SLAMF6 is a type I transmembrane protein composed of an extracellular immunoglobulin variable (V) domain, a constant (C) domain, a transmembrane region, and a cytoplasmic tail containing immunoreceptor tyrosine-based switch motifs (ITSMs) . Alternative splicing generates three isoforms:
Canonical SLAMF6: Full-length protein with intact V and C domains, associated with inhibitory signaling .
SLAMF6Δ17–65: Lacks amino acids 17–65 in the V domain, acting as a co-stimulatory receptor by recruiting SHP1 and enhancing cytotoxic T cell responses .
SLAMF6ΔExon2: Missing the entire V domain, though functional data remain limited .
Isoform | Structure | Function |
---|---|---|
Canonical SLAMF6 | Full V and C domains | Co-inhibitory (via SAP recruitment) |
SLAMF6Δ17–65 | Truncated V domain | Co-stimulatory (via SHP1 pathway) |
SLAMF6ΔExon2 | No V domain | Undetermined |
SLAMF6 is expressed on hematopoietic cells, including:
T cells: Found on CD4⁺, CD8⁺, and γδ T cells, with higher expression in progenitor-exhausted CD8⁺ T cells .
B cells and monocytes: Elevated in tumor-associated macrophages (TAMs) in hepatocellular carcinoma (HCC) .
SLAMF6 clustering at the immunological synapse enhances TCR signaling via Rap1 activation, improving T cell adhesion and cytotoxicity .
In chronic lymphocytic leukemia (CLL) and melanoma models, SLAMF6⁺ CD8⁺ T cells exhibit exhaustion (PD-1⁺), but anti-SLAMF6 antibodies restore effector functions .
In HCC, SLAMF6 promotes M2-like macrophage polarization via NF-κB suppression, accelerating tumor progression .
The co-stimulatory SLAMF6Δ17–65 isoform drives Th1/Th17 differentiation and correlates with improved responses to immune checkpoint blockade .
Hepatocellular Carcinoma: SLAMF6 overexpression in TAMs promotes tumor cell proliferation, migration, and invasion .
Melanoma and Leukemia: SLAMF6Δ17–65 enhances tumor-infiltrating lymphocyte (TIL) cytotoxicity, reducing tumor growth in murine models .
Systemic Lupus Erythematosus (SLE): Despite normal SLAMF6 expression, SLE T cells show impaired Th1 cytokine production upon SLAMF6 engagement .
High SLAMF6 expression correlates with better survival in cancer patients and is linked to TCF7 (T-cell factor 1) expression, a marker of stem-like T cells .
SLAMF6 is a type I transmembrane protein belonging to the CD2 subfamily of the immunoglobulin superfamily, encoded by the SLAMF6 gene in humans. Its structure comprises an amino terminal Ig-like variable (V) domain and a membrane proximal constant 2 (C2) domain in the extracellular portion . The intracellular portion contains two immune receptor tyrosine-based switch motifs (ITSMs) that serve as binding sites for adaptor molecules such as SLAM adaptor protein (SAP) and Ewing sarcoma associated transcript (EAT-2) .
SLAMF6 complexes form in a "head to head" fashion via interacting IgV-like domains. The receptor undergoes tyrosine phosphorylation and associates with Src homology 2 domain-containing proteins (SH2D1A) as well as with SH2 domain-containing phosphatases (SHPs) .
SLAMF6 is exclusively expressed on hematopoietic cells, specifically on natural killer (NK) cells, T lymphocytes, and B lymphocytes . Expression levels vary across different T cell subsets:
Naïve CD4+ and CD8+ T cells (CD45RA+CCR7+) express SLAMF6 at similar baseline levels
Non-naïve T cells (CD45RA- or CCR7-) have significantly higher levels (approximately 2-fold higher) than naïve cells
Non-naïve CD8+ T cells express significantly higher levels of SLAMF6 than non-naïve CD4+ T cells
Activated (HLA-DR+CD38+) and exhausted (PD-1+) CD4+ T cells express significantly higher levels of SLAMF6 than bulk non-naïve CD4+ T cells
This differential expression pattern suggests SLAMF6 plays specialized roles in different lymphocyte subpopulations and activation states.
In humans, SLAMF6 has three splice isoforms involving its V-domain that exhibit dramatically different functions:
Canonical receptor: Inhibits T-cell activation through SAP recruitment
Short isoform SLAMF6 Δ17–65: Has a strong agonistic effect, with costimulatory action dependent on protein phosphatase SHP1
Additional splice variant: (specific functions less characterized in the available data)
The short isoform leads to a cytotoxic molecular profile mediated by the expression of TBX21 and RUNX3. Notably, patients treated with immune checkpoint blockade show a shift toward SLAMF6 Δ17–65 in peripheral blood T cells, suggesting therapeutic relevance of this isoform .
This "yin-yang" relationship between the inhibitory canonical form and stimulatory short form may represent a natural balancing mechanism in immune regulation.
SLAMF6 plays a critical role in immunological synapse formation and T cell activation through multiple mechanisms:
Synapse formation: SLAMF6 is required for productive TCR downstream signaling. Biochemical and genetic experiments reveal that SLAMF6 is essential for effective TCR-mediated responses .
Clustering effect: Imaging studies demonstrate that SLAMF6 clustering, specifically with the TCR, results in dramatic increases in downstream signaling. Interestingly, the SLAMF6 ectodomain is required for its function but not for its recruitment to the immunological synapse .
Adhesion enhancement: Mechanistically, SLAMF6 enhances T cell function by increasing T cell adhesiveness through activation of the small GTPase Rap1 .
Critical residues: Flow-cytometry analysis has demonstrated that tyrosine 308 of the tail of SLAMF6 is crucial for its ability to enhance T cell function .
SLAMF6 plays a particularly important role in the formation of stable conjugates between effector CD8+ T cells and CD4+ T cell targets, both in virus-specific CTL lines and ex-vivo lymphocytes .
Several approaches have been developed to modulate SLAMF6 expression or function for experimental research:
Genetic knockout models: SLAMF6-deficient mouse models (Pmel-1 x SLAMF6-/-) have been developed to study the effects of SLAMF6 absence on T cell function. These knockout T cells show improved functional capacity with preserved proliferative responses to peptide stimulation .
Splice-switching antisense oligonucleotides (ASOs): Researchers have developed ASOs specifically designed to target SLAMF6 splice junctions. These ASOs can enhance SLAMF6 Δ17–65 expression in human tumor-infiltrating lymphocytes and improve their capacity to inhibit tumors in experimental models .
Antibody-based approaches: Anti-SLAMF6 antibodies have been used for therapeutic effects in autoimmunity and cancer models. Additionally, bispecific antibodies like CD3/SLAMF6 have been created to investigate SLAMF6 localization effects on T cell activation .
Co-localization studies: To study SLAMF6 compartmentalization, researchers have developed approaches to manipulate SLAMF6 localization, demonstrating that SLAMF6 localization within versus outside the immune synapse affects T cell function differently .
Multiple lines of evidence support SLAMF6's role as a novel T cell checkpoint regulator:
Enhanced anti-tumor activity: SLAMF6-deficient T cells demonstrate augmented tumor killing capacity. In mouse models with killer T cells that recognized skin cancer cells and lacked SLAMF6, these modified cells were more effective at fighting cancer, producing more anti-cancer cytokines and killing more cancer cells .
Prolonged anti-tumor effects: SLAMF6-deficient T cells showed lasting effects on tumors and improved survival in experimental models .
Synergy with established checkpoint inhibitors: The effects of SLAMF6 deficiency could be further enhanced by combining with other immunotherapies, suggesting potential for combination approaches .
Isoform switching during therapy: Patients treated with existing immune checkpoint blockade show a shift toward the activating SLAMF6 Δ17–65 isoform in peripheral blood T cells, suggesting SLAMF6 splicing may be a natural response to immune activation .
These findings collectively position SLAMF6 as a potential new target for cancer immunotherapy, particularly in contexts like melanoma where T cell-based approaches are already established.
SLAMF6 expression and function are altered in several disease contexts:
Systemic Lupus Erythematosus (SLE): SLAMF6 expression is increased on the surface of SLE T cells compared to normal cells, suggesting a role in autoimmune dysregulation .
HIV infection: SLAMF6 plays a critical role in the killing of HIV-1-infected CD4+ T cells by HIV-1-specific CTLs. SLAMF6 blockade leads to a reduction in the killing efficiency of infected cells, highlighting its importance in antiviral immunity .
X-linked lymphoproliferative disease: SLAMF6 can mediate inhibitory signals in NK cells from X-linked lymphoproliferative patients, indicating its involvement in this primary immunodeficiency .
Researchers employ several specialized techniques to visualize and quantify SLAMF6 localization:
Imaging studies: Advanced microscopy techniques track SLAMF6 clustering and recruitment to the immunological synapse. These approaches have revealed that while the SLAMF6 ectodomain is required for its function, it is not necessary for recruitment to the immunological synapse .
Bispecific antibody approaches: Researchers have developed bispecific CD3/SLAMF6 antibodies to investigate how forced co-localization affects T cell function. This approach has demonstrated that SLAMF6 localization within the immune synapse provides strong synergistic activation .
Flow cytometry-based conjugate assays: These assays measure the formation of stable conjugates between effector and target cells, allowing researchers to quantify how SLAMF6 manipulation affects this critical step in T cell function .
Phospho-proteomic analysis: This technique has been used to determine that during ligation of the TCR and programmed cell death 1, SLAMF6 ITSMs are differentially phosphorylated, providing insight into SLAMF6's signaling dynamics during T cell activation .
Distinguishing between SLAMF6 splice isoform effects requires specialized approaches:
Isoform-specific antibodies: Development of antibodies that specifically recognize unique epitopes in different splice variants.
RNA-based detection methods: PCR primers or RNA-seq approaches that can differentiate between splice variants based on their unique junction sequences.
Splice-switching antisense oligonucleotides (ASOs): ASOs designed to target specific SLAMF6 splice junctions can selectively enhance expression of particular isoforms like SLAMF6 Δ17–65 .
Functional readouts: Monitoring downstream effects specific to each isoform, such as the cytotoxic molecular profile (TBX21, RUNX3 expression) associated with the SLAMF6 Δ17–65 isoform .
Protein interaction studies: Analyzing differential protein-protein interactions, as the canonical receptor primarily recruits SAP while the short isoform activity depends on SHP1 .
Several experimental systems have proven valuable for investigating SLAMF6's role in cancer immunotherapy:
Genetic knockout models: SLAMF6-deficient mouse models (e.g., Pmel-1 x SLAMF6-/-) have been instrumental in demonstrating SLAMF6's role as a checkpoint regulator. These models show that SLAMF6-deficient T cells have better functional capacity against melanoma targets .
Adoptive T cell transfer models: Systems using melanoma-specific T cells (like Pmel-1) with SLAMF6 manipulation allow researchers to study how SLAMF6 alterations affect anti-tumor immunity in vivo .
Human tumor-infiltrating lymphocyte models: Patient-derived TILs modified with splice-switching ASOs to enhance SLAMF6 Δ17–65 expression have been used to study SLAMF6 manipulation in a more clinically relevant context .
Xenograft models: Human melanoma xenografts in immunocompromised mice, treated with SLAMF6-manipulated human TILs, offer insights into translational aspects of SLAMF6 targeting .
These models collectively enable investigation of both basic biological mechanisms and potential therapeutic applications of SLAMF6 manipulation.
SLAMF6 manipulation shows promising potential for integration with established immunotherapies:
Combination with checkpoint inhibitors: SLAMF6-deficient T cells show enhanced anti-tumor effects that can be further boosted by combining with other immunotherapies, suggesting synergistic potential .
Splice isoform modulation: Patients treated with immune checkpoint blockade naturally shift toward the activating SLAMF6 Δ17–65 isoform, suggesting that actively promoting this shift with splice-switching ASOs might enhance existing checkpoint inhibitor therapies .
Adoptive cell therapy enhancement: Manipulating SLAMF6 expression or function in TILs or CAR-T cells before adoptive transfer could potentially improve their anti-tumor efficacy .
Bispecific antibody approaches: Building on findings that SLAMF6 localization affects its function, bispecific antibodies targeting SLAMF6 and other relevant receptors could offer novel therapeutic strategies .
These approaches could be particularly relevant for patients who don't respond to current immunotherapies, potentially expanding the population of patients who benefit from cancer immunotherapy.
Several important considerations exist when translating SLAMF6 findings between species:
Splice isoform differences: The three splice isoforms of SLAMF6 identified in humans may not have direct counterparts in mouse models, complicating direct translation of findings .
Expression pattern variations: While SLAMF6 is expressed on similar cell types in both species, subtle differences in expression levels or regulation may exist between mice and humans.
Signaling pathway conservation: Although the basic signaling mechanisms of SLAMF6 appear conserved, species-specific differences in adapter proteins or downstream effectors could lead to functional variations.
Therapeutic antibody development: Antibodies developed against mouse SLAMF6 may not cross-react with human SLAMF6 due to sequence differences, requiring separate development of human-specific therapeutic agents.
These challenges highlight the importance of validating key findings in human systems whenever possible before advancing to clinical translation.
While SLAMF6's role in HIV infection has been investigated, its broader involvement in antiviral immunity merits further study:
Synapse formation in antiviral responses: Given SLAMF6's importance in stable immune synapse formation between CTLs and HIV-infected cells , similar roles may exist for other viral infections where CTL-mediated clearance is important.
Viral immune evasion: Some viruses may have evolved mechanisms to interfere with SLAMF6 function as part of immune evasion strategies, similar to how they target other immune receptors.
Isoform regulation during viral infection: The balance between inhibitory and stimulatory SLAMF6 isoforms may be altered during different viral infections, potentially affecting immune response efficacy.
Checkpoint function in chronic viral infection: SLAMF6's emerging role as an immune checkpoint suggests it might contribute to T cell exhaustion in chronic viral infections beyond HIV.
Understanding these broader antiviral roles could inform both basic immunology and the development of antiviral immunotherapeutic approaches.
SLAMF6 is characterized by its extracellular domain, which contains Ig-like domains, a transmembrane region, and a cytoplasmic tail. The receptor is constitutively expressed on various immune cells, including T cells, B cells, natural killer (NK) cells, and dendritic cells . Its expression is upregulated upon activation of these cells, indicating its involvement in immune modulation .
SLAMF6 functions as a co-stimulatory molecule that enhances the activation and proliferation of T cells and NK cells. It is involved in the formation of immunological synapses, which are critical for effective immune responses. SLAMF6 also plays a role in the regulation of cytokine production and the maintenance of immune homeostasis .
Recent studies have highlighted the role of SLAMF6 as an immune checkpoint regulator. It has been shown to be involved in the exhaustion of CD8+ T cells within the tumor microenvironment. T-cell exhaustion is characterized by a loss of proliferative capacity and impaired effector functions, which can hinder effective antitumor responses .
Targeting SLAMF6 has emerged as a potential therapeutic strategy for cancer immunotherapy. Inhibiting SLAMF6 can enhance the cytotoxic functions of CD8+ T cells and improve their ability to eradicate tumors. This approach has shown promise in preclinical models of chronic lymphocytic leukemia (CLL) and melanoma .
Human recombinant SLAMF6 is produced using recombinant DNA technology, which involves the insertion of the SLAMF6 gene into an expression vector, followed by the expression of the protein in a suitable host system. Recombinant SLAMF6 is used in various research applications to study its function and therapeutic potential.