AGR3 is expressed in secretory and ciliated epithelial cells, including:
Airway epithelium: Regulates mucociliary clearance and calcium-modulated ciliary beat frequency .
Reproductive tissues: Upregulated by androgens and estrogens, with roles in hormone-responsive cancers (e.g., breast, prostate) .
Gastrointestinal tract: Maintains epithelial barrier function .
Key findings from functional studies:
COPD Pathogenesis: Reduced AGR3 expression in frequent COPD exacerbators correlates with downregulated epithelial junction proteins (E-cadherin, occludin, ZO-1), increasing susceptibility to infections .
Cancer Biology:
Analysis of TCGA data identified somatic variations in AGR3 across 32 tumor types :
Variation Type | AGR3 (n=35 tumors) | AGR2 (n=32 tumors) |
---|---|---|
Missense Mutations | 84% | 77% |
Nonsense Mutations | 6% | 9% |
Frameshift Mutations | 3% | 6% |
Splice Mutations | 3% | 6% |
Notable cancer associations include cutaneous melanoma, endometrial carcinoma, and bladder cancer .
Ovarian Cancer: AGR3 positivity (>10% stained cells) correlates with longer median survival in high-grade tumors (95.5 vs. 41.9 months, p=0.008) .
COPD: Reduced AGR3 mRNA (p=0.04) and protein (p=0.009) levels predict frequent exacerbations .
AGR3 overexpression rescues cigarette smoke extract (CSE)-induced junctional protein loss in airway epithelia, suggesting potential for gene therapy in COPD . In contrast, its pro-metastatic role in breast cancer via Src kinase activation highlights context-dependent therapeutic strategies .
The AGR3 protein, also known as Anterior Gradient Protein 3 Homolog, plays a role in the development and progression of cancer. This protein is found in the cytoplasm of cells and is secreted outside the cell. AGR3 is involved in promoting the spread of cancer cells (metastasis) and interfering with the function of the p53 protein, a crucial tumor suppressor. Due to its association with hormone-dependent breast cancers, AGR3 shows potential as both a biomarker for disease detection and a target for therapeutic interventions.
This recombinant AGR3 protein is produced in E. coli, resulting in a single polypeptide chain that lacks glycosylation. It consists of 169 amino acids, with the sequence spanning from amino acid 22 to 166. The molecular weight of the protein is 19.5 kDa. For purification purposes, a 20-amino acid His-tag is added to the N-terminus. The protein is then purified using specialized chromatographic techniques.
This AGR3 protein solution has a concentration of 1 mg/ml. It is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 20% glycerol, 0.1 M NaCl, and 1 mM DTT.
For short-term storage of 2-4 weeks, the AGR3 protein solution can be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein such as HSA or BSA to a final concentration of 0.1% is advised for long-term storage. Repeated freezing and thawing of the solution should be avoided.
Anterior gradient protein 3 homolog, AG-3, AG3, hAG-3, Breast cancer membrane protein 11, AGR3, BCMP11, UNQ642/PRO1272, HAG3, PDIA18.
MGSSHHHHHH SSGLVPRGSH MGSMIAIKKE KRPPQTLSRG WGDDITWVQT YEEGLFYAQK SKKPLMVIHH LEDCQYSQAL KKVFAQNEEI QEMAQNKFIM LNLMHETTDK NLSPDGQYVP RIMFVDPSLT VRADIAGRYS NRLYTYEPRD LPLLIENMKK ALRLIQSEL.
AGR3 (also known as AG-3, BCMP11, or hAG-3) is a member of the protein disulfide isomerase (PDI) family that functions as an endoplasmic reticulum (ER)-resident molecular foldase involved in maintaining cellular homeostasis . Like its homologue AGR2, with which it shares 71% sequence identity, AGR3 is encoded at chromosomal position 7p21 .
In normal physiology, AGR3 has been identified as a key regulator in airway epithelium, where it is required for ciliary beat frequency and mucociliary clearance . Its expression pattern is similar to AGR2 in non-pathological tissues, suggesting cognate physiological functions . While AGR3 contains an ER retention signal sequence (QSEL), it has also been detected in extracellular media such as blood and urine, indicating potential functions beyond its intracellular role .
Methodologically, studying AGR3's normal function requires careful consideration of tissue-specific expression patterns and both its intracellular and extracellular roles.
Researchers employ several complementary techniques to detect and quantify AGR3 expression:
Protein Detection Methods:
Immunohistochemistry (IHC): Effective for visualizing AGR3 distribution in tissue samples
Western blotting: Used to measure AGR3 protein levels in tissue extracts
Enzyme-linked immunosorbent assay (ELISA): Enables quantification of AGR3 in serum samples
mRNA Expression Analysis:
Real-time quantitative polymerase chain reaction (RT-qPCR): Allows precise measurement of AGR3 mRNA expression levels
Materials Required for AGR3 RT-qPCR:
Real-Time PCR Detection System
RNA isolation kit (e.g., RNAprep Pure Tissue Kit)
cDNA synthesis kit (e.g., iScript cDNA Synthesis Kit)
qPCR master mix (e.g., SsoFast EvaGreen supermix)
When designing experiments to measure AGR3, researchers should include appropriate controls and consider potential tissue-specific variations in expression. Multi-method approaches combining protein and mRNA analysis provide the most comprehensive assessment of AGR3 expression.
AGR3 is a relatively small protein with the following structural characteristics:
From a methodological perspective, researchers studying AGR3 structure should consider:
X-ray crystallography or cryo-EM for detailed structural analysis
Computational modeling based on homology with AGR2
Analysis of post-translational modifications that may affect function
Investigation of protein-protein interaction domains
The structural analysis of AGR3 provides crucial insights into its functional capabilities and potential interaction partners in both normal physiology and disease states.
AGR3 has emerged as a significant factor in chronic obstructive pulmonary disease (COPD), particularly in the context of exacerbation frequency. Research has revealed several key aspects of AGR3's role in respiratory pathology:
Recent studies have demonstrated that AGR3 protein expression is decreased in patients with frequent COPD exacerbations compared to those with infrequent exacerbations . This finding suggests that AGR3 may play a protective role in the respiratory epithelium that becomes compromised in severe COPD.
Mechanistically, AGR3 regulates airway epithelial junctions, and its loss may contribute to the deterioration of epithelial integrity . This reduced barrier function could facilitate trans-epithelial permeability of pathogens in patients with frequent exacerbations, explaining the increased susceptibility to respiratory infections .
Methodological approach for studying AGR3 in COPD:
Collection of human lung tissues from:
Current-smoking patients without COPD (Control)
Patients with infrequent COPD exacerbations (IFCOPD)
Patients with frequent COPD exacerbations (FCOPD)
Analysis of:
AGR3 protein expression via immunohistochemistry and western blotting
AGR3 mRNA expression via RT-qPCR
Assessment of adherent junctions (AJs) and tight junctions (TJs) protein expression
In vitro studies using BEAS-2B cells exposed to cigarette smoke extract (CSE) to examine:
This methodological framework allows researchers to establish both correlative and causative relationships between AGR3 expression and epithelial junction integrity in COPD.
While AGR3 has traditionally been considered an intracellular protein resident in the endoplasmic reticulum, research has revealed significant extracellular functions that impact cancer progression:
Extracellular AGR3 (eAGR3) has been identified as a microenvironmental signaling molecule in tumor-associated processes . In breast cancer, eAGR3 regulates cancer cell migration via Src signaling pathways . This suggests that AGR3 not only functions within cancer cells but also participates in cell-cell communication within the tumor microenvironment.
The secretion of AGR3 appears to be a regulated process despite the presence of an ER retention signal (QSEL), as it has been detected in extracellular media including gastrointestinal mucus, blood, and urine . This suggests specific export mechanisms that might be upregulated in cancer cells.
Experimental approach for studying extracellular AGR3:
Collection and analysis of conditioned media from cancer cell lines
Protein purification and characterization of extracellular AGR3
Treatment of cancer cells with recombinant AGR3 protein to assess:
Migration capacity (wound healing assays)
Invasion capability (Boyden chamber assays)
Signal transduction activation (phosphorylation of Src and downstream targets)
Neutralization experiments using anti-AGR3 antibodies to block extracellular functions
This research direction reveals AGR3 as a potential therapeutic target not only within cancer cells but also in the tumor microenvironment. Blocking extracellular AGR3 functions could represent a novel strategy for inhibiting cancer progression and metastasis.
The genomic and transcriptomic profiles of AGR3 in cancer reveal important insights into its role in tumorigenesis:
Genomic Alterations:
Analysis of multiple cancer databases (including NCI, CCLE, and TCGA) has shown that:
No functional polymorphisms or recurrent mutations have been detected in the AGR3 gene
Copy number variations (CNVs) may occur but are not the primary mechanism of AGR3 dysregulation in cancer
Transcriptomic Features:
AGR3 expression is positively correlated with epithelial gene expression and inversely correlated with mesenchymal gene expression, suggesting involvement in epithelial-mesenchymal transition (EMT)
AGR3 expression is significantly associated with several cancer features, including TP53 or SMAD4 mutations, with relationships varying depending on cancer type
Functional Impact Assessment:
CRISPR gene extinction screens (Achilles project) revealed that AGR3 extinction does not significantly modify cell fitness, contrasting with the effects observed for oncogenes (decreased fitness) and tumor suppressor genes (increased fitness)
This suggests that AGR3 functions as a non-genetic evolutionary factor in human tumorigenesis rather than a classical oncogene or tumor suppressor
From a methodological perspective, integrative analysis combining genomic, transcriptomic, and functional data provides the most comprehensive understanding of AGR3's role in cancer. Researchers should employ multi-omics approaches to fully characterize AGR3 alterations in specific cancer types.
AGR2 and AGR3 are homologous proteins with 71% sequence identity that are encoded adjacently at chromosomal position 7p21 . Despite their structural similarities, they exhibit both overlapping and distinct functions in cancer progression:
Similarities:
Both proteins are PDI family members functioning as ER-resident molecular foldases
Both show similar expression patterns in normal and carcinomatous tissues
Both have been associated with oestrogen receptor-positive breast tumors
Both can be detected extracellularly despite having ER retention signals (KTEL for AGR2, QSEL for AGR3)
Differences:
AGR2 has been more extensively characterized as a pro-oncogenic protein associated with cancer aggressiveness and poor prognosis
AGR2 is regulated by the ER stress response and is involved in mucin production in intestinal, pulmonary, and pancreatic tissues
AGR2 and AGR3 may interact with different partner proteins, contributing to distinct functions
Methodological considerations for comparative studies:
Parallel knockdown and overexpression experiments of AGR2 and AGR3 in the same cell lines
Co-immunoprecipitation studies to identify shared and unique protein interaction partners
Dual immunohistochemistry in patient samples to assess co-expression patterns
Analysis of prognostic significance of AGR2/AGR3 ratios rather than absolute expression levels
Understanding the functional relationship between AGR2 and AGR3 may provide insights into their complementary or compensatory roles in cancer progression, potentially improving their utility as biomarkers or therapeutic targets.
For researchers working with recombinant AGR3 protein, optimal production and storage conditions are crucial for maintaining protein integrity and functionality:
Production Specifications:
Expression Host: HEK293 cells provide proper folding and post-translational modifications for human AGR3
Protein Sequence: Ile22-Leu166 (mature protein without signal peptide)
Purification Method: Affinity chromatography followed by size exclusion chromatography
Quality Control: >95% purity as determined by reducing SDS-PAGE
Storage and Handling Guidelines:
Long-term Storage: Lyophilized protein is stable for up to 12 months at -20 to -80°C
Reconstitution Buffer: 20mM Glycine-HCl, 10% Trehalose, 0.05% Tween 80, pH 3.5
Short-term Storage: Reconstituted protein can be stored at 4-8°C for 2-7 days
Aliquoting: Reconstituted samples should be divided into single-use aliquots and stored at < -20°C, stable for 3 months
Freeze-Thaw Cycles: Should be minimized to prevent protein degradation
Methodological considerations for experimental use:
Perform activity assays to confirm functionality after reconstitution
Include appropriate controls when using recombinant AGR3 in cell culture experiments
Consider the impact of the C-terminal His tag on protein function
For extracellular applications, ensure physiologically relevant concentrations
Proper production, handling, and storage of recombinant AGR3 protein are essential for obtaining reliable and reproducible experimental results.
AGR3 has demonstrated potential as a prognostic biomarker in several cancer types, with particular emphasis on ovarian carcinomas:
AGR3 can serve as a prognostic marker for survival in patients with both low-grade and high-grade serous ovarian carcinomas . The prognostic value of AGR3 appears to be related to its role in cancer cell biology and tumor microenvironment regulation.
For breast cancer, AGR3 is associated with estrogen receptor-positive tumors and may interact with metastasis-related genes, suggesting potential utility in predicting disease progression .
Methodological framework for biomarker validation:
Retrospective analysis:
Tissue microarray analysis of AGR3 expression in tumor samples
Correlation with clinical parameters and survival outcomes
Multivariate analysis to assess independent prognostic value
Liquid biopsy approach:
Integration with other biomarkers:
Assessment of AGR3 in combination with established cancer biomarkers
Development of prognostic scoring systems incorporating AGR3 status
For optimal clinical utility, standardized methods for AGR3 detection should be established, including validated antibodies for immunohistochemistry and calibrated ELISA kits for serum quantification.
Understanding the correlation between AGR3 expression and therapeutic response could guide personalized treatment approaches:
While direct data on AGR3 and treatment response is limited in the provided search results, insights can be drawn from its biological functions and related studies:
AGR3's extracellular signaling role via Src pathways suggests potential influence on response to targeted therapies, particularly those affecting growth factor receptor signaling . Its membership in the PDI family implies potential involvement in ER stress responses, which are known to affect chemotherapy resistance mechanisms.
Research methodology for investigating AGR3 in therapy response:
In vitro assessment:
Compare drug sensitivity profiles in cell lines with AGR3 knockdown or overexpression
Evaluate the impact of extracellular AGR3 on drug efficacy
Analyze changes in AGR3 expression following exposure to various therapeutic agents
Clinical correlation studies:
Retrospective analysis of AGR3 expression in responders versus non-responders
Longitudinal monitoring of AGR3 levels during treatment
Evaluation of AGR3 as a predictive biomarker for specific therapeutic modalities
Mechanistic investigations:
Assess the role of AGR3 in therapy-induced stress responses
Investigate AGR3-mediated signaling pathways that could influence drug resistance
Explore combination approaches targeting AGR3 alongside standard therapies
This research direction could potentially identify AGR3 as a predictive biomarker for therapy selection and a target for overcoming resistance mechanisms in cancer treatment.
Based on current understanding of AGR3 biology, several approaches show promise for therapeutic targeting:
Potential targeting strategies:
Antibody-based approaches:
Neutralizing antibodies against extracellular AGR3
Antibody-drug conjugates for targeted delivery to AGR3-expressing cells
Small molecule inhibitors:
Compounds disrupting AGR3 protein-protein interactions
Inhibitors of AGR3 secretion pathways
Gene therapy approaches:
siRNA or CRISPR-based knockdown of AGR3 in disease contexts
Modulation of AGR3 expression via epigenetic targeting
Peptide inhibitors:
Competitive inhibitors derived from AGR3 binding partners
Cell-penetrating peptides targeting intracellular AGR3 functions
Methodological considerations for therapeutic development:
Target validation:
Detailed characterization of AGR3 roles in specific disease contexts
Identification of patient populations most likely to benefit
Assay development:
High-throughput screening systems for inhibitor discovery
Relevant in vitro and in vivo models for efficacy testing
Delivery optimization:
Strategies for targeting extracellular versus intracellular AGR3
Tissue-specific delivery approaches
The development of AGR3-targeted therapeutics represents an emerging opportunity in precision medicine, particularly for cancers and respiratory diseases where AGR3 dysregulation has been established.
Systems biology offers powerful frameworks for integrating diverse data types to understand AGR3's role in disease:
Integrative approaches:
Multi-omics integration:
Combining genomic, transcriptomic, proteomic, and metabolomic data
Network analysis to position AGR3 within disease-relevant pathways
Identification of hub proteins that interact with AGR3
Computational modeling:
Predictive models of AGR3 regulation and function
Simulation of perturbation effects in disease networks
Virtual screening for potential AGR3 modulators
Single-cell analysis:
Characterization of AGR3 expression at single-cell resolution
Spatial transcriptomics to map AGR3 expression in tissue microenvironments
Cell-cell communication networks involving AGR3
Methodological framework:
Data collection and integration:
Standardized protocols for multi-omics data generation
Quality control procedures for heterogeneous data types
Computational pipelines for data normalization and integration
Network construction and analysis:
Protein-protein interaction networks centered on AGR3
Gene regulatory networks controlling AGR3 expression
Pathway enrichment analysis to identify biological processes
Validation and refinement:
Experimental testing of computationally derived hypotheses
Iterative model improvement based on experimental feedback
Translation of network insights into therapeutically relevant targets
Systems biology approaches provide a comprehensive framework for understanding AGR3's complex role in health and disease, potentially revealing unexpected connections and therapeutic opportunities.
AGR3 is characterized by:
This protein is predominantly expressed in ciliated airway epithelial cells and plays a crucial role in regulating ciliary beat frequency and mucociliary clearance in the airway . In mice, AGR3 is involved in the regulation of intracellular calcium in tracheal epithelial cells .
AGR3 has several important biological functions: