SIRPG is selectively expressed on adaptive immune cells:
SIRPG localizes to the plasma membrane and polarizes to the immune synapse during T cell-antigen-presenting cell (APC) interactions, facilitating CD47-mediated adhesion .
SIRPG interacts with CD47, a ubiquitously expressed ligand, to regulate immune responses:
Polymorphisms in SIRPG influence disease susceptibility and immune function:
rs2281808 TT: Linked to reduced SIRPG expression, leading to impaired T cell memory and increased susceptibility to autoimmunity .
Splicing Regulation: RNA-seq data show rs6043409 A allele decreases isoforms 1/2 (involved in CD47 binding) and increases isoform 3 (unknown function) .
SIRPG promotes lung squamous cell carcinoma (LUSC) progression by:
Activating M1 Macrophages: Upregulates pro-inflammatory cytokines (e.g., IFN-γ, TNF-α) and chemokines (CCL2, CCL3) .
Disrupting JAK-STAT Signaling: Alters stem cell differentiation pathways, favoring tumor growth .
Immune Cell Infiltration: Correlates with reduced T helper cells and regulatory T cells (Tregs) .
Clinical Relevance: High SIRPG expression in LUSC is associated with poor prognosis, suggesting its utility as a biomarker .
SIRPγ (encoded by the SIRPG gene) is uniquely expressed by human T-cells, unlike other immunomodulatory SIRP family members. It appears to function as an important checkpoint regulator of human effector T-cells. Research has demonstrated that SIRPγ expression levels can compartmentalize CD8 T-cells into distinct phenotypic and functional populations with different effector capabilities . While other SIRP family members are expressed on various immune cells, SIRPγ's T-cell specificity makes it particularly interesting for T-cell-focused immunological research.
SIRPγ expression is commonly measured using flow cytometry to detect surface expression on CD4 and CD8 T-cells. Researchers typically evaluate both the percentage of SIRPγ-expressing cells and the mean fluorescence intensity (MFI) to quantify expression levels. RNA sequencing can also be employed to analyze SIRPG gene expression and identify differentially expressed genes between SIRPγ-high and SIRPγ-low cell populations . When designing experiments to measure SIRPγ, it's important to include appropriate controls and consider the bimodal distribution pattern often observed on CD8 T-cells.
The SNP rs2281808, located within the SIRPG gene, significantly influences SIRPγ expression on T-cells. Research across 79 healthy donors revealed distinct expression patterns based on genotype:
Genotype | SIRPγ Expression Pattern | SIRPγ-MFI on CD4 T-cells | SIRPγ-MFI on CD8 T-cells |
---|---|---|---|
CC | High expression on >80% T-cells | 526 ± 244 | 439 ± 170 |
CT | Intermediate expression with 25-50% SIRPγ-low T-cells | 350 ± 123 | 275 ± 93 |
TT | Low expression on >80% T-cells | 203 ± 10.8 | 160 ± 7.9 |
The rs2281808 TT variant results in significantly reduced surface expression of SIRPγ on both CD4 and CD8 T-cells compared to CC carriers (p<0.01) . This genetic variation appears to cause an imbalance in the ratio of SIRPγ-low vs. SIRPγ-high CD8 T-cells in CT/TT individuals, potentially contributing to autoimmune disease susceptibility.
When genotyping rs2281808 in SIRPG research, standard SNP genotyping methods like PCR-RFLP, TaqMan assays, or next-generation sequencing approaches can be employed. The research literature indicates that genotyping should be combined with phenotypic assessment of SIRPγ expression through flow cytometry to establish the functional correlation between genotype and expression levels . For large cohort studies, high-throughput methods like SNP arrays or targeted sequencing panels that include rs2281808 are recommended to ensure consistent and reliable genotyping across samples.
SIRPγ expression levels profoundly influence CD8 T-cell function, creating distinct functional populations. SIRPγ-low CD8 T-cells display:
Heightened effector state with lower activation threshold
Increased expression of genes associated with cytotoxic potential (granzymes, IFN-γ)
Enhanced migratory capacity (higher expression of integrins, CCL3, CCL4, CCL5)
Activation of effector-associated genes (T-bet, EOMES, CD244, CD247, SLAMF7)
Deficiency in transcription factors associated with long-term memory formation
RNA sequencing analysis revealed 399 genes significantly upregulated and 593 genes downregulated in SIRPγ-low versus SIRPγ-high CD8 T-cells (log2 fold-change ≥1, adjusted p-value <0.05) . The heightened effector state of SIRPγ-low cells suggests SIRPγ functions as a checkpoint that restrains T-cell effector functions, with reduced expression potentially contributing to hyperresponsive T-cells in autoimmune conditions.
In human T-cells, SIRPγ appears to engage the Hippo/YAP signaling pathway. Research indicates that SIRPγ can bridge MST1 and PP2A to facilitate MST1 dephosphorylation, which results in Hippo/YAP activation . This pathway activation leads to cytokine release that can influence the tumor microenvironment in cancer contexts. Additionally, SIRPγ-low CD8 T-cells show heightened expression of MAP3K8, a serine/threonine kinase selectively expressed by effector CTLs in humans . Understanding these signaling mechanisms is crucial for developing targeted interventions that modulate SIRPγ function in therapeutic contexts.
When designing experiments to study SIRPγ in human T-cells, researchers should consider:
Genotyping participants for rs2281808 to account for genetic influence on expression
Using flow cytometry with appropriate gating strategies to distinguish SIRPγ-high vs. SIRPγ-low populations
Considering the bimodal distribution of SIRPγ on CD8 T-cells, particularly pronounced in CT carriers
Including functional assays to assess T-cell activation, cytokine production, and cytotoxic potential
Controlling for T-cell differentiation state (naïve, central memory, effector memory, terminally differentiated)
Implementing appropriate control groups based on experimental design principles
For interventional studies, researchers should clearly operationalize variables and consider using blinded study designs to control for experimenter bias and participant expectations .
For SIRPγ knockdown studies in primary human T-cells, researchers can employ several approaches:
siRNA/shRNA: Using transfection or transduction methods optimized for primary T-cells
CRISPR-Cas9: For more permanent genetic modification, though efficiency may vary in primary cells
Neutralizing antibodies: Using SIRPγ-specific neutralizing antibodies to block function without genetic modification
Key considerations include:
Confirming knockdown efficiency through flow cytometry and qPCR
Assessing cell viability post-intervention
Including appropriate controls (scrambled siRNA, isotype antibodies)
Measuring functional outcomes such as cytokine production (IFNγ, TNFα, granzyme B)
Comparing results between genetically different donors (CC vs. CT vs. TT) to understand interaction effects
Research has shown that SIRPγ knockdown in primary human T-cells increases secretion of IFNγ, TNFα, and granzyme B from CD8 T-cells and IFNγ from CD4 T-cells upon activation .
Multiple genome-wide association studies (GWAS) have linked the SNP rs2281808 TT variant in the SIRPG gene to autoimmune diseases, particularly Type 1 Diabetes . The functional evidence suggests this association may be mediated through:
Reduced SIRPγ expression on T-cells in TT carriers
Enhanced effector function of SIRPγ-low CD8 T-cells
Lower activation threshold of SIRPγ-low T-cells, potentially increasing reactivity to self-antigens
Deficiency in long-term memory formation transcription factors in SIRPγ-low CD8 T-cells
This evidence collectively suggests that SIRPγ functions as an immune checkpoint that, when compromised by genetic variation, may contribute to dysregulated immune responses characteristic of autoimmune conditions.
SIRPγ plays a complex role in cancer immune evasion. Research has shown that in lung adenocarcinoma (LUAD):
SIRPγ is upregulated and its overexpression predicts poor survival outcomes
SIRPγ-high cells serve as cancer stem-like cells (CSLCs) and tumor immune checkpoint-initiating cells
SIRPγ bridges MST1 and PP2A to facilitate Hippo/YAP activation, leading to cytokine release
These cytokines stimulate CD47 expression in LUAD cells
Increased CD47 expression inhibits tumor cell phagocytosis, promoting immune escape
Importantly, targeting SIRPγ through genetic knockdown or neutralizing antibodies inhibited CSLC phenotypes and elicited phagocytosis that suppressed tumor growth in vivo. This suggests SIRPγ as a potential therapeutic target combining both immune and cancer stem-like cell targeting strategies.
Single-cell technologies offer powerful approaches to understand SIRPγ heterogeneity:
Single-cell RNA sequencing (scRNA-seq): Can reveal transcriptional profiles associated with different SIRPγ expression levels across T-cell subsets, potentially identifying novel SIRPγ-regulated pathways
CyTOF/mass cytometry: Enables simultaneous detection of SIRPγ with dozens of other surface and intracellular markers to comprehensively map phenotypic relationships
Single-cell ATAC-seq: Could identify chromatin accessibility differences between SIRPγ-high and SIRPγ-low populations to understand epigenetic regulation
Spatial transcriptomics: May reveal tissue-specific SIRPγ expression patterns in disease contexts
These approaches would extend beyond bulk RNA sequencing analyses that have already identified hundreds of differentially expressed genes between SIRPγ-high and SIRPγ-low CD8 T-cells , providing higher resolution understanding of functional heterogeneity.
Developing therapeutic approaches targeting SIRPγ presents several methodological challenges:
Specificity: Ensuring targeted approaches specifically affect SIRPγ without cross-reactivity with other SIRP family members
Cell type selectivity: Developing methods to target SIRPγ specifically on relevant T-cell subsets while sparing others
Context-dependent function: Addressing SIRPγ's potentially opposing roles in autoimmunity versus cancer
Therapeutic window: Determining optimal degree of SIRPγ inhibition that enhances anti-tumor immunity without triggering autoimmunity
Biomarker development: Establishing reliable methods to monitor SIRPγ expression/function during treatment
Combination strategies: Designing rational combination approaches with existing immunotherapies
Current research using genetic knockdown and neutralizing antibodies against SIRPγ has shown promise in inhibiting cancer stem-like cell phenotypes and eliciting anti-tumor immune responses , but translating these approaches to clinical applications requires addressing these methodological challenges.
Signal-Regulatory Protein Gamma (SIRPγ) is a member of the signal-regulatory protein (SIRP) family, which belongs to the immunoglobulin superfamily. SIRPγ is a type I transmembrane glycoprotein that plays a crucial role in the regulation of immune responses. It is uniquely expressed on T lymphocytes and is involved in the integrin-independent adhesion of lymphocytes to antigen-presenting cells .
The preparation of human recombinant SIRPγ typically involves the expression of the protein in a suitable host system, such as Escherichia coli (E. coli). The gene encoding SIRPγ is cloned into an expression vector, which is then introduced into the host cells. The host cells are cultured under conditions that promote the expression of the recombinant protein. After sufficient expression, the cells are harvested, and the protein is purified using techniques such as affinity chromatography .
SIRPγ interacts with its physiological ligand, CD47, which is also known as the “don’t eat me” signal. This interaction mediates cell-cell adhesion and plays a role in the modulation of immune responses. The binding of SIRPγ to CD47 enhances antigen-specific T-cell proliferation and costimulates T-cell activation . The crystal structure of SIRPγ in complex with an antibody Fab fragment has been determined, revealing that the epitope for the Fab fragment is formed at the interface of the first and second domains of SIRPγ. This interaction stabilizes a conformation that favors SIRP dimer formation in the crystal structure .