The AGTRAP antibody is a research tool designed to detect the Angiotensin II Receptor-Associated Protein (AGTRAP), a transmembrane protein that interacts with the type 1 angiotensin II receptor (AT1R) to regulate the renin-angiotensin system (RAS). AGTRAP has gained attention for its role in modulating tumor progression, immune infiltration, and metabolic disorders . This article synthesizes data on its structure, applications, commercial availability, and research findings.
AGTRAP is encoded by the AGTRAP gene and functions as a negative regulator of AT1R-mediated signaling by controlling receptor internalization . Its expression is localized to the plasma membrane and perinuclear vesicles . In cancer contexts, AGTRAP overexpression has been linked to pathways such as endoplasmic reticulum (ER) transport, JAK-STAT signaling, and chemokine signaling .
a. Cancer Research
AGTRAP antibodies are critical for studying cancer biology. For example, immunohistochemistry (IHC) studies reveal high AGTRAP expression in glioblastoma, hepatocellular carcinoma (HCC), and pancreatic adenocarcinoma . In HCC, AGTRAP correlates with T-cell exhaustion markers and poor prognosis .
b. Immunohistochemistry and ELISA
AGTRAP antibodies enable tissue-level detection via IHC (e.g., 1:500–1:1000 dilution) and quantification via ELISA . Proteintech’s AGTRAP antibody (11559-1-AP) has been validated for IHC in human pancreas cancer tissue .
b. Immune Infiltration
AGTRAP expression positively correlates with M2 macrophage infiltration in 25 cancers, promoting tumor-associated macrophage (TAM) activity and immune evasion .
c. Immunotherapy Prediction
AGTRAP overexpression predicts favorable responses to immunotherapy in adrenocortical carcinoma (ACC) and glioblastoma multiforme (GBM) .
AGTRAP (Angiotensin II Receptor-Associated Protein) functions primarily by interacting directly with the Angiotensin II Type 1 Receptor (AT1R) to modulate its signaling . It appears to be a negative regulator of type-1 angiotensin II receptor-mediated signaling by regulating receptor internalization and mechanisms of receptor desensitization such as phosphorylation .
Research has revealed that AGTRAP plays a substantial role in tumor progression, with high expression significantly related to poor prognosis in multiple cancer types, including glioma, liver cancer, and kidney chromophobe . Pan-cancer analysis has shown that AGTRAP is highly expressed in breast cancer, lung adenocarcinoma, and several other cancers, making it an important target for oncology research .
AGTRAP is involved in several key signaling pathways:
Immune-related pathways: Natural killer cell-mediated cytotoxicity, chemokine signaling, RIG-I-like receptor signaling, toll-like receptor signaling, and JAK-STAT signaling pathway
Metabolism-related pathways: Taurine and hypotaurine metabolism, glycine, serine, and threonine metabolism
Additional pathways: NF-κB and MAPK signaling pathways in hepatocellular carcinoma
Gene Set Enrichment Analysis (GSEA) has demonstrated that AGTRAP enrichment in these pathways may contribute to tumor development through immune modulation and metabolic reprogramming . The enrichment of chemokine signaling pathway was consistent with the abundant correlation between immunomodulators and AGTRAP, suggesting a function in regulating immunomodulators and promoting tumor progression .
Commercial AGTRAP antibodies target various regions of the protein, with several common epitopes:
The C-terminal region (particularly around amino acids 108-159) appears to be a frequently targeted epitope, suggesting this region may be particularly immunogenic or accessible for antibody binding .
Rigorous validation is essential before using AGTRAP antibodies in experiments:
Western blot validation: Confirm the antibody detects a protein of the expected molecular weight. Note that when using recombinant proteins with tags (e.g., GST tag adds 26 KDa), the observed molecular weight may differ from the predicted value for native AGTRAP .
Cross-reactivity testing: Some antibodies are validated against protein arrays containing hundreds of human recombinant protein fragments to ensure specificity. The Prestige Antibodies, for example, are tested on protein arrays of 364 human recombinant protein fragments .
Tissue validation: Test the antibody on tissues known to express or lack AGTRAP. Commercial antibodies often undergo validation on tissue arrays of 44 normal human tissues and 20 common cancer types .
Positive and negative controls: Include appropriate controls in each experiment, such as tissues with known AGTRAP expression patterns and secondary-only controls to assess background staining.
Multiple antibody approach: When possible, use multiple antibodies targeting different epitopes to confirm findings, especially for novel observations.
Based on published methodologies for AGTRAP IHC staining:
Sample preparation:
Antigen retrieval and blocking:
Antibody incubation:
Detection and scoring:
For optimal Western blot results when detecting AGTRAP:
Protein extraction:
Gel electrophoresis and transfer:
Antibody incubation:
Detection:
AGTRAP expression shows significant correlation with immune cell infiltration across multiple cancer types:
Positive correlation with infiltration of B cells (correlation = 0.225, p = 2.43e-05), CD8+ T cells (correlation = 0.321, p = 1.29e-09), CD4+ T cells (correlation = 0.292, p = 3.52e-08), macrophages (correlation = 0.417, p = 8.18e-16), neutrophils (correlation = 0.345, p = 4.22e-11), and dendritic cells (correlation = 0.406, p = 6.14e-15) in hepatocellular carcinoma .
Association with immune scores: Significantly positive associations were found between AGTRAP and ImmuneScore in multiple cancers, including LGG (lower-grade glioma), NB (neuroblastoma), BLCA (bladder cancer), and several other cancer types .
M2 macrophage correlation: AGTRAP expression was positively related to M2 macrophage infiltration among 25 cancer types . M2 macrophages promote angiogenesis, tissue reconstruction, and tumor progression .
Several bioinformatic approaches have been successfully employed to study AGTRAP:
Expression analysis databases:
Correlation analyses:
Survival analyses:
Pathway analyses:
AGTRAP expression shows significant correlations with T-cell exhaustion markers and immunotherapy response:
T-cell exhaustion markers: AGTRAP expression positively correlates with PD-1 (PDCD1), GZMB, LAG-3, CTLA-4, and HAVCR2 (TIM-3) in hepatocellular carcinoma, suggesting involvement in T-cell exhaustion pathways .
Immunotherapy response prediction: Overexpression of AGTRAP predicts better immunotherapy responses in specific cancer types, including ACC (adrenocortical carcinoma), KICH (kidney chromophobe), GBM (glioblastoma), DLBC (diffuse large B-cell lymphoma), and COAD (colon adenocarcinoma) .
Biomarker potential: The correlation with tumor mutation burden (TMB), microsatellite instability (MSI), and neoantigens suggests that AGTRAP could serve as a predictive biomarker for immunotherapy efficacy .
AGTRAP expression has significant prognostic implications across multiple cancer types:
AGTRAP expression varies significantly between tumor and normal tissues across different cancer types:
These expression patterns have been confirmed through multiple methods, including bioinformatic analyses of TCGA data, immunohistochemistry, and Western blot validation on clinical specimens .
Based on current research findings, AGTRAP shows potential as a therapeutic target through several mechanisms:
Modulation of immune responses: Given its correlation with immune cell infiltration and T-cell exhaustion markers, targeting AGTRAP might enhance anti-tumor immunity .
Combination with immunotherapy: The correlation with immunotherapy response biomarkers suggests potential for combination approaches targeting AGTRAP alongside immune checkpoint inhibitors .
Targeting associated signaling pathways: Interventions aimed at the NF-κB and MAPK signaling pathways that AGTRAP influences may provide therapeutic benefit in cancers where AGTRAP is overexpressed .
RNA interference approaches: The validation of AGTRAP antibodies for RNAi applications suggests this as a potential approach for therapeutic development .
Researchers face several challenges when studying AGTRAP in complex tissues:
Cell-type specificity: AGTRAP is expressed in multiple cell types, including Kupffer cells and T cells in the liver, requiring careful interpretation of bulk tissue analysis .
Subcellular localization: AGTRAP is primarily localized in cell plasma, which may require specialized techniques for accurate visualization and quantification .
Post-translational modifications: Current data on AGTRAP post-translational modifications is limited, which may impact antibody recognition and functional studies .
Isoform specificity: Multiple AGTRAP antibodies recognize different epitopes, potentially capturing different isoforms or modified forms of the protein, necessitating careful antibody selection .
Cross-reactivity concerns: While most commercial antibodies are validated for human AGTRAP, cross-reactivity with mouse or rat orthologs varies, requiring validation for cross-species studies .