cells. These CAR-T or CAR-NK cells would recognize stress-induced ligands commonly upregulated on malignant cells.
Bispecific Antibody Engineering: Bispecific antibodies linking T cells to RAET1L-expressing tumor cells represent another promising approach. These could be designed to overcome potential immune evasion mechanisms like RAET1L shedding or downregulation.
Checkpoint Inhibition Combination: RAET1L-targeting therapies might synergize with established checkpoint inhibitors like anti-PD-1/PD-L1, potentially converting "cold" tumors to "hot" immunogenic ones by simultaneously releasing brakes and applying accelerators to immune responses.
Personalized Approaches: Given the known polymorphisms in RAET1L and their impact on immune responses , genotyping patients could help predict responsiveness to NKG2D-based immunotherapies and guide personalized treatment strategies.
For research in this area, developing preclinical models that accurately recapitulate human RAET1L expression patterns and regulation will be essential for translational success.
While HCMV's interaction with RAET1L is well-documented , the relationship between RAET1L and other viral infections represents an important frontier for research:
Broad Viral Survey: Systematic studies should examine RAET1L expression patterns during infection with diverse virus families (herpesviruses, papillomaviruses, influenza, hepatitis viruses, coronaviruses). Initial evidence suggesting RAET1L expression in HPV-positive cervical carcinoma cell lines warrants deeper investigation .
Immune Evasion Mechanisms: Beyond HCMV's UL16, other viruses likely employ distinct mechanisms to counteract RAET1L-mediated immune activation. Comparative viral proteomics approaches could identify novel viral immunomodulatory proteins targeting RAET1L.
Genetic Association Studies: Analysis of RAET1L polymorphisms in relation to viral disease susceptibility, severity, and outcomes could reveal important clinical correlations, similar to findings in transplantation contexts .
Tissue-Specific Responses: Given RAET1L's restricted expression pattern , investigation of tissue-specific RAET1L responses to viral infection in organs like the respiratory tract (where RAET1L shows natural expression) could reveal specialized roles.
Therapeutic Implications: Understanding virus-specific regulation of RAET1L could inform the development of broad-spectrum antiviral approaches that enhance natural immune surveillance mechanisms rather than targeting specific viral proteins that can rapidly evolve.
Research methodologies should include ex vivo infection of primary human tissues, humanized mouse models, and retrospective analysis of patient samples from various viral infections to comprehensively address these questions.
Despite significant advances in understanding RAET1L biology, several critical questions remain:
Transcriptional Regulation: What specific transcription factors and epigenetic mechanisms control the tissue-restricted expression of RAET1L, and how are these altered during stress, malignancy, and infection?
Evolutionary Significance: Why has the human immune system evolved multiple NKG2D ligands with different expression patterns and structures (GPI-anchored vs. transmembrane), and what unique role does RAET1L play within this diverse repertoire?
Signaling Complexity: How does RAET1L binding to NKG2D integrate with other immune receptor signaling pathways to fine-tune immune responses in different physiological and pathological contexts?
Clinical Translation: How can the growing understanding of RAET1L polymorphisms and expression patterns be leveraged for improved clinical outcomes in transplantation, cancer immunotherapy, and viral infection management?
Beyond Immune Surveillance: Does RAET1L have functions beyond immune recognition, potentially in tissue homeostasis, development, or cellular stress responses?
Addressing these questions will require interdisciplinary approaches combining structural biology, advanced imaging, systems immunology, and clinical studies. The answers will significantly advance our understanding of immune surveillance mechanisms and inform new therapeutic strategies.
Emerging technologies will transform RAET1L research in several key areas:
Single-Cell Multi-omics: Integration of single-cell transcriptomics, proteomics, and epigenomics will reveal cell-specific RAET1L expression patterns and regulatory mechanisms at unprecedented resolution, especially in heterogeneous tissues and tumor microenvironments.
Spatial Transcriptomics/Proteomics: These technologies will map RAET1L expression in intact tissues while preserving spatial relationships with immune cells and other tissue components, providing crucial contextual information about RAET1L function.
Cryo-Electron Microscopy: Advanced structural studies will elucidate the atomic-level details of RAET1L interactions with NKG2D and viral proteins like UL16, informing structure-based drug design.
Organoid Models: Patient-derived organoids will enable personalized studies of RAET1L expression and function in physiologically relevant 3D tissue architectures.
AI/Machine Learning: Computational approaches will facilitate integration of multi-dimensional RAET1L data with broader immune parameters, potentially revealing previously unrecognized patterns and correlations.
Base Editing and Prime Editing: These refined CRISPR technologies will enable precise modification of RAET1L regulatory elements and coding sequences without double-strand breaks, advancing functional genomics studies.
In situ Protein-Protein Interaction Detection: Technologies like APEX proximity labeling will map the RAET1L interactome in living cells, potentially uncovering novel binding partners beyond NKG2D and UL16.
These technological advances will collectively drive the field toward a more comprehensive understanding of RAET1L biology and its therapeutic implications.
The RAET1L gene encodes glycoproteins that contain extracellular alpha-1 and alpha-2 domains but lack the membrane-proximal Ig-like alpha-3 domain . Most RAET1 glycoproteins are anchored to the membrane via glycosylphosphatidylinositol (GPI) linkage . The RAET1L protein binds and activates the KLRK1/NKG2D receptor, mediating natural killer cell cytotoxicity .
RAET1L plays a significant role in the immune system. It is involved in the activation of natural killer (NK) cells and certain T-cells, which are crucial for the body’s innate immune response . The protein’s ability to bind to the KLRK1/NKG2D receptor makes it an essential player in the immune surveillance against tumors and virally infected cells .