EYA2 Human, formally known as Eyes Absent Homolog 2, is a recombinant protein encoded by the EYA2 gene and produced in E. coli for laboratory research. It consists of 295 amino acids (residues 244–514) with a molecular mass of 33.2 kDa, fused to an N-terminal His-tag for purification . EYA2 belongs to the HAD-family tyrosine phosphatases and functions as both a transcriptional coactivator and a phosphatase, playing critical roles in DNA repair, organogenesis, and cancer progression .
EYA2 contains two primary functional regions:
N-terminal domain: Exhibits serine/threonine phosphatase activity, implicated in immune regulation .
C-terminal EYA domain (ED): Contains tyrosine phosphatase activity, critical for dephosphorylating histone H2AX at Tyr-142 (H2AXY142ph) to facilitate DNA repair .
Structural analyses reveal that EYA2 interacts with homeobox transcription factors (e.g., SIX1–SIX5) to form transcriptional complexes essential for organ development and cancer metastasis .
EYA2 promotes the recruitment of DNA repair complexes (e.g., MDC1) by dephosphorylating H2AXY142ph, a key step in distinguishing repair pathways from apoptotic responses .
As a coactivator for SIX proteins, EYA2 enhances the transcription of genes involved in cell proliferation (e.g., cyclin D1, cyclin E) and survival .
EYA2 is overexpressed in multiple malignancies and drives oncogenic processes:
EYA2 expression correlates with tumor aggressiveness:
Small-molecule inhibitors: Blocking EYA2’s phosphatase activity disrupts cancer cell proliferation .
Epigenetic modulation: BRD4 inhibitors (e.g., JQ1) reduce EYA2 expression in glioblastoma stem cells .
EYA2 Human is utilized in:
EYA2 contains two primary functional domains: a highly conserved C-terminal Eya Domain (ED) that shares 83-89% sequence identity with other human Eya family members, and a less conserved N-terminal region. The ED functions both as a protein interaction domain that binds to Six family proteins and as a phosphatase domain with catalytic activity. Unlike most cellular tyrosine phosphatases that use cysteine as their active site residue, EYA2 belongs to the haloacid dehalogenase (HAD) hydrolase family and utilizes aspartate as its active site residue .
The crystal structure of the EYA2 ED in complex with Six1 has been determined, revealing the molecular details of this interaction and providing insights into how mutations in this domain can lead to developmental disorders like branchio-oto-renal syndrome . The structural analysis confirms that EYA2 functions as a HAD family phosphatase, yet uniquely targets tyrosine residues rather than serine/threonine residues typically targeted by other HAD phosphatases .
To investigate EYA2's dual functionality as both a phosphatase and transcriptional co-activator, researchers employ multiple complementary approaches:
Phosphatase activity assays: Researchers can measure EYA2's ability to dephosphorylate known substrates such as histone H2AX at Y142 or estrogen receptor β at Y36 . Site-directed mutagenesis of the catalytic aspartate residue can create phosphatase-dead mutants to distinguish phosphatase-dependent from phosphatase-independent functions.
Transcriptional activity assays: Co-transfection of EYA2 with Six1 followed by reporter gene assays can demonstrate EYA2's role as a transcriptional co-activator .
Protein-protein interaction studies: Immunoprecipitation experiments have confirmed the physical interaction between EYA2 and Six1 proteins in cancer cell lines like U251, which exhibit high expression of both proteins . This methodology helps elucidate how EYA2 forms functional complexes with transcription factors.
Functional domain separation: Expression of truncated versions of EYA2 containing either the N-terminal transactivation domain or the C-terminal phosphatase domain allows researchers to dissect which cellular functions depend on each activity .
EYA2 expression follows distinct patterns in normal development versus cancer contexts:
In normal tissues, EYA2 and other EYA family members are typically expressed during early embryogenesis and are essential for the development of numerous organs . After organ development is complete, EYA2 expression is normally downregulated . Immunohistochemical analysis shows negative or low expression of EYA2 in normal glial cells of adult brain tissue .
In cancer, EYA2 becomes re-expressed in multiple tumor types, including astrocytoma, ovarian cancer, breast cancer, glioblastomas, leukemia, and Wilms' tumor . In astrocytoma specifically, EYA2 overexpression was observed in 36.7% (33/90) of specimens examined, with expression rates increasing with tumor grade: 0% in grade I, 26.7% in grade II, 45.5% in grade III, and 50% in grade IV . This pattern suggests EYA2 may play a role in the progression of these malignancies rather than their initiation.
EYA2 expression shows a significant positive correlation with astrocytoma tumor grade. Immunohistochemical analysis of 90 astrocytoma specimens revealed the following distribution pattern:
| Grade | Patients | EYA2 Low | EYA2 High | Percentage High |
|---|---|---|---|---|
| I | 8 | 8 | 0 | 0% |
| II | 30 | 22 | 8 | 26.7% |
| III | 22 | 12 | 10 | 45.5% |
| IV | 30 | 15 | 15 | 50% |
This correlation was statistically significant (p=0.03), with higher-grade tumors (III-IV) showing 48.1% EYA2 overexpression compared to only 21.1% in lower-grade tumors (I-II) . No significant correlation was found between EYA2 expression and patient age (p=0.62) or gender (p=0.82) . The progressive increase in EYA2 expression with tumor grade suggests it may contribute to the aggressive behavior and progression of astrocytomas rather than tumor initiation.
EYA2 promotes cancer cell proliferation through several interconnected molecular mechanisms:
Cell cycle regulation: EYA2 facilitates cell cycle progression, particularly at the G1/S checkpoint . Experimental evidence shows that EYA2 overexpression in A172 cells increases the percentage of cells in S phase, while EYA2 knockdown in U251 cells decreases this percentage .
Cyclin regulation: EYA2 positively regulates the expression of key cell cycle proteins, including cyclin D1 and cyclin E, at both mRNA and protein levels . These cyclins are critical for G1/S transition and are established markers of astrocytoma progression .
Transcriptional co-activation: Through its interaction with Six1, EYA2 helps activate the expression of genes involved in cell proliferation . This complex formation is essential for EYA2's oncogenic functions.
ERK pathway activation: EYA2 overexpression upregulates ERK phosphorylation, which contributes to cell proliferation in addition to invasion . The ERK pathway is a well-established regulator of cell growth in various cancers.
DNA damage response modulation: EYA proteins (including EYA2) can dephosphorylate histone H2AX at Y142, directing cells toward DNA repair rather than apoptosis after DNA damage . This function may help cancer cells survive conditions that would normally trigger cell death.
Multiple experimental approaches have established EYA2's role in promoting cancer cell invasion:
Gain and loss of function studies: Transfection of A172 cells (low endogenous EYA2) with an EYA2 expression plasmid significantly enhanced their invasive capacity in Matrigel invasion assays . Conversely, knockdown of EYA2 in U251 cells (high endogenous EYA2) using siRNA substantially decreased their invasive ability .
Molecular pathway analysis: EYA2 overexpression was shown to upregulate both ERK phosphorylation and MMP9 expression (at mRNA and protein levels), both key mediators of cancer cell invasion . The ERK pathway is well-established in promoting invasion by upregulating matrix metalloproteinases.
Inhibitor studies: Treatment with the ERK inhibitor PD98059 abolished the invasion-promoting effects of EYA2 on A172 cells and prevented EYA2-induced MMP9 upregulation . This demonstrates that ERK signaling is a critical downstream mediator of EYA2's effects on invasion.
Co-factor dependency: When Six1 was silenced by siRNA, EYA2 failed to upregulate MMP9 expression in A172 cells . This indicates that the interaction between EYA2 and Six1 is necessary for EYA2's invasion-promoting effects.
Tyrosine phosphatase activity: While not directly shown in astrocytoma, research has demonstrated that EYA's tyrosine phosphatase activity regulates cell motility in other contexts by altering Rho and Rac/cdc42 activities , suggesting additional mechanisms through which EYA2 might promote invasion.
Based on the available research, effective cellular models for studying EYA2 function include:
Cell lines with differential EYA2 expression: A complementary approach using cell lines with contrasting endogenous EYA2 levels provides robust experimental systems. For example:
Cell lines with known Six1 expression: Since EYA2 functionally interacts with Six1, cell lines expressing both proteins allow for studying this interaction. The U251 cell line exhibits high expression of both proteins and has been used successfully to demonstrate their physical interaction through immunoprecipitation .
Inducible expression systems: While not specifically mentioned in the search results, inducible expression systems would allow for temporal control of EYA2 expression, enabling studies of immediate versus long-term effects of EYA2 activation.
3D culture systems: For invasion studies, three-dimensional culture systems that better mimic the tumor microenvironment would provide more physiologically relevant data than traditional 2D cultures.
Patient-derived xenograft models: For in vivo studies, patient-derived xenografts with various levels of EYA2 expression could provide insights into EYA2's role in tumor growth and metastasis under more physiological conditions.
Effective methodological approaches for analyzing EYA2's phosphatase activity include:
Substrate-specific assays: Using known substrates such as phosphorylated histone H2AX (pY142) or estrogen receptor β (pY36) to directly measure EYA2's ability to remove phosphate groups . These assays can be performed with purified proteins or in cellular contexts.
Phosphatase-dead mutants: Creating EYA2 mutants with alterations in the catalytic aspartate residue to generate phosphatase-inactive variants. Comparing the effects of wild-type versus phosphatase-dead EYA2 helps distinguish phosphatase-dependent from phosphatase-independent functions .
Phospho-specific antibodies: Using antibodies that recognize specifically phosphorylated forms of known substrates (such as pY142-H2AX) to monitor EYA2 activity through immunoblotting or immunofluorescence .
In vitro phosphatase assays: Using purified EYA2 protein with synthetic phosphopeptides or purified phosphorylated substrates to measure phosphate release under controlled conditions.
Phosphoproteomic analysis: Mass spectrometry-based approaches to identify novel substrates by comparing the phosphoproteome in cells with and without EYA2 expression or activity.
Small molecule inhibitors: Using specific inhibitors of EYA2's phosphatase activity to assess which cellular functions depend on this enzymatic activity. This approach has therapeutic implications as well .
Several technical considerations are critical when manipulating EYA2 expression:
Expression level control: Ensuring physiologically relevant expression levels is crucial. Extreme overexpression might lead to non-specific effects, while insufficient knockdown might miss functional impacts. Quantitative PCR and western blotting should be used to confirm expression levels .
Domain-specific manipulations: Given EYA2's dual functionality, researchers should consider whether to manipulate the entire protein or specific domains. Constructs expressing only the phosphatase domain or lacking phosphatase activity can help distinguish between EYA2's different functions .
Timing considerations: Acute versus chronic manipulation of EYA2 may yield different results. Inducible systems can help address this issue.
Co-factor expression: Since EYA2 functions in concert with Six1, the endogenous levels of Six1 in the chosen model system will influence experimental outcomes. In some cases, co-transfection of Six1 might be necessary to observe EYA2's full effects .
Functional readouts: Selecting appropriate assays to measure the specific processes of interest is essential. For EYA2, these typically include:
Controls: Including phosphatase-dead mutants as controls when studying enzymatic functions, and using scrambled siRNA sequences for knockdown studies .
The interplay between EYA2's dual functions creates a complex regulatory system that impacts cancer progression through multiple mechanisms:
The phosphatase activity of EYA2 has been shown to dephosphorylate histone H2AX at Y142, directing cells toward DNA repair rather than apoptosis following DNA damage . This function potentially enhances cancer cell survival after genotoxic stress. Additionally, EYA2 dephosphorylates estrogen receptor β at Y36, inhibiting its anti-tumor activity in breast cancer . These direct enzymatic effects operate alongside EYA2's role as a transcriptional co-activator.
As a co-activator, EYA2 interacts with Six1 to regulate gene expression programs that promote proliferation and invasion . The transcriptional effects include upregulation of cyclins D1 and E (promoting cell cycle progression) and MMP9 (enhancing invasion) . Importantly, there appears to be interdependence between these functions, as EYA2 fails to upregulate MMP9 when Six1 is silenced .
The ERK pathway represents a critical nexus in this crosstalk, as EYA2 promotes ERK phosphorylation, and ERK signaling is necessary for EYA2's effects on invasion and MMP9 expression . Whether this ERK regulation depends on EYA2's phosphatase activity, transcriptional function, or both remains an important research question.
This complex interplay creates challenges for therapeutic targeting, as inhibiting only one function of EYA2 might not fully suppress its cancer-promoting effects. A comprehensive understanding of how these functions coordinate in different cancer contexts is essential for developing effective EYA2-targeted therapies.
Current literature suggests potentially diverse roles for EYA2 across cancer types, requiring sophisticated methodological approaches to resolve these apparent contradictions:
Multi-cancer comparative analyses: Systematically comparing EYA2's expression, interacting partners, and downstream targets across multiple cancer types using identical experimental platforms. This should include:
Tissue microarrays spanning multiple cancer types with standardized immunohistochemistry protocols
RNA-seq and ChIP-seq across cancer cell lines representing different tissues of origin
Proteomics and phosphoproteomics to identify cancer-specific substrates and interacting proteins
Context-dependent pathway mapping: Since EYA2 may interact with different signaling networks depending on the cellular context, researchers should:
Employ systems biology approaches to map EYA2-dependent signaling networks in different cancer types
Use CRISPR screens to identify synthetic lethal interactions with EYA2 across cancer types
Investigate tissue-specific co-factors that might modify EYA2 function
In vivo modeling with tissue specificity: Developing animal models with tissue-specific EYA2 manipulation would help determine whether apparent contradictions are due to intrinsic tissue differences:
Conditional EYA2 transgenic or knockout models in different tissue types
Patient-derived xenografts from multiple cancer types with EYA2 modulation
Single-cell approaches: Single-cell RNA-seq and proteomics could reveal whether heterogeneity within tumor populations explains apparent contradictions in EYA2 function.
Structural biology approaches: Determining whether EYA2 undergoes different post-translational modifications or adopts different conformations in various cancer contexts.
These methodological approaches would help determine whether EYA2 truly plays different roles in different cancer types or whether these apparent differences reflect gaps in our understanding of its fundamental mechanisms.
Developing specific inhibitors for EYA2 presents distinct challenges depending on the targeted function:
For EYA2's phosphatase activity:
Unique catalytic mechanism: EYA2 uses an aspartate-based catalytic mechanism unlike conventional cysteine-based protein tyrosine phosphatases . This unusual mechanism requires specialized approaches for inhibitor design.
Substrate specificity: EYA2 shows specificity for tyrosine residues in particular protein contexts (like H2AX and ERβ) . Creating inhibitors that block the active site while maintaining specificity for EYA2 over other phosphatases remains challenging.
Structural considerations: The HAD family phosphatase domain of EYA2 may undergo conformational changes upon substrate binding, complicating structure-based drug design approaches.
For EYA2's protein-protein interactions:
Large interaction surfaces: Protein-protein interactions typically involve large, flat interfaces that are difficult to disrupt with small molecules. The interaction between EYA2 and Six1 likely presents this challenge.
Specificity concerns: Ensuring that inhibitors disrupt only the EYA2-Six1 interaction without affecting other protein-protein interactions of either protein.
Context dependence: The EYA2-Six1 interaction may be modified by cellular context, potentially requiring different inhibition strategies in different tissues.
For both targets:
Redundancy with other EYA family members: EYA1, 2, and 3 share functional overlap in some contexts, such as dephosphorylating H2AX . Inhibitors may need to target multiple EYA proteins or be highly specific to EYA2.
Functional interdependence: Since EYA2's phosphatase activity and transcriptional co-activation are interconnected, inhibiting only one function might lead to compensatory increases in the other.
Biomarker development: Identifying which patients would benefit from EYA2 inhibition requires developing reliable biomarkers of EYA2 dependency.
Despite these challenges, the crystal structure of the EYA2 ED-Six1 complex provides a foundation for rational drug design approaches . Both functions represent potentially valuable therapeutic targets, particularly in cancers where EYA2 overexpression correlates with higher disease grade and poorer outcomes.
EYA2 shows considerable promise as a prognostic biomarker based on several key observations:
To establish EYA2 as a validated clinical biomarker, researchers should:
The current data showing EYA2's association with higher tumor grades provides a strong rationale for further exploration of its prognostic significance in prospective clinical studies.
The EYA2-Six1 interaction represents a promising therapeutic target based on several important observations:
Disrupting the interaction between Six1 and EYA2 significantly reduces Six1-mediated metastasis and increases survival in xenograft breast cancer models . This demonstrates that targeting this protein-protein interaction can have meaningful biological effects. Additionally, EYA2 fails to upregulate MMP9 (a key mediator of invasion) when Six1 is silenced , indicating that this interaction is functionally important for EYA2's oncogenic effects.
Novel therapeutic strategies could include:
Small molecule inhibitors: Developing compounds that bind at the interface between EYA2 and Six1, preventing their interaction. The availability of crystal structure data for the EYA2 ED-Six1 complex provides a foundation for structure-based drug design .
Peptide-based inhibitors: Designing peptides that mimic critical regions of the interaction interface to competitively inhibit complex formation.
Degradation-based approaches: Employing proteolysis-targeting chimeras (PROTACs) or molecular glues to selectively degrade EYA2 or Six1 proteins.
Combination approaches: Targeting both the EYA2-Six1 interaction and EYA2's phosphatase activity simultaneously to overcome potential compensatory mechanisms.
Context-specific strategies: Developing therapies that target the EYA2-Six1 complex specifically in cancer cells by exploiting cancer-specific vulnerabilities or delivery mechanisms.
These approaches could be particularly valuable in cancers like high-grade astrocytoma where EYA2 overexpression is common and correlates with disease progression .
Validating EYA2 as a therapeutic target requires rigorous methodological approaches across multiple dimensions:
Target validation in human tissues:
Comprehensive analysis of EYA2 expression across large, clinically annotated patient cohorts
Correlation of expression with treatment response and survival outcomes
Multi-omics approaches to identify patient subsets most likely to benefit from EYA2 targeting
Mechanistic validation:
Demonstrating that EYA2 inhibition affects critical cancer hallmarks (proliferation, invasion, survival)
Identifying which function of EYA2 (phosphatase activity vs. transcriptional co-activation) is more critical in specific cancer contexts
Mapping the key downstream effectors (like MMP9 and cyclins) that mediate EYA2's oncogenic effects
Preclinical model considerations:
Using models with appropriate EYA2 and Six1 expression levels
Testing EYA2 inhibition in both immunocompetent and immunodeficient models to assess potential immune effects
Evaluating effectiveness against metastatic disease, not just primary tumors
Pharmacological considerations:
Developing highly specific inhibitors with demonstrable on-target activity
Assessing pharmacokinetics and ability to cross the blood-brain barrier (critical for astrocytoma)
Identifying predictive biomarkers of response to EYA2 inhibition
Potential resistance mechanisms:
Investigating whether redundancy among EYA family members might limit effectiveness
Determining if alternate pathways activate ERK signaling or MMP9 expression in the absence of EYA2 function
Identifying rational combination therapies to prevent or overcome resistance
The existing evidence showing EYA2's correlation with tumor grade and its demonstrated roles in promoting proliferation and invasion through well-characterized mechanisms provides a strong foundation for these validation efforts.
The EYA2 gene is located on chromosome 20 (20q13.12) in humans . The gene produces multiple transcript variants encoding different isoforms of the EYA2 protein . The full-length human recombinant EYA2 protein consists of 538 amino acids and is often tagged with GST (Glutathione S-transferase) at the N-terminal for research purposes .
EYA2 functions as a transcriptional coactivator and a phosphatase. It has been shown to play a crucial role in eye development and other developmental processes. In particular, EYA2 is involved in the regulation of transcription, DNA repair, and cellular response to DNA damage .
One of the key functions of EYA2 is its role as a tyrosine phosphatase. It specifically dephosphorylates ‘Tyr-142’ of histone H2AX (H2AXY142ph), which is essential for efficient DNA repair. By dephosphorylating H2AX, EYA2 promotes the recruitment of DNA repair complexes containing MDC1, thereby facilitating the DNA repair process .
EYA2 is involved in several biological processes, including:
Recombinant human EYA2 protein is widely used in research to study its function and role in various biological processes. It is often used in assays such as Western blot, ELISA, and protein arrays . However, it is important to note that recombinant EYA2 protein may contain endotoxins and is not suitable for use with live cells .