Recombinant Rat Arf-GAP with GTPase, ANK repeat and PH domain-containing protein 2 (Agap2), partial

Shipped with Ice Packs
In Stock

Description

Research Applications

The recombinant protein is used to study Agap2’s role in cellular processes and validate its interactions with signaling molecules.

Key Applications

  • Western Blotting (WB): Detects Agap2 in human, mouse, and rat tissues (brain, gliomas) .

  • Immunoprecipitation (IP): Identifies binding partners like FAK (focal adhesion kinase) .

  • Functional Assays:

    • Focal Adhesion Dynamics: Agap2 overexpression disrupts focal adhesions, promoting cell migration .

    • Retrograde Transport: Required for trafficking of Shiga toxin, cholera toxin, and TGN46 .

    • Phagocytosis: Enhances Fcγ receptor-mediated uptake in neutrophils via GAP domain-dependent recruitment .

Functional Mechanisms

Agap2 regulates membrane trafficking and cytoskeletal remodeling through distinct domains:

Membrane Trafficking

  • Retrograde Transport: Agap2 interacts with AP-1 adaptors, facilitating early endosome-to-TGN transport of toxins and cargo proteins (e.g., TGN46, CI-MPR) .

  • Endosomal Recycling: Regulates transferrin recycling via Rab4-dependent pathways .

Cytoskeletal Remodeling

  • Focal Adhesions: Binds FAK, increasing its Tyr-397 phosphorylation and promoting adhesion disassembly .

  • Phagocytosis: Localizes to phagocytic cups in neutrophils, enhancing opsonized particle uptake independently of GAP activity .

Cancer and Disease Implications

  • Oncogenic Role: Overexpressed in glioblastomas and hepatocellular carcinomas (HCC), promoting invasion and survival via Akt activation .

  • Transcriptional Regulation: SP1 and RARα drive Agap2 expression in cancer cells, linking retinoid signaling to tumorigenesis .

Table 1: Agap2 Interactions and Functional Outcomes

Interaction/ProcessPartner MoleculeOutcomeSource
FAK BindingFAKIncreased FAK phosphorylation, adhesion disassembly
AP-1 AdaptorsAP-1Retrograde transport of STxB, CTxB
Akt ActivationAktEnhanced cell survival, migration in cancer

Caveats and Limitations

Future Directions

  • Cancer Therapeutics: Targeting Agap2’s PH domain or GAP activity to inhibit tumor invasion .

  • Infectious Disease Models: Investigating Agap2’s role in retrograde transport of bacterial toxins .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. Please specify your desired tag type for preferential development.
Synonyms
Agap2; Centg1Arf-GAP with GTPase; ANK repeat and PH domain-containing protein 2; AGAP-2; Centaurin-gamma-1; Cnt-g1; Phosphatidylinositol 3-kinase enhancer; PIKE
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Rattus norvegicus (Rat)
Target Names
Uniprot No.

Target Background

Function
This protein is a GTPase-activating protein (GAP) for ARF1 and ARF5, exhibiting strong GTPase activity. It participates in neuronal apoptosis prevention by enhancing PI3 kinase activity. It facilitates the coupling of metabotropic glutamate receptor 1 (GRM1) to cytoplasmic PI3 kinase through interaction with Homer scaffolding proteins and mediates the anti-apoptotic effects of NGF by activating nuclear PI3 kinase.
Gene References Into Functions
  1. PIKE plays crucial roles in oligodendroglia development and myelinogenesis via Akt/mTOR activation. PMID: 24449917
  2. PIKE is a critical regulator of synaptic AMPA receptor insertion. PMID: 21847098
  3. Pike/nuclear PI 3-kinase signaling mediates the anti-apoptotic effects of nerve growth factor in the cell nucleus. PMID: 15385964
Database Links
Protein Families
Centaurin gamma-like family
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Present in cortex, hippocampus and olfactory bulb but absent in cerebellum (at protein level).

Q&A

What is AGAP2 and what are its functional domains?

AGAP2 (Arf-GAP with GTPase, ANK repeat and PH domain-containing protein 2) is a multidomain protein belonging to the centaurin gamma-like family. It contains several functional domains including:

  • GTPase domain (GLD)

  • Ankyrin repeat domain (ANK)

  • Pleckstrin homology domain (PH)

These domains work collectively to enable AGAP2's diverse cellular functions. The PH domain of AGAP2 has been identified as crucial for binding to focal adhesion kinase (FAK), as demonstrated through immunoprecipitation and GST pulldown assays . The protein is also known as PIKE-L in some research contexts and functions as a scaffold protein that coordinates membrane and actin cytoskeleton dynamics essential for normal physiological functions including development, wound healing, and phagocytosis .

To study domain-specific functions, researchers typically generate truncated forms of AGAP2 including PH2 (isolated PH domain), PZA2 (partial construct), and GLD2 (isolated GTPase domain) for comparative binding and functional assays .

What cellular pathways does AGAP2 regulate?

AGAP2 participates in multiple cellular pathways with significance for both normal physiology and disease states:

  • Receptor trafficking pathway: AGAP2 promotes the fast recycling of transferrin receptors and regulates β2-adrenergic receptor trafficking through interaction with β-arrestin1 and β-arrestin2 .

  • Cell survival signaling: The protein mediates anti-apoptotic effects of nerve growth factor by activating nuclear phosphoinositide 3-kinase, providing protective effects against apoptosis in certain cell types .

  • Cell migration and invasion pathways: AGAP2 regulates focal adhesion dynamics through its interaction with FAK, influencing cell motility and invasion potential .

  • MAPK/ERK signaling: AGAP2 forms complexes with endogenous ERK and can potentiate ERK phosphorylation induced by β2-adrenergic receptors, affecting downstream signal transduction .

  • Akt pathway interactions: Research has shown that AGAP2 binds to activated Akt, which increases invasion of glioblastoma cells and provides protection against apoptosis .

Methodologically, these pathways are best studied through a combination of co-immunoprecipitation, phosphorylation assays, and cellular phenotype assessments following AGAP2 manipulation.

How is AGAP2 expression altered in cancer compared to normal tissues?

AGAP2 expression exhibits significant differences between normal and cancerous tissues. Based on TCGA database analyses and immunohistochemical verification:

  • AGAP2 is frequently overexpressed in various cancer cells compared to corresponding normal tissues .

  • In clear cell renal cell carcinoma (ccRCC), AGAP2 expression correlates with clinical cancer stages and tumor progression. Higher expression levels are associated with:

CharacteristicLow expression of AGAP2High expression of AGAP2p-value
T stage0.015
T1153 (28.4%)125 (23.2%)
T239 (7.2%)32 (5.9%)
T373 (13.5%)106 (19.7%)
T44 (0.7%)7 (1.3%)
M stage0.014
M0227 (44.9%)201 (39.7%)
M129 (5.7%)49 (9.7%)
Status event0.029
Alive195 (36.2%)171 (31.7%)
Dead74 (13.7%)99 (18.4%)

These data indicate that higher AGAP2 expression is associated with more advanced T stages, higher metastasis rates, and poorer survival outcomes .

For accurate assessment of AGAP2 expression in research contexts, immunohistochemistry validation following initial database mining is essential to confirm expression patterns across tissue types.

What methods are most effective for studying AGAP2 protein interactions?

When investigating AGAP2 protein interactions, researchers should consider multiple complementary approaches:

  • Co-immunoprecipitation (Co-IP): This technique has been successfully employed to demonstrate AGAP2's interactions with FAK, β-arrestin1, and β-arrestin2. For optimal results, use FLAG epitope-tagged AGAP2 constructs and M2 anti-FLAG antibody for immunoprecipitation, followed by immunoblotting for potential binding partners .

  • GST pulldown assays: Generate GST fusion proteins of AGAP2 (full-length or domain-specific constructs) in BL21 bacterial cells and purify using glutathione-Sepharose 4B beads. This approach effectively identified the PH2 domain as the critical region for FAK binding .

  • Domain mapping: Express truncated versions of AGAP2 containing specific functional domains (GLD2, PH2, PZA2) to identify binding sites for interaction partners. This approach revealed that the PH2 domain, not the GLD2 domain, mediates binding to FAK .

  • Cellular co-localization: Use fluorescence microscopy with appropriate antibodies or fluorescently tagged proteins to visualize AGAP2 co-localization with potential binding partners, such as β-arrestin2 on plasma membranes and β2-adrenergic receptors on endosomes .

When designing these experiments, include appropriate controls such as non-binding domains (e.g., isolated GLD2) and related family members (e.g., comparing AGAP1 and AGAP2 binding) to establish specificity of interactions .

What are proven techniques for AGAP2 knockdown in experimental models?

Several effective approaches for AGAP2 knockdown have been validated in research settings:

  • siRNA-mediated knockdown:

    • Use two complementary siRNA duplexes targeting different regions of AGAP2 mRNA

    • Validated sequences include: 5′-AGA CAC AUC UGG UGC UAA U-3′ and 5′-GUA AUG GCU UUC UAC UCU A-3′

    • Transfect at 50 nM concentration each using Lipofectamine RNAiMax

    • Include non-targeting control siRNA (e.g., Dharmacon non-targeting siRNA pool) as negative control

    • Verify knockdown efficiency by Western blotting after 48-72 hours

  • shRNA-mediated stable knockdown:

    • Generate lentiviral particles containing AGAP2-targeting shRNA

    • Infect target cells for 2 hours followed by medium change

    • Select stable integrants using puromycin (2 μg/ml initially, 1 μg/ml for maintenance)

    • Use uninfected cells as selection control

    • Confirm knockdown efficiency by Western blotting

These methodologies have been validated in multiple cell lines including U87 glioblastoma and HEK293 cells. When implementing these techniques, it's crucial to verify knockdown at both mRNA and protein levels and to include appropriate controls to rule out off-target effects.

What assays best measure AGAP2 functional effects in cellular models?

Based on AGAP2's known functions, the following assays are recommended to comprehensively assess its cellular effects:

  • Cell migration assays:

    • Use migration chambers with appropriate chemoattractants (e.g., EGF at 50 nM)

    • Seed 1.0 × 10^5 starved cells in Opti-MEM

    • Quantify migration using fluorescent dye staining and measurement

    • Compare migration with and without stimulation across control and AGAP2-manipulated cells

  • Receptor trafficking assays:

    • For β2-adrenergic receptor trafficking: monitor receptor internalization and recycling using fluorescently-labeled receptors

    • Assess recycling kinetics in cells overexpressing AGAP2 versus knockdown conditions

    • Track receptor accumulation in perinuclear recycling endosomes versus plasma membrane localization

  • Signaling pathway activation:

    • Measure ERK phosphorylation levels following receptor activation

    • Compare phosphorylation kinetics in control versus AGAP2-overexpressing or knockdown cells

    • Use Western blotting with phospho-specific antibodies for quantification

  • Focal adhesion dynamics:

    • Visualize focal adhesions using antibodies against markers like vinculin, paxillin, or FAK

    • Assess focal adhesion turnover rates in AGAP2-manipulated cells

    • Correlate changes in focal adhesion dynamics with cell migration capacity

For all functional assays, include appropriate positive and negative controls, and when possible, rescue experiments by reintroducing wild-type or mutant AGAP2 into knockdown cells to confirm specificity of observed phenotypes.

How does AGAP2 mechanistically regulate cell migration and invasion?

AGAP2 regulates cell migration and invasion through multiple interconnected mechanisms:

  • Focal adhesion regulation: AGAP2 physically interacts with focal adhesion kinase (FAK) via its PH2 domain, as demonstrated through co-immunoprecipitation and GST pulldown assays. This interaction affects focal adhesion dynamics essential for cell motility, including assembly and disassembly rates of adhesion complexes .

  • Cytoskeletal remodeling: As a member of the AGAP family, AGAP2 coordinates changes in membrane and actin cytoskeleton under several motor cell structures. This coordination is essential for cellular processes including development and wound healing under normal conditions, but can contribute to pathological behaviors like cancer cell invasion and metastasis in disease states .

  • Akt pathway activation: AGAP2 binds directly to activated Akt, enhancing invasion potential particularly in glioblastoma cells. This interaction provides a molecular link between AGAP2 overexpression and increased invasiveness in certain cancer types .

  • Receptor trafficking influence: Through its interaction with β-arrestin2, AGAP2 regulates the trafficking of receptors such as β2-adrenergic receptors. This function impacts cellular signaling cascades that contribute to migration behavior .

In experimental settings, these mechanisms can be investigated using a combination of protein interaction studies, live-cell imaging of focal adhesion dynamics, and functional migration/invasion assays following genetic manipulation of AGAP2. Researchers should pay particular attention to cell type-specific effects, as AGAP2's impact may vary across different tissue contexts.

What is the relationship between AGAP2 and tumor microenvironment?

The relationship between AGAP2 and the tumor microenvironment is an emerging area of research with significant implications for cancer biology:

  • Immune cell infiltration correlation: Analysis of TCGA dataset and Tumor Immune Estimation Resource (TIME) suggests a relationship between AGAP2 expression and immune cell infiltration patterns in tumors. This was specifically investigated in clear cell renal cell carcinoma, where AGAP2 expression correlates with specific immune cell populations .

  • Pathway enrichment: Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of AGAP2-related genes have revealed enrichment in biological pathways relevant to the tumor microenvironment. While specific pathway details aren't fully detailed in the search results, these analyses suggest AGAP2 may influence tumor-stroma interactions .

  • Prognostic significance: The correlation between high AGAP2 expression and poor prognosis in ccRCC (with increased rates of advanced T stage, metastasis, and mortality) suggests its potential role in modifying tumor behavior within the microenvironment context .

Methodologically, researchers investigating this relationship should employ:

  • Single-cell RNA sequencing to delineate cell type-specific expression

  • Spatial transcriptomics to map AGAP2 expression patterns relative to stromal and immune cells

  • Multiplex immunohistochemistry to simultaneously visualize AGAP2-expressing cells and immune infiltrates

  • Co-culture systems to model interactions between AGAP2-expressing tumor cells and stromal/immune components

This approach will help elucidate how AGAP2 might influence the complex cellular ecosystem within tumors.

How can AGAP2 be leveraged as a prognostic biomarker in cancer research?

AGAP2 shows promise as a prognostic biomarker, particularly in clear cell renal cell carcinoma (ccRCC), with potential applications in other cancer types:

  • Expression analysis methodology:

    • Utilize TCGA database mining followed by validation in independent cohorts

    • Verify with immunohistochemistry in patient samples

    • Quantify expression using standardized scoring systems (H-score or percentage positive cells)

  • Clinical correlation approach:

    • Stratify patients by AGAP2 expression levels (high vs. low)

    • Correlate with clinicopathological features including tumor stage, grade, and metastatic status

    • Perform Kaplan-Meier survival analysis to assess prognostic significance

    • Use multivariate analysis to determine independence from other prognostic factors

  • Integration with other biomarkers:

    • Combine AGAP2 expression with other molecular markers for improved prognostic accuracy

    • Develop composite scoring systems incorporating multiple biomarkers

    • Validate in prospective cohorts to confirm clinical utility

As shown in ccRCC studies, AGAP2 expression correlates significantly with T stage (p=0.015), M stage (p=0.014), and patient survival (p=0.029), with high expression associated with poorer outcomes . When designing biomarker studies, researchers should:

  • Include adequate sample sizes with appropriate statistical power

  • Account for intratumoral heterogeneity through multiple sampling

  • Standardize tissue processing and immunostaining protocols

  • Utilize digital pathology for objective quantification when possible

  • Validate findings across diverse patient populations

What are common challenges in AGAP2 protein purification and how can they be addressed?

Researchers working with recombinant AGAP2 often encounter several technical challenges during protein purification:

  • Protein solubility issues:

    • Challenge: Full-length AGAP2 may exhibit poor solubility due to its multiple domains.

    • Solution: Express domain-specific constructs (GLD2, PH2, PZA2) separately as demonstrated in successful studies. For GST fusion proteins, expression in BL21 cells at lower temperatures (16-18°C) can improve solubility .

  • Protein stability concerns:

    • Challenge: Multidomain proteins like AGAP2 can be prone to degradation during purification.

    • Solution: Include protease inhibitors throughout the purification process. For GST-AGAP2 fusion proteins, maintain protein-bound to glutathione-Sepharose 4B beads until use in binding assays to minimize handling-related degradation .

  • Binding specificity verification:

    • Challenge: Ensuring observed protein interactions are specific rather than non-specific binding.

    • Solution: Include appropriate controls such as isolated domains known not to interact (e.g., GLD2 domain) and related family members (e.g., AGAP1) for comparison .

  • Appropriate buffer conditions:

    • Challenge: Identifying optimal buffer conditions for maintaining AGAP2 activity.

    • Solution: When working with purified GST-AGAP2 fusion proteins, wash protein-bound beads thoroughly with PBS before incubation with cell lysates. For cell-based assays, prepare lysates in buffers that preserve protein-protein interactions while minimizing non-specific binding .

For researchers new to working with AGAP2, beginning with domain-specific constructs rather than the full-length protein may provide higher success rates while developing purification expertise.

How should contradictory results regarding AGAP2 function be interpreted?

When facing contradictory results in AGAP2 research, consider these methodological approaches for resolution:

  • Cell type-specific effects:

    • AGAP2 may function differently across cell types. For example, while AGAP2 forms complexes with FAK in multiple cell lines (U87, HEK293, MCF-7), the functional consequences may vary .

    • Resolution approach: Systematically compare AGAP2 function across multiple cell lines under identical experimental conditions, and clearly report cell type alongside results.

  • Isoform variations:

    • AGAP2 has multiple isoforms (e.g., PIKE-L) with potentially distinct functions .

    • Resolution approach: Clearly identify which isoform is being studied and use isoform-specific reagents when possible. Consider comparative studies of different isoforms.

  • Experimental context differences:

    • AGAP2's function in receptor trafficking varies depending on the receptor type and cellular context .

    • Resolution approach: Precisely define experimental conditions including cell density, passage number, and stimulation parameters to ensure reproducibility.

  • Methodological variations:

    • Different knockdown approaches (transient siRNA vs. stable shRNA) may yield varying phenotypes due to compensation mechanisms .

    • Resolution approach: Employ multiple independent methods to manipulate AGAP2 expression/function and compare results.

When publishing seemingly contradictory findings, researchers should explicitly discuss differences from previous studies, paying particular attention to methodological variations, cell types used, and specific experimental conditions that might explain discrepancies.

What statistical analyses are most appropriate for AGAP2 expression data in clinical samples?

When analyzing AGAP2 expression in clinical samples, researchers should employ these statistical approaches:

  • For comparing expression levels between groups:

    • Non-parametric tests (Mann-Whitney U or Kruskal-Wallis) are often appropriate as expression data frequently violate normality assumptions

    • When comparing across multiple groups (e.g., tumor stages), use ANOVA with appropriate post-hoc tests while checking assumptions

    • For categorical analyses (e.g., high vs. low expression), use chi-square or Fisher's exact test depending on sample size

  • For survival analyses:

    • Kaplan-Meier method with log-rank test to compare survival between high and low AGAP2 expression groups

    • Cox proportional hazards regression for multivariate analysis to determine if AGAP2 is an independent prognostic factor

    • Time-dependent ROC curve analysis to assess prognostic performance at different time points

  • For correlating with clinical parameters:

    • Spearman's rank correlation for continuous variables

    • Contingency table analysis for categorical variables (as shown in the T stage, M stage, and survival status table)

  • For gene expression correlation studies:

    • Pearson or Spearman correlation depending on data distribution

    • Gene set enrichment analysis (GSEA) to identify pathways associated with AGAP2 expression

    • Heat map visualization with hierarchical clustering to identify expression patterns

When reporting results, include:

  • Sample size and power calculations

  • Effect size estimates alongside p-values

  • Multiple testing corrections when appropriate (e.g., Benjamini-Hochberg procedure)

  • Transparent reporting of all statistical methodologies and software packages used

What is the potential of AGAP2 as a therapeutic target in cancer?

AGAP2 shows promising characteristics as a potential therapeutic target in cancer, particularly based on these research findings:

  • Overexpression in multiple cancers: AGAP2 is overexpressed in various cancer types and promotes cancer cell invasion, suggesting it as a common mechanism supporting tumor progression .

  • Association with poor prognosis: Higher AGAP2 expression correlates with advanced tumor stage, increased metastasis, and decreased survival in clear cell renal cell carcinoma, indicating clinical relevance of targeting this protein .

  • Multiple druggable mechanisms:

    • Inhibition of AGAP2's interaction with FAK could disrupt focal adhesion dynamics and reduce invasiveness

    • Targeting AGAP2-β-arrestin2 interactions might impair receptor trafficking and downstream signaling

    • Disrupting AGAP2-Akt binding could reduce protective effects against apoptosis in cancer cells

  • Pathway convergence: AGAP2 interacts with multiple cancer-associated pathways including ERK signaling and receptor trafficking, potentially providing a node for therapeutic intervention .

For researchers pursuing AGAP2 as a therapeutic target, these methodological approaches should be considered:

  • Structure-based drug design targeting specific AGAP2 domains (particularly the PH2 domain)

  • High-throughput screening for small molecules disrupting key protein-protein interactions

  • Peptide-based inhibitors designed to compete with natural binding partners

  • Evaluation of selective vulnerability in AGAP2-overexpressing cancer cells versus normal cells

  • Combination approaches targeting AGAP2 alongside established therapeutic targets

Pre-clinical validation should include careful assessment of effects in both AGAP2-overexpressing and normal cell lines to establish therapeutic window and potential toxicities.

How is AGAP2 functionally connected to other GTPase-regulating proteins?

AGAP2, as an Arf GTPase-activating protein, operates within a complex network of GTPase-regulating proteins with interconnected functions:

  • Arf GTPase regulation network:

    • AGAP2 functions as a GTPase-activating protein (GAP) for Arf GTPases, promoting GTP hydrolysis and inactivation

    • This activity positions AGAP2 within the broader Arf regulatory network that includes guanine nucleotide exchange factors (GEFs) that activate Arfs and other GAPs with potentially overlapping specificities

    • Understanding these interactions requires comparative studies of multiple Arf regulators in the same experimental systems

  • Comparative function within the AGAP family:

    • AGAP2 belongs to the AGAP family that includes related proteins such as AGAP1

    • While both AGAP1 and AGAP2 can interact with FAK, AGAP2 shows stronger binding capacity, suggesting functional specialization within the family

    • Methodological approach: Comparative binding and functional assays with multiple AGAP family members can elucidate specific versus redundant functions

  • Intersection with small GTPase signaling pathways:

    • Beyond Arf regulation, AGAP2 functionally connects to other GTPase-dependent pathways including those regulated by Rho-family GTPases involved in cytoskeletal dynamics

    • The coordination of membrane and actin cytoskeleton by AGAP2 likely involves cross-talk between multiple GTPase-regulated pathways

For researchers investigating these connections, recommended approaches include:

  • Proteomics studies to identify the complete interactome of AGAP2

  • Comparative functional assays following knockdown of multiple GTPase regulators

  • Live-cell imaging with fluorescent biosensors to monitor spatial and temporal activation patterns of different GTPases in relation to AGAP2 activity

Understanding these interconnections may reveal synthetic lethal interactions that could be exploited therapeutically, particularly in cancer contexts where AGAP2 is overexpressed.

What novel techniques show promise for studying AGAP2 regulation in vivo?

Advancing our understanding of AGAP2 function requires implementation of cutting-edge techniques that allow for dynamic, in vivo analysis:

  • CRISPR-based approaches:

    • CRISPR-Cas9 genome editing for generating AGAP2 knockout or domain-specific mutant animal models

    • CRISPR interference (CRISPRi) or CRISPR activation (CRISPRa) systems for temporal control of AGAP2 expression in specific tissues

    • CRISPR-based lineage tracing to monitor effects of AGAP2 manipulation on cell fate in vivo

  • Advanced imaging technologies:

    • Fluorescence resonance energy transfer (FRET) biosensors to monitor AGAP2 activation states and protein interactions in living cells

    • Lattice light-sheet microscopy for high-resolution spatiotemporal analysis of AGAP2 dynamics during cellular processes

    • Intravital microscopy to visualize AGAP2-expressing cells in tumor microenvironments in real-time

  • Single-cell and spatial transcriptomics:

    • Single-cell RNA sequencing to identify cell populations with distinctive AGAP2 expression patterns

    • Spatial transcriptomics to map AGAP2 expression relative to microenvironmental features in tissues

    • Multi-omics approaches integrating transcriptomic, proteomic, and phosphoproteomic data

  • Functional proteomics:

    • Proximity labeling techniques (BioID, APEX) to identify context-specific AGAP2 interactors

    • Phosphoproteomics to map AGAP2-dependent signaling networks

    • Targeted protein degradation approaches (e.g., dTAG, PROTAC) for acute AGAP2 depletion to distinguish direct from compensatory effects

These methodologies should be implemented in physiologically relevant model systems, including patient-derived organoids and genetically engineered mouse models that recapitulate AGAP2 expression patterns observed in human cancers. Such approaches will provide more nuanced insights into AGAP2 regulation and function than traditional in vitro systems alone.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.