TNFRSF14 functions as a bidirectional receptor-ligand, mediating immune activation and inhibition:
Pro-inflammatory signaling: Binds TNFSF14/LIGHT to activate NF-κB and AP-1, promoting T-cell proliferation and cytokine production (e.g., TNF-α, IL-8) .
Inhibitory signaling: Engages immunoglobulin superfamily members BTLA and CD160 to suppress T-cell responses .
Viral entry: Serves as a receptor for herpes simplex virus (HSV) glycoprotein D, facilitating viral entry into cells .
Matrix metalloproteinase (MMP) induction: Enhances MMP-1, -9, and -13 expression in macrophages, contributing to plaque destabilization in atherosclerosis .
T-cell modulation: TNFRSF14 Human, His enables studies of costimulatory/inhibitory signals in CD4+/CD8+ T cells .
NK cell activation: Enhances IFN-γ production via CD160 interaction, boosting antitumor responses .
Cell viability: TNFRSF14 overexpression reduced T24 bladder cancer cell proliferation by 40% at 72 hours (P < 0.05) .
Flow cytometry: Anti-TNFRSF14 antibodies (e.g., MAB3563) confirm protein expression in monocytes, B cells, and NK cells .
TNFRSF14, also known as Herpesvirus Entry Mediator (HVEM), is a human cell surface receptor belonging to the TNF-receptor superfamily. It serves as a receptor for two TNF superfamily ligands: lymphotoxin α (TNF-β) and LIGHT (TNFSF14), which initiate positive signaling pathways . The protein consists of an extracellular domain (amino acids Leu39-Val202 in the mature protein) that mediates ligand binding, a transmembrane domain, and a cytoplasmic region that facilitates downstream signaling . When expressed as a recombinant protein with a histidine tag, the theoretical molecular weight is approximately 18.5 kDa, though the observed weight in human tissues is around 26.5 kDa due to post-translational modifications .
TNFRSF14 plays a complex role in immune regulation by delivering negative signals to T cells through interaction with the B and T Lymphocyte Attenuator (BTLA) molecule . This interaction is critical for maintaining immune homeostasis and preventing excessive T-cell activation. Simultaneously, TNFRSF14 can mediate positive signaling through interactions with LIGHT and lymphotoxin α. The receptor is expressed at high levels on activated monocytes, macrophages, and THP-1 cells (a human macrophage-like cell line), suggesting a significant role in myeloid cell function . Research has shown that TNFRSF14 activation in monocytes/macrophages can induce the expression of proatherogenic cytokines and matrix metalloproteinases, highlighting its importance in inflammatory processes .
For quantitative analysis of TNFRSF14 expression in tissue samples, researchers should employ:
Flow cytometry: Using fluorescently-labeled antibodies against TNFRSF14, expression can be quantified at the single-cell level. This approach has successfully demonstrated that approximately 50% of follicular lymphoma B cells express HVEM at levels above isotype control .
Immunohistochemistry: This method is particularly useful for analyzing TNFRSF14 expression patterns within tissue architecture. Studies have utilized this technique to reveal high expression levels in the shoulder regions of atherosclerotic plaques, particularly in areas rich in foam cells and HLA-DR–positive cells .
Western blot analysis: This approach has been effective for comparing TNFRSF14 expression between different regions of atherosclerotic plaques, revealing higher expression in atheromatous regions compared to fibrous regions .
Each method provides distinct advantages, and selection should be based on experimental requirements for spatial resolution versus quantitative precision.
TNFRSF14 genetic aberrations, found in approximately 40% of follicular lymphoma (FL) patients, significantly impact immune function in several ways:
Expression levels: FL B cells with dual TNFRSF14 aberrations show virtually undetectable HVEM expression compared to wild-type cells where ~50% express HVEM at levels above isotype control. Single aberrations result in intermediate expression levels .
Allogeneic T-cell stimulation: FL B cells with dual TNFRSF14 aberrations demonstrate enhanced capacity to stimulate allogeneic T-cell responses. Specifically:
Clinical implications: These aberrations associate with higher incidence of acute graft-versus-host disease (GVHD) in patients undergoing allogeneic hematopoietic stem cell transplantation, suggesting practical applications for stratified transplantation approaches .
This represents the first demonstration of how an acquired genetic lesion impacts tumor cells' ability to stimulate allogeneic T-cell immune responses, with significant implications for adoptive immunotherapy strategies.
Researchers evaluating TNFRSF14 as an immune checkpoint target should incorporate these methodological considerations:
Genetic modification models: Use CRISPR/Cas9 genetic editing to establish TNFRSF14-deficient tumor cell lines for comparison with TNFRSF14-expressing lines in appropriate animal models (e.g., PBMC-humanized NSG mice) .
Antibody characterization: When developing blocking antibodies, thoroughly assess:
Controls: Include tumor lines deficient for TNFRSF14 when testing therapeutic antibodies to confirm that effects are dependent on target expression .
Microenvironment factors: Consider that the tumor microenvironment may influence TNFRSF14 signaling outcomes, as studies have shown different effects in humanized versus non-humanized mouse models .
In vitro validation: Complement in vivo studies with in vitro analyses of antibody effects on tumor survival, with and without relevant immune cell populations .
To ensure high-quality recombinant TNFRSF14-His protein for research applications, implement this comprehensive quality control pipeline:
Quality Control Test | Methodology | Acceptance Criteria | Research Application |
---|---|---|---|
Purity assessment | Tris-Bis PAGE | >95% purity under reduced conditions | Ensures experimental results aren't affected by contaminants |
Secondary verification | SEC-HPLC | >95% purity | Confirms homogeneity of protein preparation |
Functional validation | ELISA binding assay | Verified binding to known ligands (e.g., CD160) with specific EC50 values | Confirms biological activity of purified protein |
Contaminant testing | LAL method | <1 EU per 1 μg of protein | Prevents endotoxin interference in immunological studies |
These quality control measures, as implemented for commercial TNFRSF14-His preparations, ensure that experimental results accurately reflect the biological properties of the protein rather than artifacts from the production process .
TNFRSF14 contributes to atherosclerosis through several coordinated mechanisms:
Expression pattern: Immunohistochemical analysis reveals high TNFRSF14 expression in atherosclerotic plaque regions rich in macrophages/foam cells, particularly in shoulder regions. Expression is specifically observed in CD68-positive foam cells and is absent in normal human aorta .
Cytokine induction: Activation of TNFRSF14 in THP-1 cells (macrophage-like cell line), particularly in combination with IFN-γ, induces synergistic production of proatherogenic cytokines including TNF-α and interleukin-8 .
Matrix remodeling: TNFRSF14 activation induces expression of matrix metalloproteinases (MMP-1, MMP-9, MMP-13) and tissue inhibitors of metalloproteinase-1 and -2, potentially decreasing plaque stability by promoting extracellular matrix degradation .
Ligand interaction: TNFSF14 (LIGHT), the ligand for TNFRSF14, is expressed at higher levels in inflammatory regions of atherosclerotic plaques. This interaction likely stimulates TNFRSF14-mediated inflammatory reactions in foam cells .
Cellular activation: Upregulation of TNFRSF14 occurs concomitantly with CD14 (a monocyte activation marker) in activated peripheral blood monocytes, suggesting its regulation is linked to monocyte activation states .
These findings collectively indicate that TNFRSF14 mediates atherosclerosis by orchestrating proatherogenic cytokine production and matrix degradation in activated monocytes/macrophages within plaque microenvironments.
TNFRSF14 functions as a targetable immune checkpoint through these mechanisms:
Negative T-cell signaling: TNFRSF14 delivers negative signals to T cells through interaction with BTLA, creating an immunosuppressive environment that benefits tumor growth .
Prognostic association: TNFRSF14 expression has been associated with worse prognosis in various malignancies, supporting its role in tumor immune evasion .
Experimental validation: Genetic deletion of TNFRSF14 or blocking TNFRSF14/BTLA interaction with monoclonal antibodies profoundly impacts tumor growth in mice reconstituted with human T cells, demonstrating its potential as a therapeutic target .
Mechanism of action: Blocking TNFRSF14 increases proliferation and number of tumor-infiltrating lymphocytes, enhancing anti-tumor immune responses .
Expression requirement: Therapeutic effects of anti-TNFRSF14 antibodies are lost with TNFRSF14-deficient tumors, confirming that expression by tumor cells is necessary for checkpoint blockade efficacy .
Macrophage involvement: Surprisingly, anti-TNFRSF14 antibodies also show effects in non-humanized mouse models, suggesting additional mechanisms potentially involving myeloid cells .
These findings establish TNFRSF14 as a novel immune checkpoint with therapeutic potential through both T cell-dependent and potentially myeloid cell-dependent mechanisms.
For optimal production of functional TNFRSF14-His protein:
Expression system: Use human HEK293 cells for expression, as this mammalian system provides appropriate post-translational modifications essential for proper folding and function of human TNFRSF14 .
Construct design:
Purification strategy: Implement a multi-step purification protocol that achieves >95% purity as determined by both Tris-Bis PAGE and HPLC analysis .
Quality assessment: Validate functionality through binding assays with known ligands (e.g., CD160) to ensure that the purified protein maintains its biological activity .
Storage formulation: Lyophilize the purified protein in appropriate buffer conditions that maintain stability and activity upon reconstitution .
This approach has successfully produced recombinant TNFRSF14-His with a theoretical molecular weight of 18.5 kDa and high biological activity as measured by ligand binding assays .
To effectively identify and characterize TNFRSF14 aberrations in patient samples:
Next-generation sequencing: Sequence the TNFRSF14 gene to identify mutations, particularly those resulting in altered protein expression or function. This approach has successfully identified aberrations in 40% of follicular lymphoma patients .
Flow cytometry: Quantitatively assess HVEM protein expression on cell surfaces using fluorescently-labeled antibodies. This approach effectively distinguishes between wild-type, single aberration, and dual aberration expression patterns .
Functional validation: Implement in vitro alloreactivity assays to assess the functional impact of identified aberrations. Compare the capacity of wild-type and aberrant TNFRSF14-expressing cells to stimulate allogeneic T-cell responses .
Phenotypic characterization: Analyze additional cell surface molecules associated with antigen-presenting capacity (MHC class I, MHC class II, CD80, CD86, CD58) to determine if aberrations specifically affect TNFRSF14 or have broader impacts on cell phenotype .
Clinical correlation: Associate identified aberrations with clinical outcomes, such as incidence of acute GVHD in transplantation settings, to establish clinical relevance .
This multi-modal approach provides comprehensive characterization of both the genetic basis and functional consequences of TNFRSF14 aberrations.
To effectively model TNFRSF14-mediated T-cell regulation in vitro:
Allostimulation assays: Use a mixed lymphocyte reaction system where TNFRSF14-expressing or TNFRSF14-deficient cells stimulate allogeneic T cells. This approach has successfully demonstrated that:
T-cell activation monitoring:
Influence of inflammatory environment: Model the proinflammatory post-transplant environment through in vitro activation conditions and assess how this affects TNFRSF14 expression and function .
Antibody blocking studies: Incorporate blocking antibodies against TNFRSF14 or its ligands to dissect specific interaction effects on T-cell activation .
This system effectively recapitulates the impact of TNFRSF14 on T-cell regulation and provides a platform for testing therapeutic interventions targeting this pathway.
TNFRSF14-targeted therapies could substantially impact transplantation outcomes through several mechanisms:
Stratified immunosuppression: Patients with TNFRSF14 aberrations experience higher incidence of acute GVHD, suggesting they could benefit from more aggressive immunosuppression regimens to reduce harmful GVHD after transplantation .
Enhanced donor-recipient matching: Incorporating TNFRSF14 status into matching algorithms could improve transplant compatibility beyond traditional HLA matching, potentially reducing GVHD risk .
Targeted blockade approach: Developing TNFRSF14 blocking agents could specifically inhibit excessive alloreactive T-cell responses while preserving beneficial immune functions. This approach is supported by in vitro data showing that FL B cells with TNFRSF14 aberrations stimulate greater frequencies of alloreactive effector T cells .
Preservation of anti-tumor effects: Careful modulation of TNFRSF14 signaling could potentially maintain beneficial graft-versus-tumor effects while reducing harmful GVHD, as TNFRSF14 influences both processes .
Personalized therapy: TNFRSF14 genetic testing could allow for personalized immunosuppression strategies based on predicted GVHD risk, optimizing the balance between GVHD prevention and maintenance of beneficial immune functions .
These approaches represent promising avenues for improving transplantation outcomes through targeted manipulation of TNFRSF14 signaling pathways.
Emerging research on TNFRSF14 in cardiovascular disease suggests these promising research directions:
Plaque vulnerability biomarkers: TNFRSF14 expression patterns overlap with MMP-1, -9, and -13 in atherosclerotic plaques, suggesting potential as a biomarker for plaque instability and rupture risk .
Targeted anti-inflammatory therapies: Development of agents that specifically inhibit TNFRSF14-induced production of proatherogenic cytokines (TNF-α, IL-8) in macrophages could provide more selective anti-inflammatory approaches than current therapies .
Matrix remodeling pathway intervention: TNFRSF14 activation induces both matrix metalloproteinases and their inhibitors, suggesting complex regulation of matrix remodeling. Understanding and modulating this balance could stabilize vulnerable plaques .
TNFSF14-TNFRSF14 axis targeting: The observed higher expression of TNFSF14 (LIGHT) in inflammatory regions of plaques suggests that disrupting this ligand-receptor interaction could reduce inflammatory processes driving atherosclerosis .
Monocyte activation modulation: The concomitant upregulation of TNFRSF14 and CD14 during monocyte activation indicates that targeted intervention in this pathway might influence early stages of foam cell formation .
These research directions hold promise for developing novel diagnostic and therapeutic approaches for atherosclerosis by targeting the TNFRSF14 signaling network.
Despite promising results, several challenges must be addressed for successful clinical translation of TNFRSF14 research:
Dual signaling complexity: TNFRSF14 can mediate both positive and negative signaling depending on which ligand it engages. Therapeutic interventions must carefully consider this dual nature to avoid unintended consequences .
Context-dependent effects: Research indicates that TNFRSF14 blockade has different effects in different models (humanized versus non-humanized), suggesting complex interactions with the microenvironment that must be understood for successful translation .
Expression heterogeneity: Variations in TNFRSF14 expression between patients and even within different regions of the same tissue (as seen in atherosclerotic plaques) complicate standardization of therapeutic approaches .
Target specificity: Anti-TNFRSF14 antibodies must be highly specific to avoid off-target effects on related TNF-receptor family members, requiring extensive validation before clinical application .
Biomarker development: Establishing reliable biomarkers to identify patients most likely to benefit from TNFRSF14-targeted therapies remains challenging but essential for personalized medicine approaches .
Addressing these challenges requires rigorous preclinical validation using diverse experimental models and careful correlation with human pathophysiology.
The recombinant human HVEM protein is typically expressed in HEK293 cells and includes a polyhistidine (His) tag at the C-terminus for purification purposes . The extracellular domain (ECD) of HVEM, which spans from Met1 to Val202, is the primary region used in recombinant protein production . The recombinant protein has a predicted molecular weight of approximately 19 kDa, but due to glycosylation, it appears as 33-38 kDa on SDS-PAGE under reducing conditions .