TNFRSF14 orchestrates bidirectional immune signaling through interactions with ligands from distinct superfamilies:
LIGHT (TNFSF14) Binding: Triggers costimulatory signals in T cells, enhancing proliferation, cytokine production (e.g., IFN-γ), and antitumor immunity .
Lymphotoxin-α (LTα) Binding: Promotes inflammatory responses and lymphoid tissue organization .
BTLA/CD160 Binding: Recruits SHP1/2 phosphatases via immunoreceptor tyrosine-based inhibitory motifs (ITIMs), dampening T cell receptor (TCR) and CD28 signaling .
Serves as a receptor for herpes simplex virus (HSV) glycoprotein D (gD), facilitating viral entry into T cells and dendritic cells .
TNFRSF14 dysregulation is implicated in multiple pathologies:
Emerging strategies focus on modulating TNFRSF14 pathways:
Dual Signaling Mechanisms: Clarify how TNFRSF14 balances stimulatory (LIGHT) and inhibitory (BTLA) signals in tumor microenvironments .
Mutation-Specific Therapies: Develop agents targeting TNFRSF14 mutations in lymphoma .
Biomarker Validation: Confirm TNFRSF14’s prognostic utility in bladder cancer using multi-center cohorts .
TNFRSF14 (Tumor Necrosis Factor Receptor Superfamily Member 14), also known as HVEM (Herpes Virus Entry Mediator), is a cell surface receptor that plays critical roles in immune regulation. This receptor is expressed on various immune cells including mast cells and B cells. TNFRSF14 functions primarily through interactions with its ligand TNFSF14 (LIGHT) and can modulate immune responses by influencing T-cell activation. The receptor belongs to the TNF receptor superfamily and has pleiotropic functions that can either foster or inhibit immune responses depending on the cellular context. TNFRSF14 is particularly important in regulating T-cell-mediated immunity, with significant implications for both normal immune function and pathological conditions. TNFRSF14:TNFSF14 interactions have been shown to support the generation and longevity of Th2 cells and promote Th2 memory through Akt activation .
TNFRSF14 expression has been documented on multiple human immune cell populations. Strong expression has been detected on human mast cells, including those from the LAD2 human mast cell line and in vitro-derived human peripheral blood cultured mast cells (huPBCMCs) from CD34+ mononuclear precursors . TNFRSF14 is also expressed on B cells, particularly in follicular lymphoma B cells, where its expression levels impact antigen-presenting capacity . The receptor is additionally found on various T cell subsets, where it participates in regulation of T cell activation and immune responses. Unlike some other receptors like TNFRSF3, which was not detected on human mast cells in studies, TNFRSF14 appears to have a broader expression pattern across immune cell lineages, suggesting its diverse roles in human immune regulation .
TNFRSF14 signaling exhibits distinctive characteristics compared to other TNF receptor family members. Unlike some TNF receptors that function independently, TNFRSF14 engagement alone typically does not activate cellular responses but rather acts in concert with other activation signals. For instance, in mast cells, TNFRSF14 engagement by TNFSF14 enhances IgE-dependent activation but does not induce mediator production as a single signal . This cooperative signaling mechanism distinguishes TNFRSF14 from other family members that can directly initiate cellular responses.
Additionally, TNFRSF14 has unusual versatility in its ligand interactions. In humans, TNFSF14 can interact with three receptors: TNFRSF14 (HVEM), TNFRSF3 (lymphotoxin-beta receptor), and TNFRSF6B (soluble decoy receptor 3), while in mice it interacts with TNFRSF14 and TNFRSF3 . This multi-receptor interaction capability creates complexity in understanding TNFRSF14 signaling networks compared to more straightforward receptor-ligand pairs in the TNF family.
For accurate TNFRSF14 detection in human tissue samples, researchers should employ complementary techniques to confirm expression patterns. Flow cytometry represents the gold standard for quantifying surface TNFRSF14 expression on specific cell populations, allowing for comparison between different cell types and disease states. This approach successfully distinguished TNFRSF14 expression levels between wild-type follicular lymphoma B cells and those with genetic aberrations .
Immunohistochemistry provides spatial context for TNFRSF14 expression within tissue architecture, though proper validation with specific antibodies is crucial. For genetic analysis, RT-PCR can detect TNFRSF14 mRNA expression, while next-generation sequencing allows comprehensive identification of TNFRSF14 aberrations and mutations. In research presented in the literature, fluorescently labeled TNFSF14 (TNFSF14-A594) has been used to visualize TNFRSF14 receptor clustering and engagement on cell surfaces .
For functional validation of protein expression, receptor-ligand binding assays using AlexaFluor-conjugated TNFSF14 can confirm the presence of functional TNFRSF14 receptors, as demonstrated in studies visualizing receptor clustering .
To effectively model TNFRSF14 signaling dynamics in vitro, researchers should establish systems that recapitulate the receptor's co-stimulatory function. Cell culture models using human mast cell lines (such as LAD2) or primary cells (like peripheral blood-derived cultured mast cells) provide physiologically relevant systems for studying TNFRSF14 signaling . For B cell studies, primary follicular lymphoma B cells with characterized TNFRSF14 mutation status can be isolated and maintained for functional assays .
Signaling dynamics are best captured through time-course experiments measuring downstream effectors following TNFRSF14 engagement. This approach should include analysis of both immediate responses (minutes to hours) and delayed effects (hours to days). Co-stimulation protocols are essential, as TNFRSF14 signaling often requires concurrent activation of other pathways. For example, in mast cells, TNFRSF14 engagement enhances FcεRI-mediated activation but has minimal effects alone .
Fluorescent labeling of both TNFRSF14 and its signaling partners provides visual confirmation of receptor clustering and co-localization. Researchers have successfully employed AlexaFluor-conjugated TNFSF14 (TNFSF14-A594) simultaneously with labeled anti-IgE antibodies to visualize receptor interactions during signaling events . Genetic manipulation through CRISPR-Cas9 or siRNA knockdown can further validate specificity of observed signaling effects.
Distinguishing direct TNFRSF14 effects from indirect pathway activation presents several methodological challenges in research. The pleiotropic nature of TNFRSF14 signaling, which can either promote or inhibit immune responses depending on context, complicates interpretation of experimental results . One significant challenge is that TNFRSF14 effects often manifest through co-stimulation with other receptors rather than as direct single-signal effects. For example, in mast cells, TNFRSF14 engagement alone produces minimal activation, while it significantly enhances FcεRI-mediated responses, making it difficult to isolate TNFRSF14-specific contributions .
Another challenge is the complexity of TNFSF14 ligand interactions, as TNFSF14 can bind multiple receptors in humans (TNFRSF14, TNFRSF3, and TNFRSF6B) and mice (TNFRSF14 and TNFRSF3) . This creates potential for indirect effects through alternative receptor engagement. Research designs must account for these complexities through careful controls and genetic approaches.
Researchers should employ genetic knockout models (e.g., TNFRSF14-deficient cells) compared with wild-type counterparts to identify receptor-specific effects. Receptor-specific blocking antibodies provide another approach to isolate direct TNFRSF14 contributions, while biochemical analysis of signaling intermediates can help map direct signaling cascades versus secondary activation events.
TNFRSF14 aberrations occur in approximately 40% of follicular lymphoma (FL) patients and significantly impact disease biology through modulation of immune interactions . These genetic lesions, which include homozygous deletions and nonsense mutations, result in markedly reduced or absent HVEM (TNFRSF14) expression on lymphoma B cells . The functional consequence is profound, as HVEM normally limits T-cell activation through interaction with the coinhibitory receptor BTLA (B- and T-lymphocyte attenuator) .
In FL B cells with TNFRSF14 aberrations, the loss of this inhibitory interaction creates a more immunostimulatory phenotype. Flow cytometry analysis has confirmed that while wild-type FL B cells express HVEM on approximately 50% of cells, expression is virtually undetectable in cells with dual TNFRSF14 aberrations . Importantly, other molecules associated with antigen presentation (MHC class I, MHC class II, CD80, CD86, and CD58) remain normally expressed, indicating a specific effect on HVEM expression rather than global changes in immune regulatory molecule expression .
Functional studies demonstrate that FL B cells with TNFRSF14 aberrations stimulate greater frequencies of alloreactive effector T cells . This enhanced immunostimulatory capacity likely contributes to lymphoma progression by creating a more favorable microenvironment for lymphoma cell survival and expansion, through altered T-cell interactions that may promote tumor growth rather than anti-tumor immunity.
TNFRSF14 plays a significant role in asthma pathology through its expression on mast cells and subsequent enhancement of IgE-mediated inflammatory responses. Research has revealed that TNFSF14:TNFRSF14 interactions potentiate IgE-dependent mast cell activation, amplifying allergic inflammatory cascades that contribute to asthma features . This mechanism has been demonstrated in both human and mouse mast cells, where TNFRSF14 engagement enhances FcεRI-mediated degranulation and production of inflammatory mediators including histamine, leukotrienes, and cytokines .
The pathological significance of this pathway has been confirmed in mouse models of asthma, where TNFRSF14 blockade with neutralizing antibodies or genetic deletion significantly diminished multiple features of asthma pathology, including airway hyperreactivity, inflammation, and remodeling . Importantly, therapeutic intervention targeting TNFRSF14 showed efficacy even when administered after initial antigen sensitization, suggesting potential relevance for treating established disease .
The translational significance is supported by clinical data showing a positive correlation between elevated TNFSF14 levels in the sputum of asthma patients and impaired lung function . These findings collectively suggest that targeting the TNFRSF14 pathway may represent a promising therapeutic approach for asthma, particularly through inhibiting mast cell-dependent inflammatory mechanisms. Future therapeutic strategies might include monoclonal antibodies against TNFRSF14 or small molecule inhibitors of downstream signaling pathways, which could potentially modulate allergic responses without broadly suppressing immune function.
TNFRSF14 expression and genetic status significantly impact outcomes in allogeneic hematopoietic stem cell transplantation (AHSCT), particularly in follicular lymphoma patients. Research has demonstrated that TNFRSF14 aberrations, which reduce HVEM expression, increase the alloantigen-presenting capacity of lymphoma B cells . This enhanced immunostimulatory phenotype has direct clinical consequences in the transplantation setting.
Patients with follicular lymphoma harboring TNFRSF14 aberrations show higher incidence of acute graft-versus-host disease (GVHD) following AHSCT compared to patients with wild-type TNFRSF14 . This connection between a specific genetic lesion and clinical alloreactivity provides a potential biomarker for risk stratification in transplantation. The mechanism involves disruption of the normal inhibitory interaction between HVEM on antigen-presenting cells and BTLA on T cells, which typically limits T-cell activation and proliferation .
These findings have important clinical implications for transplantation approaches. Patients with TNFRSF14 aberrations may benefit from more aggressive immunosuppression strategies to reduce harmful GVHD after transplantation . Additionally, the TNFRSF14-BTLA axis could represent a therapeutic target for modulating alloreactivity in the transplant setting. This represents the first demonstration of how an acquired genetic lesion can influence tumor cells' capacity to stimulate allogeneic T-cell immune responses, with broader implications for adoptive immunotherapy strategies .
Single-cell analysis approaches provide crucial insights into TNFRSF14 signaling heterogeneity across immune cell populations. Single-cell RNA sequencing (scRNA-seq) can identify differential expression patterns of TNFRSF14 and associated signaling components across diverse immune cells, revealing cell type-specific signaling potentials not apparent in bulk analysis. This technique could identify previously unrecognized TNFRSF14-expressing cell populations beyond the well-documented mast cells and B cells .
Mass cytometry (CyTOF) offers comprehensive protein-level analysis of TNFRSF14 signaling networks by simultaneously measuring multiple phosphorylation events and surface markers at single-cell resolution. This approach could map how TNFRSF14 engagement differentially activates downstream pathways across immune cell subsets. For example, researchers could examine how TNFRSF14 stimulation affects signaling in naive versus memory T cells or in different B cell subpopulations.
Imaging mass cytometry extends this approach by preserving spatial information, enabling visualization of TNFRSF14 receptor clustering and co-localization with other signaling components within tissue microenvironments. This technique could reveal how TNFRSF14-expressing cells interact with other immune cells in lymphoid tissues or inflammatory sites.
Single-cell phospho-flow cytometry provides a direct measure of phosphorylation events downstream of TNFRSF14 activation, allowing researchers to quantify signaling kinetics at individual cell resolution. This approach could be particularly valuable for studying the co-stimulatory function of TNFRSF14 in different contexts, such as its enhancement of FcεRI-mediated mast cell activation .
Studying TNFRSF14-mediated co-stimulatory effects in complex immune interactions requires sophisticated experimental systems that preserve physiological complexity while allowing specific pathway interrogation. Multiparameter flow cytometry with simultaneous detection of multiple activation markers represents an essential approach for capturing the diverse outcomes of TNFRSF14 co-stimulation. This method should incorporate markers of immediate activation (calcium flux, phosphorylation events), intermediate responses (surface activation markers), and long-term effects (proliferation, cytokine production) .
3D co-culture systems that incorporate multiple relevant cell types provide more physiologically relevant contexts than traditional monocultures. For studying mast cell responses, researchers should consider co-cultures with T cells and other inflammatory cells to model asthmatic environments . Similarly, for B cell studies, co-cultures with T cells can model the lymphoma microenvironment .
Live-cell imaging using fluorescently labeled receptors and signaling components allows direct visualization of TNFRSF14 clustering and co-localization with other receptors during co-stimulation. Previous research has successfully employed AlexaFluor-conjugated TNFSF14 (TNFSF14-A594) simultaneously with labeled anti-IgE antibodies to visualize receptor interactions during signaling events .
Genetic approaches using CRISPR-Cas9 to create precise modifications in TNFRSF14 or interacting partners help establish causality in observed co-stimulatory effects. For validation in primary human samples, ex vivo stimulation of cells from patients with different TNFRSF14 genetic backgrounds (normal versus aberrant) provides clinically relevant insights into co-stimulatory mechanisms .
Integrating genomic and proteomic approaches creates a comprehensive map of TNFRSF14 signaling networks that connects genetic variation to functional outcomes. Next-generation sequencing should be employed to identify TNFRSF14 mutations, copy number variations, and expression patterns across different cell types and disease states. This genomic foundation establishes the baseline variation in TNFRSF14 biology, as demonstrated in studies of follicular lymphoma where TNFRSF14 aberrations significantly impact function .
Phosphoproteomics provides direct measurement of signaling cascade activation following TNFRSF14 engagement, capturing both canonical and non-canonical pathways. Time-course experiments following TNFRSF14 stimulation can reveal the temporal dynamics of signaling networks, particularly important for co-stimulatory contexts where TNFRSF14 enhances other pathways like FcεRI signaling in mast cells .
Interactome analysis using proximity labeling techniques (BioID, APEX) can identify novel TNFRSF14-interacting proteins within living cells, expanding our understanding of the receptor's signaling complexes. For validation, CRISPR-Cas9 screening targeting components of identified networks can establish their functional relevance in TNFRSF14-mediated responses.
Data integration across platforms requires computational approaches including pathway analysis, network modeling, and machine learning to synthesize disparate datasets. These methods can identify critical nodes and potential therapeutic targets within TNFRSF14 networks. Correlation of integrated network maps with clinical outcomes, as seen in the association between TNFRSF14 aberrations and GVHD incidence , connects molecular mechanisms to disease relevance.
Therapeutic targeting of the TNFRSF14 pathway in inflammatory conditions like asthma offers multiple promising intervention points. Monoclonal antibodies against TNFRSF14 represent a direct approach with demonstrated efficacy in preclinical models. Research has shown that anti-TNFRSF14 antibody treatment administered after antigen sensitization significantly reduced multiple features of asthma pathology in mouse models, including airway hyperreactivity, inflammation, and remodeling . This approach blocks the co-stimulatory effect of TNFRSF14 on mast cell activation, thereby attenuating allergic inflammatory responses.
Recombinant soluble TNFRSF14 proteins could act as decoy receptors, sequestering TNFSF14 and preventing its interaction with cell-bound TNFRSF14. This approach might be particularly effective in asthma where TNFSF14 levels correlate with disease severity . Small molecule inhibitors targeting downstream signaling components unique to the TNFRSF14 pathway could provide more selective modulation than broader immunosuppressive approaches.
Cell-specific delivery systems could target therapeutic agents specifically to TNFRSF14-expressing cells like mast cells, potentially increasing efficacy while reducing systemic effects. Combined therapeutic approaches targeting both TNFRSF14 and classical allergic pathways (like anti-IgE therapy) might provide synergistic benefits in severe asthma by addressing multiple mechanistic aspects simultaneously.
Ongoing research should focus on developing biomarkers to identify patients most likely to benefit from TNFRSF14-targeted therapies, potentially including genetic testing for TNFRSF14 polymorphisms or measurement of soluble TNFSF14 levels in biological fluids.
TNFRSF14 status offers valuable opportunities for personalizing allogeneic transplantation approaches in lymphoma patients. Pretransplant genetic screening for TNFRSF14 aberrations should become a standard risk stratification tool, as follicular lymphoma patients with TNFRSF14 aberrations demonstrate higher incidence of acute graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation . This genetic information can guide individualized transplantation protocols.
For patients with TNFRSF14 aberrations, intensified GVHD prophylaxis regimens are warranted given their heightened risk profile. These might include more potent immunosuppressive combinations or extended prophylaxis duration compared to standard approaches . Conversely, patients with wild-type TNFRSF14 might safely receive less intensive immunosuppression, potentially improving graft-versus-lymphoma effects.
Novel targeted approaches could include antibodies against BTLA, the binding partner of HVEM (TNFRSF14), which could compensate for reduced HVEM expression in patients with TNFRSF14 aberrations. Murine models have shown that agonistic antibody-mediated BTLA stimulation reduces donor T-cell–mediated acute GVHD, suggesting therapeutic potential in human transplantation .
Post-transplant monitoring strategies should be tailored based on TNFRSF14 status, with more frequent and intensive monitoring for GVHD in high-risk patients with TNFRSF14 aberrations. This stratified approach to transplantation based on TNFRSF14 status represents a significant advance toward precision medicine in hematologic malignancies, potentially improving outcomes by balancing GVHD risk against anti-lymphoma efficacy.
Development of research tools targeting TNFRSF14 pathway components requires strategic considerations to ensure specificity, versatility, and translational relevance. Antibody development should prioritize highly specific monoclonal antibodies that can distinguish between TNFRSF14 and related TNF receptor family members, with separate antibodies for detection, functional blocking, and agonistic stimulation. Validation across multiple techniques (flow cytometry, immunohistochemistry, Western blotting) ensures broad applicability .
Recombinant proteins should include both the full-length extracellular domain of TNFRSF14 and truncated variants representing specific binding regions. Fluorescent labeling (as demonstrated with TNFSF14-A594) enables visualization of receptor-ligand interactions and clustering dynamics . For maximum utility, proteins should be available with various tags (His, GST, Fc) to facilitate different experimental approaches.
Genetic tools development should focus on conditional knockout systems that allow tissue-specific or inducible deletion of TNFRSF14 to study context-dependent functions. CRISPR-Cas9 libraries targeting pathway components enable systematic screening approaches. For structural biology applications, expression systems that produce properly folded TNFRSF14 and interacting partners facilitate crystallography and cryo-EM studies to guide rational drug design.
Cell line development should establish reporter systems where TNFRSF14 pathway activation triggers fluorescent protein expression or luciferase activity, enabling high-throughput screening of potential modulators. Finally, standardized protocols for functional assays measuring TNFRSF14-dependent responses across different cell types are essential for comparing results across research groups.
Despite significant advances, several critical knowledge gaps persist in TNFRSF14 biology that warrant focused investigation. The cell type-specific signaling mechanisms downstream of TNFRSF14 remain incompletely characterized across diverse immune populations. While studies have detailed certain aspects in mast cells and B cells , comprehensive signaling pathway mapping is needed to understand context-dependent outcomes of receptor engagement.
The regulatory mechanisms controlling TNFRSF14 expression under physiological and pathological conditions remain poorly understood. Research should investigate transcriptional, post-transcriptional, and epigenetic regulation of TNFRSF14 expression, which could reveal new opportunities for therapeutic modulation. Additionally, the role of alternative splicing variants of TNFRSF14 in modifying receptor function requires systematic investigation.
While TNFRSF14 aberrations clearly impact follicular lymphoma biology , the potential involvement of TNFRSF14 in other malignancies remains underexplored. Comprehensive analysis across cancer types could identify additional settings where TNFRSF14 pathway targeting might prove beneficial. Similarly, the role of TNFRSF14 in non-allergic inflammatory conditions beyond asthma requires investigation to determine the pathway's broader relevance.
The complex interplay between TNFRSF14 and other co-stimulatory/co-inhibitory pathways within the immunological synapse needs deeper investigation. Understanding how TNFRSF14 signaling integrates with other immune checkpoint molecules could inform more effective combinatorial therapeutic approaches. Finally, longitudinal studies examining how TNFRSF14 function changes during disease progression or in response to therapy would provide valuable insights into dynamic roles of this receptor.
Emerging single-cell and spatial biology techniques promise to revolutionize our understanding of TNFRSF14 in human diseases by providing unprecedented resolution of cellular heterogeneity and tissue context. Single-cell RNA sequencing combined with TCR/BCR repertoire analysis can reveal how TNFRSF14 expression correlates with immune cell clonality and antigen specificity in disease settings. This approach could identify specialized immune cell subsets with unique TNFRSF14 expression patterns and functions that are obscured in bulk analyses .
Spatial transcriptomics techniques like Visium or MERFISH enable mapping of TNFRSF14 expression patterns within tissue microenvironments while preserving architectural context. This approach could reveal how TNFRSF14-expressing cells are distributed relative to other immune and stromal cells in lymphoid tissues, inflammatory sites, or tumor microenvironments.
Multiplexed ion beam imaging (MIBI) or Co-detection by indexing (CODEX) allow simultaneous visualization of dozens of proteins, enabling comprehensive mapping of TNFRSF14 alongside its ligands, signaling partners, and functional markers within intact tissues. These technologies could reveal previously unappreciated cellular interactions mediated by TNFRSF14 in both health and disease.
Single-cell ATAC-seq combined with transcriptomics can illuminate the epigenetic regulation of TNFRSF14 across cell states, potentially identifying regulatory elements that control context-specific expression. Integration of multi-omic datasets using computational approaches like trajectory inference and gene regulatory network analysis will likely reveal new insights into how TNFRSF14 functions within complex cellular ecosystems in human diseases. These advanced techniques will ultimately enable more precise targeting of TNFRSF14 pathways in personalized medicine approaches.
Breakthrough insights in TNFRSF14 research will likely emerge from innovative interdisciplinary approaches that integrate diverse scientific perspectives. Systems biology approaches combining multi-omic data (genomics, transcriptomics, proteomics, metabolomics) with computational modeling can map comprehensive TNFRSF14 signaling networks and predict therapeutic vulnerabilities. This holistic perspective could reveal emergent properties not apparent from studying individual components in isolation.
Structural biology integrated with computational drug design represents another promising interdisciplinary approach. Cryo-electron microscopy and X-ray crystallography of TNFRSF14 complexes with its various binding partners could guide rational design of selective modulators with therapeutic potential. Molecular dynamics simulations could further predict how genetic variants affect receptor function and ligand interactions.
Engineering approaches including protein engineering and synthetic biology offer tools to create modified TNFRSF14 variants with enhanced or altered functions. These engineered receptors could serve as research tools or potential therapeutics with optimized properties. Similarly, nanobiotechnology could enable development of targeted delivery systems for TNFRSF14 pathway modulators to specific cell populations.
Clinical-basic science partnerships are essential for translational advances. Biobanking initiatives collecting patient samples with comprehensive clinical annotation and long-term follow-up, combined with advanced molecular analyses, could connect TNFRSF14 biology to clinical outcomes . Finally, artificial intelligence and machine learning applied to large-scale TNFRSF14-related datasets could identify patterns and relationships not apparent through traditional analysis methods, potentially revealing novel biomarkers or therapeutic approaches.
HVEM-Fc (Human Recombinant) is a fusion protein that combines the extracellular domain of the Herpesvirus Entry Mediator (HVEM) with the Fc region of human immunoglobulin G1 (IgG1). This recombinant protein is used in various research applications, particularly in immunology and virology, due to its role in modulating immune responses.
Herpesvirus Entry Mediator (HVEM), also known as Tumor Necrosis Factor Receptor Superfamily Member 14 (TNFRSF14) and CD270, is a type I transmembrane protein belonging to the TNF receptor superfamily . HVEM is expressed on various immune cells, including T cells, B cells, natural killer (NK) cells, monocytes, and neutrophils . It plays a crucial role in both promoting and inhibiting T cell activity .
The mature human HVEM protein consists of a 164 amino acid extracellular domain (ECD) with three cysteine-rich domains (CRDs), a 21 amino acid transmembrane segment, and a 60 amino acid cytoplasmic tail containing a TRAF interaction domain . The extracellular domain of HVEM is responsible for binding to its ligands, such as LIGHT (TNFSF14), BTLA (B and T Lymphocyte Attenuator), and CD160 .
The Fc region of human immunoglobulin G1 (IgG1) is a constant region that mediates various effector functions, including antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). The Fc region also enhances the stability and half-life of the fusion protein in vivo .
The HVEM-Fc fusion protein is created by fusing the extracellular domain of HVEM to the Fc region of human IgG1. This fusion enhances the stability and solubility of HVEM, making it suitable for various experimental applications . The recombinant HVEM-Fc protein is typically produced in mammalian cell lines, such as HEK293 or NS0 cells, to ensure proper folding and post-translational modifications .
HVEM-Fc is widely used in research to study the interactions between HVEM and its ligands. It is particularly useful in investigating the modulation of immune responses, as HVEM can both activate and inhibit T cell activity depending on the context . Additionally, HVEM-Fc is employed in assays to block or neutralize the activity of HVEM ligands, providing insights into the mechanisms of immune regulation .