AZGP1 stimulates lipid degradation in adipocytes, contributing to fat loss in cancer cachexia . It binds polyunsaturated fatty acids and activates β3-adrenergic receptors to promote lipolysis .
In kidney and heart tissues, AZGP1 inhibits TGF-β signaling, reducing fibrosis by:
Prognostic Biomarker: Low AZGP1 expression correlates with poor survival in gastric adenocarcinoma (HR = 1.68, P = 0.011) .
Tumor Suppression: Downregulation in cancer tissues (52.8% of gastric cancers) associates with advanced T stage (P = 0.008) and poor differentiation (P = 0.005) .
Serum AZGP1 inversely correlates with pulse wave velocity (P < 0.05), indicating a protective role against arterial stiffness post-kidney transplantation .
Levels decline with improved renal function (eGFR correlation: r = -0.45, P = 0.002) .
AZGP1 deficiency exacerbates renal fibrosis in murine models (P < 0.05 for collagen deposition) .
Recombinant AZGP1 rescues fibrotic phenotypes, suggesting therapeutic potential .
AZGP1 inhibits amine oxidase copper-containing 3 (AOC3), reducing oxidative stress in adipocytes and endothelial cells .
While Sf9 cells (insect cell line) are not referenced in the provided data, recombinant AZGP1 is typically produced in:
Parameter | E. coli-Expressed AZGP1 | Mammalian-Expressed AZGP1 |
---|---|---|
Glycosylation | No | Yes (18.2% carbohydrate) |
Purity | >95% by chromatography | Varies by protocol |
Functional Assays | Lipid degradation | TGF-β inhibition |
Alpha-2-Glycoprotein 1, Zinc-Binding, Zinc-Alpha-2-Glycoprotein, Zn-Alpha-2-Glycoprotein, Zn-Alpha-2-GP, ZAG, Testicular Tissue Protein Li 227, Alpha-2-Glycoprotein 1, Zinc, Alpha-2-Glycoprotein, Zinc, Zn-Alpha2-Glycoprotein, ZNGP1, ZA2G.
Sf9, Baculovirus cells.
QENQDGRYSL TYIYTGLSKH VEDVPAFQAL GSLNDLQFFR YNSKDRKSQP MGLWRQVEGM EDWKQDSQLQ KAREDIFMET LKDIVEYYND SNGSHVLQGR FGCEIENNRS SGAFWKYYYD GKDYIEFNKE IPAWVPFDPA AQITKQKWEA EPVYVQRAKA YLEEECPATL RKYLKYSKNI LDRQDPPSVV VTSHQAPGEK KKLKCLAYDF YPGKIDVHWT RAGEVQEPEL RGDVLHNGNG TYQSWVVVAV PPQDTAPYSC HVQHSSLAQP LVVPWEASLE HHHHHH.
AZGP1 functions as an important inhibitor of amine oxidase copper-containing 3 (AOC3), acting through a non-competitive, allosteric mechanism. Studies demonstrate that AZGP1 binds to AOC3 at a site distinct from the catalytic center, effectively reducing enzyme activity without competing with substrate molecules . Beyond enzyme inhibition, AZGP1 has been identified as a tumor suppressor in several cancer types, including pancreatic cancer where it induces mesenchymal-to-epithelial transition . It is also significantly elevated in the plasma of cancer patients experiencing progressive weight loss (cachexia), particularly in gastrointestinal, breast, and prostate malignancies . This association with cachexia suggests AZGP1 plays roles in metabolism and energy expenditure regulation, potentially contributing to the wasting syndrome observed in these devastating conditions.
Validation of AZGP1 expression in Sf9 cells requires multiple complementary approaches. Western blotting using specific anti-AZGP1 antibodies represents the primary validation method, where properly expressed human AZGP1 should appear at approximately 41 kDa, though this may vary depending on post-translational modifications . For secreted variants, analyze both cell lysates and culture medium. Additionally, immunofluorescence microscopy can confirm subcellular localization, with AZGP1 expected to show membrane-associated localization as observed in human tissue samples . Functional validation through activity assays measuring inhibition of AOC3 provides crucial confirmation that the expressed protein maintains biological activity. GST-pulldown experiments, as described in the literature, can verify that recombinant AZGP1 maintains binding capacity to its partners like AOC3 . For definitive structural validation, differential scanning fluorimetry (DSF) using label-free nanoDSF technology can assess proper protein folding by measuring thermal denaturation profiles .
When investigating AZGP1 in cell culture, several essential controls must be incorporated:
Expression controls: Include both empty vector-transfected cells and untransfected cells to account for transfection-related effects and background expression.
Activity verification: For inhibition studies, include the established AOC3 inhibitor LJP1586 as a positive control, as demonstrated in previous research .
Glycosylation controls: Since insect cells produce different glycosylation patterns than human cells, include treatments with glycosidases (PNGase F or Endo H) to assess the impact of glycosylation on function.
Localization controls: For immunofluorescence, include markers for relevant cellular compartments (membrane, secretory pathway) to confirm proper trafficking.
Tissue-derived AZGP1: When possible, compare with AZGP1 derived from human tissues or mammalian expression systems to identify any functional differences.
These controls ensure that observed effects are specifically attributable to AZGP1 and not artifacts of the expression system or experimental conditions.
AZGP1 expressed in Sf9 insect cells exhibits significant differences in post-translational modifications compared to native human protein, potentially affecting research interpretations. The most notable difference involves glycosylation patterns—Sf9 cells produce primarily high-mannose and paucimannose-type N-glycans lacking the complex terminal sialylation found in human glycoproteins . This glycosylation divergence can affect multiple aspects of AZGP1 function, including:
Protein-protein interactions: The altered glycan structure may modify AZGP1's binding affinity to AOC3, potentially affecting the observed Ki values or kinetics of inhibition .
Protein stability: Different glycosylation patterns can influence thermal stability and proteolytic resistance, requiring careful interpretation of degradation studies.
Allosteric modulation: Since AZGP1 functions as an allosteric inhibitor of AOC3, changes in glycosylation may alter the conformational changes transmitted to the active site.
Optimizing AZGP1 expression in Sf9 cells requires addressing multiple parameters to enhance both yield and functional quality:
Vector design optimization:
Incorporate strong promoters specifically suited for insect cell expression (polyhedrin or p10)
Include efficient secretion signals (native AZGP1 signal or insect-optimized signals)
Consider codon optimization for insect cell preference
Strategic placement of purification tags to minimize interference with functional domains
Expression condition optimization:
Determine optimal multiplicity of infection (MOI) through systematic testing
Fine-tune harvest timing (typically 48-72 hours post-infection)
Evaluate temperature modulation during expression phase (lower temperatures may improve folding)
Supplementation with chemical chaperones to enhance proper folding
Purification strategy development:
Functional validation methods:
By systematically addressing these parameters, researchers can significantly improve both yield and functional quality of Sf9-expressed AZGP1 for downstream applications.
Recent research presents an intriguing contradiction: computational docking predicted binding between AZGP1 and lenvatinib with a binding free energy of -1.65, while differential scanning fluorimetry (DSF) assays showed no direct interaction . This discrepancy requires methodical investigation through complementary approaches:
Advanced biophysical interaction analyses:
Surface Plasmon Resonance (SPR) to detect transient or weak interactions through real-time binding kinetics
Microscale Thermophoresis (MST) for detection of subtle binding events in near-native conditions
Isothermal Titration Calorimetry (ITC) for direct thermodynamic characterization of binding
NMR spectroscopy to map potential binding interfaces through chemical shift perturbations
Computational refinement strategies:
Molecular dynamics simulations to assess stability of predicted binding conformations
Ensemble docking approaches considering protein flexibility
More rigorous binding free energy calculations with explicit solvent models
Identification of potential water-mediated interactions not captured in initial docking
Mechanistic investigations:
Evaluation of lenvatinib's effects on AZGP1 expression through epigenetic mechanisms, as suggested by findings that lenvatinib promotes H3K27Ac enrichment in the AZGP1 promoter region
Assessment of potential intermediate proteins that might bridge lenvatinib and AZGP1 interaction
Cellular localization studies to determine if lenvatinib affects AZGP1 trafficking or compartmentalization
This systematic approach would clarify whether the interaction is direct but below detection thresholds of DSF, indirect through intermediate molecules, or primarily mediated through transcriptional regulation rather than physical binding.
Differential scanning fluorimetry (DSF) using label-free nanoDSF technology provides valuable insights into AZGP1 stability and interactions. Based on published protocols, the optimal parameters include:
Sample preparation:
Instrument settings:
Temperature range: 20-95°C
Scan rate: 1°C/min for optimal resolution
Excitation wavelength: 280 nm (for intrinsic tryptophan/tyrosine fluorescence)
Dual emission monitoring: 330 nm and 350 nm to calculate ratio changes
Controls and validation:
Data interpretation guidelines:
Calculate first derivative of fluorescence ratio to determine melting temperature (Tm)
Significant Tm shifts (>0.5°C) generally indicate binding
For negative results (as with lenvatinib-AZGP1), validate with orthogonal techniques
Consider concentration-dependent experiments to determine binding affinity
When properly implemented, this approach provides robust data on AZGP1 thermal stability changes upon ligand binding, though researchers should remain aware that some interaction types might not significantly affect thermal stability profiles.
Accurate quantification of AZGP1 in clinical samples requires complementary approaches to address the complexity of tumor heterogeneity:
RT-qPCR for transcript quantification:
Extract RNA using methods optimized for FFPE or fresh tissue
Implement rigorous quality control for RNA integrity
Select appropriate reference genes validated for stability in the specific cancer type
Apply the ΔΔCt method with multiple reference genes for normalization
Include positive controls from tissues known to express AZGP1
Immunohistochemistry for protein detection and localization:
Use validated antibodies with confirmed specificity
Implement semi-quantitative scoring by multiple independent pathologists
Score based on staining intensity and percentage of positive cells
Include adjacent normal tissue as internal control
Consider automated image analysis for objective quantification
Western blotting for protein quantification:
Extract proteins using buffers containing protease inhibitors
Include recombinant standards for absolute quantification
Normalize to housekeeping proteins or total protein stains
Verify antibody specificity with appropriate controls
Multiplexed immunofluorescence:
Designing experiments to accurately measure AZGP1's inhibition of AOC3 requires careful consideration of multiple factors:
Enzyme source selection:
Activity assay optimization:
For cell-based studies, radioactive assays eliminate non-specific background signals
Include selective AOC3 inhibitor LJP1586 as positive control
Implement dose-response curves with multiple AZGP1 concentrations
Account for species differences (higher concentrations needed for human AOC3 inhibition)
Kinetic analysis approaches:
Controls and normalization:
Data interpretation guidelines:
Plot dose-dependent inhibition curves
Differentiate between direct and allosteric inhibition mechanisms
Consider species-specific differences in inhibition potency
Correlate in vitro findings with physiological AZGP1 concentrations
This experimental design captures the allosteric inhibitory mechanism of AZGP1 on AOC3, providing quantitative measurements that can inform both basic understanding and therapeutic development.
AZGP1 expression shows significant correlations with patient survival across various cancer types, with particularly well-documented prognostic value in intrahepatic cholangiocarcinoma (ICC):
These findings position AZGP1 as a valuable prognostic biomarker, particularly in ICC where low expression predicts significantly worse survival outcomes. Importantly, the correlation remains significant even after adjusting for other clinical and pathological variables, confirming its independent prognostic value .
Lenvatinib increases AZGP1 expression through an epigenetic mechanism involving histone acetylation, providing insight into a novel aspect of this drug's anticancer activity:
This epigenetic regulation pathway presents several potential therapeutic implications, including the possibility of using AZGP1 expression as a biomarker for lenvatinib response and developing combination therapies targeting both histone acetylation and AZGP1-related pathways to enhance efficacy.
The AZGP1-AOC3 interaction presents a promising target for therapeutic development in both cachexia and cancer, with several potential approaches:
Therapeutic approaches for cachexia:
Development of AZGP1 mimetics that retain AOC3 inhibitory capacity without pro-lipolytic effects
AOC3 inhibitors as alternative approach when AZGP1 levels are insufficient
Combined interventions targeting both the AZGP1-AOC3 axis and other pathways involved in cachexia
Recombinant AZGP1 administration in cachexia models to evaluate efficacy
Cancer therapeutic strategies:
Epigenetic therapies to upregulate AZGP1 expression, following the model of lenvatinib's action
Combination of HDAC inhibitors with agents that induce AZGP1 expression
Development of specific modulators of the AZGP1 promoter region
AOC3 inhibitors as complementary therapy in cancers with low AZGP1 expression
Biomarker applications:
AZGP1 expression levels as predictive markers for response to therapies targeting this pathway
Monitoring AOC3 activity as a surrogate marker for treatment efficacy
H3K27Ac levels at the AZGP1 promoter as a potential predictive biomarker
Combined analysis of AZGP1 expression and metabolic parameters for patient stratification
Mechanistic considerations for drug development:
AZGP1 functions as a non-competitive, allosteric inhibitor of AOC3
This mechanism offers advantages for drug design compared to competitive inhibition
The allosteric binding site could be targeted for development of small molecule mimetics
Understanding the structural basis of the interaction would facilitate rational drug design
The strong correlation between AZGP1 expression and survival outcomes , combined with its mechanistic role in AOC3 inhibition , positions this pathway as a promising therapeutic target with potential applications in multiple cancer types and cancer-associated cachexia.
Several emerging technologies could enhance the structural and functional fidelity of human AZGP1 expressed in Sf9 insect cells:
Glycoengineering approaches:
CRISPR-Cas9 modification of Sf9 glycosylation pathways to humanize glycan structures
Introduction of human glycosyltransferases (sialyltransferases, galactosyltransferases)
Knockout of insect-specific glycosylation enzymes
Development of designer Sf9 cell lines with humanized glycosylation capabilities
Co-expression strategies:
Simultaneous expression of human chaperones to assist proper folding
Co-expression of interaction partners that stabilize native conformations
Integration of human post-translational modification enzymes
Development of polycistronic expression vectors for coordinated expression
Expression condition optimization:
Implementation of temperature cycling to optimize folding kinetics
Development of specialized media formulations to support human-like post-translational modifications
Controlled pH and oxygen tension to mimic human cellular environments
Application of mechanical stress patterns that simulate physiological conditions
Structural validation technologies:
Integration of real-time folding sensors into expression constructs
Application of hydrogen-deuterium exchange mass spectrometry for conformational analysis
Development of conformation-specific antibodies as folding quality markers
Implementation of automated purification coupled with structural screening
These technological advances would address the primary limitations of Sf9 expression systems—particularly glycosylation differences—while maintaining the advantages of high yield and eukaryotic processing capabilities, ultimately producing research-grade AZGP1 with greater structural and functional fidelity to the native human protein.
Despite significant advances in AZGP1 research, several critical questions remain unresolved:
Mechanistic role in cancer progression:
How does AZGP1 function as a tumor suppressor at the molecular level?
What signaling pathways mediate its effects on epithelial-mesenchymal transition?
How does epigenetic regulation of AZGP1 differ across cancer types?
What factors determine whether AZGP1 functions as a tumor suppressor or promotes progression?
Cachexia and metabolic regulation:
Is the elevation of AZGP1 in cachexia a cause or consequence of the wasting syndrome?
How does AZGP1 coordinate with other factors in regulating energy expenditure?
What is the relationship between AZGP1's inhibition of AOC3 and its metabolic effects?
Could AZGP1-based interventions effectively treat cancer cachexia?
AOC3 inhibition consequences:
What are the physiological consequences of AZGP1-mediated AOC3 inhibition in different tissues?
How does this inhibition affect inflammation, vascular adhesion, and immune cell trafficking?
Are the effects of AZGP1 on AOC3 activity tissue-specific or systemic?
What structural features of AZGP1 mediate its allosteric inhibition of AOC3?
Translational research priorities:
Can AZGP1 expression be effectively modulated for therapeutic benefit?
What biomarkers predict response to therapies targeting the AZGP1 pathway?
How might combination therapies targeting AZGP1 and related pathways be optimized?
What patient populations would most benefit from AZGP1-targeted interventions?
Addressing these questions will require integrated approaches combining structural biology, functional genomics, metabolic studies, and clinical investigations to fully understand AZGP1's multifaceted roles and therapeutic potential.
To establish causality between AZGP1 expression and cancer patient survival, a comprehensive experimental approach is required:
Mechanistic studies in cellular and animal models:
CRISPR-Cas9 knockout and knockin models with varying AZGP1 expression levels
Orthotopic tumor models with inducible AZGP1 expression
Patient-derived xenografts stratified by AZGP1 expression
Transgenic models with tissue-specific AZGP1 modulation
Detailed analysis of effects on tumor growth, metastasis, and survival outcomes
Molecular pathway analysis:
Multi-omics profiling (transcriptomics, proteomics, metabolomics) of tumors with varied AZGP1 expression
Network analysis to identify key pathways influenced by AZGP1
Evaluation of AOC3 activity and related inflammatory markers
Examination of metabolic alterations associated with AZGP1 levels
Correlation with epigenetic modifications, particularly H3K27 acetylation
Clinical investigation strategies:
Prospective cohort studies with baseline AZGP1 measurement and long-term follow-up
Development of standardized AZGP1 quantification methods for clinical samples
Integration with existing biobanks and clinical trial datasets
Correlative studies with treatment response and survival
Mendelian randomization studies using AZGP1-associated genetic variants
Therapeutic intervention studies:
Methodological considerations:
Careful control for confounding variables in clinical studies
Standardization of AZGP1 measurement across research groups
Integration of tissue microenvironment analysis
Consideration of temporal changes in AZGP1 expression during disease progression
Development of clinically applicable AZGP1 testing methods
This comprehensive approach would establish whether the observed correlation between AZGP1 expression and survival (as demonstrated in ICC where high expression correlates with significantly better outcomes ) represents a causal relationship, providing foundation for development of AZGP1-targeted therapeutic strategies.
The AZGP1 gene is located on chromosome 7q22.1 in humans . The protein is widely expressed in various tissues and body fluids, including the breast, stomach, liver, prostate, plasma, urine, and saliva . The human recombinant version of this protein, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain containing 286 amino acids (21-298 a.a) and has a molecular mass of 33.2 kDa . It is fused to an 8 amino acid His-tag at the C-terminus and purified by proprietary chromatographic techniques .
Recent studies have investigated the immunological function of AZGP1 in regulating tumor response, particularly in the breast cancer microenvironment . AZGP1 expression has been found to be negatively correlated with multiple immunological processes and specific immune cell infiltration, including macrophages . It is suggested to be a novel immunoregulatory factor affecting the macrophage phenotype in breast cancer tissues .