Human ANXA2 is a 36-kDa protein encoded by the ANXA2 gene, with an expected mass of 38.6 kDa and two reported isoforms . The protein can exist in multiple configurations in vivo, including as a monomer, heterodimer, or heterotetramer . ANXA2 has four forms: secretory, membrane-bound, cytoplasmic, and nuclear .
ANXA2 is also known by several alternative names in the literature:
Methodologically, researchers typically identify ANXA2 through Western blotting, ELISA, immunohistochemistry, or immunofluorescence using specific anti-ANXA2 antibodies. Currently, there are over 617 commercial ANXA2 antibodies available across 34 suppliers, indicating the protein's significance in research .
ANXA2 is expressed on the surface of multiple cell types, including:
Within cells, ANXA2 can be found in multiple compartments, reflecting its diverse functions. The four main localization patterns include:
Localization | Function | Detection Method |
---|---|---|
Secretory | Extracellular signaling | ELISA of culture media or body fluids |
Membrane-bound | Cell surface activities | Flow cytometry, membrane fractionation |
Cytoplasmic | Intracellular signaling | Immunocytochemistry, cell fractionation |
Nuclear | Gene regulation | Nuclear fractionation, ChIP assays |
For accurate localization studies, researchers should employ subcellular fractionation followed by Western blotting or immunofluorescence with compartment-specific markers to confirm ANXA2 distribution patterns .
ANXA2 orchestrates multiple cellular functions in normal human physiology:
Vesicle transport - regulates endocytosis and exocytosis
Cell division - contributes to cytokinesis processes
Calcium signaling - responds to calcium fluctuations
At the endothelial cell surface, ANXA2 forms a heterotetramer with S100A10 (p11), which accelerates tissue plasminogen activator-dependent activation of the fibrinolytic protease plasmin, playing a crucial role in vascular homeostasis and fibrinolysis .
Methodologically, researchers can study these functions through:
Calcium binding assays to assess ANXA2 activation
Co-immunoprecipitation to identify binding partners
RNA interference to evaluate loss-of-function effects
Plasmin generation assays to measure fibrinolytic activity
ANXA2 functions are regulated by various posttranslational modifications, making it a dynamic responder to cellular conditions .
ANXA2 expression demonstrates significant correlation with glioma progression:
This correlation has been validated in multiple large datasets:
The Cancer Genome Atlas (TCGA) database (699 glioma samples)
Chinese Glioma Genome Atlas (CGGA) database (325 glioma samples)
Methodologically, multivariate Cox regression analysis confirms ANXA2 as an independent risk factor for poor clinical prognosis in glioma patients, suggesting its value as a negative prognostic marker that could be incorporated into molecular diagnostic panels .
ANXA2 exhibits significant connections to immune responses in the glioma microenvironment:
Immune cell correlation: ANXA2 expression positively correlates with biomarker genes of six key immune cell types in glioma, with particularly strong associations with immunosuppressive cells:
Immunosuppression mechanisms: ANXA2 contributes to glioma immune escape through:
T cell inhibition: ANXA2 regulates the actin cytoskeleton, which negatively affects T cell aggregation, potentially reducing T cell activation and creating immune imbalance in the tumor microenvironment .
Methodologically, researchers can investigate these immune interactions using:
Single-cell RNA sequencing of tumor samples
Immune cell co-culture systems with ANXA2 manipulation
Flow cytometric analysis of immune cell populations
Spatial transcriptomics to map ANXA2 and immune cell distributions
These findings suggest ANXA2-targeted immunotherapy could potentially reduce immunosuppression in glioma and improve patient outcomes .
ANXA2 demonstrates important functions in hypoxia response and neovascularization:
Hypoxic regulation: ANXA2 is upregulated under hypoxic conditions, suggesting its role as part of the cellular adaptation to low oxygen environments .
Vascular impact: Evidence from animal models shows that:
Clinical implications: These findings suggest ANXA2 may be involved in human retinal vascular proliferation disorders and other pathological angiogenesis conditions .
Methodologically, researchers studying ANXA2 in hypoxia and angiogenesis should consider:
Hypoxic chamber experiments with ANXA2 knockdown/overexpression
Endothelial tube formation assays to assess angiogenic potential
In vivo models using ANXA2-deficient animals
Analysis of hypoxia-inducible factor (HIF) binding sites in the ANXA2 promoter
Understanding these mechanisms could lead to novel anti-angiogenic therapeutic approaches for conditions characterized by pathological vessel growth .
ANXA2 undergoes various post-translational modifications that significantly influence its functions. Research approaches should include:
Identification methods:
Mass spectrometry to map specific modification sites
Phospho-specific or other modification-specific antibodies
2D gel electrophoresis to separate modified forms
Functional analysis:
Site-directed mutagenesis of modification sites
Pharmacological inhibitors of specific modifying enzymes
In vitro modification assays to assess catalytic changes
Cellular impact assessment:
Microscopy to track localization changes of modified ANXA2
Co-immunoprecipitation to identify altered protein interactions
Activity assays (e.g., plasmin generation) to measure functional consequences
Modification Type | Common Sites | Detection Method | Functional Impact |
---|---|---|---|
Phosphorylation | Tyr23, Ser25 | Phospho-specific antibodies | Membrane translocation |
Acetylation | N-terminal | Mass spectrometry | Secretion regulation |
S-glutathionylation | Cys residues | Redox proteomics | Oxidative stress response |
Ubiquitination | Lys residues | Ubiquitin pull-down | Protein turnover |
Many ANXA2 functions are regulated by these modifications, making them critical targets for understanding ANXA2's role in both normal physiology and disease states .
Studying the ANXA2-S100A10 heterotetramer (A2- p11) requires specialized approaches:
Complex isolation and characterization:
Co-immunoprecipitation with either ANXA2 or S100A10 antibodies
Size exclusion chromatography to isolate the heterotetrameric complex
Cross-linking mass spectrometry to map interaction interfaces
Surface plasmon resonance to measure binding kinetics
Functional analysis:
Plasmin generation assays to measure fibrinolytic activity
Mutagenesis of key binding residues to disrupt complex formation
Competitive peptides to inhibit complex assembly
Cell-based assays in endothelial cells where the complex naturally functions
Visualization techniques:
Proximity ligation assays to visualize interactions in situ
FRET/BRET to measure real-time complex dynamics
Super-resolution microscopy to localize the complex at the cell surface
This complex accelerates tissue plasminogen activator-dependent activation of plasmin, playing crucial roles in fibrinolysis and vascular function . Understanding the heterotetramer's assembly, regulation, and activity is essential for developing potential therapeutic interventions for thrombotic or hemorrhagic disorders.
For robust analysis of ANXA2 in patient samples, researchers should employ:
Multi-omic approaches:
RNA-seq for transcriptional analysis
Proteomics for protein level quantification
Integrate with genomic data (mutations, CNVs)
Single-cell approaches for cellular heterogeneity assessment
Statistical methods for clinical correlation:
Kaplan-Meier survival analysis stratified by ANXA2 expression
Cox regression to determine independent prognostic value
Correlation analysis with established biomarkers
Machine learning algorithms for pattern recognition
Validation strategies:
When analyzing expression patterns in glioma, researchers should consider:
Stratification by WHO grade (II-IV)
IDH mutation status
Molecular subtypes (especially mesenchymal)
Patient demographics and treatment history
ANXA2 presents several promising avenues for therapeutic targeting:
Target rationale:
Potential therapeutic approaches:
Monoclonal antibodies against cell-surface ANXA2
Small molecule inhibitors of ANXA2-protein interactions
Peptide-based disruptors of ANXA2-S100A10 complex
ANXA2-targeted immunotherapy approaches
Combination strategies:
ANXA2 inhibition + conventional chemotherapy
ANXA2 inhibition + immune checkpoint blockade
ANXA2 inhibition + anti-angiogenic therapy
ANXA2 inhibition + radiation therapy
ANXA2 demonstrates significant potential as a clinical biomarker:
ANXA2's use as a biomarker has precedent in multiple cancers. For example, it has been used as an immunosensor for lung cancer detection and for early diagnosis of liver cancer via ELISA . Implementation in clinical practice would require standardized detection methods and establishment of validated cutoff values for high vs. low expression.
ANXA2 plays significant roles in coagulation and fibrinolysis across multiple conditions:
Thrombotic conditions:
Hemorrhagic conditions:
Mechanistic basis:
Methodologically, researchers investigating ANXA2 in coagulation should consider:
Plasmin generation assays
Clot lysis time measurements
Thromboelastography
In vivo thrombosis models with ANXA2 manipulation
Analysis of ANXA2 autoantibodies in patient plasma
Understanding these mechanisms could lead to novel diagnostic and therapeutic approaches for both thrombotic and hemorrhagic disorders .
ANXA2 has been implicated in multiple therapy resistance pathways:
Resistance mechanisms:
Research approaches:
Drug-resistant cell line models with ANXA2 modulation
Patient-derived xenografts from treatment-resistant tumors
Temporal analysis of ANXA2 expression during treatment courses
Combination therapy testing with ANXA2 inhibition
Clinical correlations:
Analysis of pre- and post-treatment ANXA2 expression
Correlation of ANXA2 levels with time to recurrence
Examination of ANXA2 in matched primary and recurrent tumors
Patient stratification based on ANXA2 status for therapy selection
Understanding ANXA2's role in therapy resistance could potentially lead to strategies that re-sensitize tumors to existing therapies, particularly in aggressive cancers like glioblastoma where treatment options remain limited .
ANXA2 coordinates complex interactions within the tumor microenvironment:
Immune cell interactions:
Strong correlations with tumor-associated macrophages (r = 0.74 in TCGA, r = 0.65 in CGGA)
Associations with regulatory T cells (r = 0.63 in TCGA, r = 0.55 in CGGA)
Relationships with myeloid-derived suppressor cells (r = 0.68 in TCGA, r = 0.29 in CGGA)
Interaction with DC-SIGN (CD209) leading to immunosuppression
Extracellular matrix remodeling:
ANXA2's role in plasmin activation influences ECM degradation
May facilitate cancer cell invasion and migration
Potentially contributes to pre-metastatic niche formation
Vascular interactions:
Research methodologies to investigate these interactions include:
3D co-culture systems with multiple cell types
Extracellular vesicle isolation and characterization
Spatial transcriptomics/proteomics of tumor sections
Intravital microscopy to observe real-time interactions
These microenvironment interactions make ANXA2 a potential master regulator of tumor ecology, suggesting that targeting ANXA2 could disrupt multiple tumor-supporting networks simultaneously .
The ANXA2 gene is located on chromosome 15q22.2 and has three pseudogenes on chromosomes 4, 9, and 10 . Multiple alternatively spliced transcript variants encoding different isoforms have been identified for this gene . Annexin A2 can exist as a monomer or as part of a heterotetramer complex, often interacting with other proteins such as S100A10 .
Annexin A2 is involved in several key cellular functions:
Annexin A2 has been implicated in various pathological conditions: