FGL1 is implicated in liver regeneration, metabolic diseases, and cancer progression:
FGL1 is upregulated in multiple cancers, including:
FGL1 suppresses anti-tumor immunity by:
Blocking LAG-3: Inhibits CD8⁺ T-cell activation and NK cell cytotoxicity .
Recruiting MDSCs: Increases CD11b⁺/Ly6G⁺ cell infiltration in tumors, dampening immune responses .
Synergizing with PD-L1: Engineered extracellular vesicles co-expressing FGL1/PD-L1 enhance immunosuppression in transplantation models .
ccRCC models: FGL1 knockdown reduces migration/invasion (Transwell assay) and tumor growth in orthotopic xenografts .
Liver metastasis: Fgl1 knockout mice show reduced metastatic burden and increased CD8⁺ T/NK cell activity .
Mechanistic pathways: FGL1 upregulates EMT factors (e.g., Snail, Slug) and cytokines (IL-6, CXCL2) in tumors .
FGL1 is quantified using sandwich ELISA with mAb-based matched pair detection sets:
Fibrinogen-like protein 1, HP-041, Hepassocin, Hepatocyte-derived fibrinogen-related protein 1, HFREP-1, Liver fibrinogen-related protein 1, LFIRE-1, FGL1, HFREP1.
MKHHHHHHAS LEDCAQEQMR LRAQVRLLET RVKQQQVKIK QLLQENEVQF LDKGDENTVI DLGSKRQYAD CSEIFNDGYK LSGFYKIKPL QSPAEFSVYC DMSDGGGWTV IQRRSDGSEN FNRGWKDYEN GFGNFVQKHG EYWLGNKNLH FLTTQEDYTL KIDLADFEKN SRYAQYKNFK VGDEKNFYEL NIGEYSGTAG DSLAGNFHPE VQWWASHQRM KFSTWDRDHD NYEGNCAEED QSGWWFNRCH SANLNGVYYS GPYTAKTDNG IVWYTWHGWW YSLKSVVMKI RPNDFIPNVI.
Fibrinogen-like protein 1 (FGL1) is a protein structurally related to fibrinogen. In humans, it is encoded by the FGL1 gene. FGL1 belongs to the fibrinogen family of proteins, which includes fibrinogen, fibrinogen-like protein 2, and clotting factors V, VIII, and XIII. FGL1 serves as a major immune regulatory protein in the human body.
FGL1 is predominantly secreted by hepatocytes in the liver. The expression pattern varies in pathological conditions, with significant implications for both liver disorders and immune regulation. Research indicates that liver-specific expression of FGL1 is crucial for understanding its biological functions in both normal physiological states and disease conditions.
Contrary to some earlier hypotheses, FGL1 expression shows a biphasic pattern in metabolic liver disease progression. Studies in mouse models demonstrate that Fgl1 mRNA expression is initially induced after 4 weeks on a Western diet but significantly decreases between weeks 4-8 of continued dietary challenge. Similar expression patterns have been observed in human patients with metabolic dysfunction-associated steatohepatitis (MASH) and hepatocellular carcinoma (HCC), where FGL1 expression is significantly reduced compared to healthy controls.
Research has identified four distinct LAG-3/FGL1 expression subtypes in tumors that significantly impact prognosis:
These expression patterns provide important stratification criteria for predicting immunotherapy outcomes.
Tumors with high FGL1 expression demonstrate multiple immunosuppressive characteristics:
Elevated expression of T-cell exhaustion markers (TIM-3, CD39, and NRP1)
Increased CD4+ regulatory T-cell signatures
Enhanced M2-like macrophage infiltration
Upregulation of dendritic cell signatures
Activation of immunosuppressive pathways including epithelial-mesenchymal transition (EMT), angiogenesis, and TGFβ signaling
These features create an immune-evasive microenvironment that contributes to therapy resistance.
Multiplex immunohistochemistry (mIHC) represents the gold standard for comprehensive FGL1 analysis in tumor tissues. This approach allows for simultaneous evaluation of multiple markers including:
FGL1 expression in tumor cells
LAG-3 expression on immune cells
CD8+ T-cell infiltration
CD4+ T-cell distribution
Foxp3+ regulatory T-cells
PanCK (for tumor cell identification)
Cell segmentation based on membrane staining (except for Foxp3, which requires nuclear staining) enables quantitative spatial analysis of the tumor microenvironment. Researchers should quantify cell density (cells/mm²) for all markers, including double-positive populations in both tumor and stromal compartments.
Despite the importance of FGL1 in various pathologies, standardized methods for reliable quantification of FGL1 protein levels in human samples remain underdeveloped. As noted in recent literature, "The study of FGL1 protein level in human will await the development of new analytical tools and standardized methods." Researchers should be aware of this methodological gap when designing FGL1-focused studies and consider validating any new analytical approaches thoroughly before implementation.
Several genetic mouse models have been developed for studying FGL1 function:
Fgl1tm1b(EUCOMM)Hmgu mice: Global knockout model available from the international mouse phenotyping consortium
Fgl1fl/fl mice: Generated by crossing with FLPO recombinase-expressing mice to remove the EUCOMM cassette
Fgl1LKO mice: Liver-specific knockout created by breeding Fgl1fl/fl mice with mice expressing CRE recombinase under the control of the albumin promoter
These models enable tissue-specific investigation of FGL1 function in metabolic disorders and cancer. Statistical analysis of results should employ appropriate tests (Student's t-test, ANOVA) with p<0.05 considered significant.
Quantitative spatial analysis using multiplex immunohistochemistry has revealed critical insights into how FGL1 expression affects immune cell organization:
| Cell Population | Median Density in Tumor Microenvironment |
|---|---|
| Tumor FGL1+ cells | 554.97-615.8 cells/mm² |
| Stromal CD4+ cells | 166.87-328.44 cells/mm² |
| Stromal CD8+ cells | 145.31-217.99 cells/mm² |
| Stromal Foxp3+ cells | 134.66-209.21 cells/mm² |
The spatial relationship between stromal CD8+LAG-3+ cells and tumor FGL1+ cells is particularly significant for predicting response to PD-1 blockade. Patients categorized as stromal CD8+LAG-3+ high/tumor FGL1+ low demonstrated superior progression-free survival compared to those with stromal CD8+LAG-3+ high/tumor FGL1+ high profiles (p=0.006).
Differential gene expression analysis between LAG-3 high FGL1 low and LAG-3 high FGL1 high tumors has revealed several key molecular drivers of FGL1-mediated immunosuppression:
Tumor microenvironment-related genes: Upregulation of NRP1, ECM2, STAT3, and CD163 in FGL1-high tumors
TGF-β signaling pathway components: Elevated TGFB1, TGFB3, and TGFBR1 expression
Epithelial-mesenchymal transition markers: Increased VIM, SNAL1, COL4A1/2, and ACTA2 expression
Gene Set Enrichment Analysis (GSEA): Confirmation of enriched pathways including EMT, KRAS signaling, angiogenesis, and CD8+ T-cell exhaustion
Additionally, NRP1 expression (critical for regulatory T-cell function) shows positive correlation with CD4+Treg signature (r=0.493, p<0.001), suggesting a mechanism for maintaining immunosuppressive regulatory T-cell populations.
Distinct genetic alteration patterns correlate with different LAG-3/FGL1 expression subtypes:
RB1 alterations (potential targets for CDK4/6 inhibition) show highest frequency in the LAG-3 high FGL1 low subgroup, followed by LAG-3 low FGL1 high and LAG-3 low FGL1 high subgroups (p=0.012)
FGFR3 variations (targets for erdafitinib) are associated with LAG-3-low groups regardless of FGL1 expression level (p<0.001)
These genetic associations suggest potential combination therapy opportunities, integrating targeted therapies with immunotherapies based on LAG-3/FGL1 expression profiles.
FGL1 expression demonstrates dynamic changes during disease progression that contradict some previous reports. In metabolic liver disease models, Fgl1 mRNA expression is initially induced but subsequently decreases significantly during disease progression. This biphasic expression pattern has important implications:
Early induction may represent a compensatory response
Later suppression could contribute to disease progression
Timing of therapeutic interventions targeting FGL1 may be crucial for efficacy
These temporal dynamics suggest that longitudinal monitoring of FGL1 expression could provide valuable prognostic information in both cancer and metabolic disorders.
The LAG-3/FGL1 pathway represents a promising target for enhancing immunotherapy efficacy, particularly in patients resistant to PD-(L)1 blockade. Research suggests several strategic approaches:
LAG-3/FGL1 blockade may reverse immunosuppressive contextures, particularly in LAG-3 high FGL1 high tumors
Combination therapies targeting both LAG-3/FGL1 and PD-(L)1 pathways could provide synergistic benefits
Patient stratification based on LAG-3/FGL1 expression subtypes may optimize treatment selection
Given that LAG-3 expression and high FGL1 coexpression are important predictive factors of adverse oncological outcomes, therapeutic interventions targeting this pathway warrant further mechanistic and clinical studies.
Studies in liver-specific FGL1 knockout (Fgl1LKO) mice have revealed important connections between FGL1 and metabolic dysfunction:
Fgl1LKO mice display increased body weight compared to wild-type littermates
Under western diet conditions, Fgl1LKO mice exhibit increased liver damage
Elevated AST, ALT, and triglyceride levels suggest enhanced sensitivity to diet-induced damage
FGL1 expression patterns change during metabolic disease progression in both mice and humans
These findings indicate that FGL1 plays a protective role against metabolic liver injury, with important implications for conditions like metabolic dysfunction-associated steatohepatitis (MASH) and hepatocellular carcinoma (HCC).
FGL1 plays a significant role in liver function. It can induce hepatocyte proliferation and aid in liver injury recovery by activating the epidermal growth factor receptor (EGFR)/EGFR kinase (ERK) and the Src-dependent pathway . This makes it a crucial protein in liver regeneration and repair processes.
Recent studies have highlighted FGL1 as a newly emerging checkpoint ligand of lymphocyte activation gene 3 (LAG3), emphasizing its potential as a target for immune checkpoint therapy . Immune checkpoints are essential in modulating the immune response and mediating T cell dysfunction in autoimmunity and inflammation . However, these inhibitory pathways can be exploited by tumor cells to promote immune escape .
The targeting of FGL1/LAG3 is considered the next generation of immune checkpoint therapy . This approach aims to reverse T cell exhaustion by targeting immune checkpoints, allowing cytotoxic T cells to attack tumor cells . This has significant implications for cancer treatment, especially in overcoming resistance to current therapies like anti-PD-1/PD-L1 monoclonal antibodies .