Actin Polymerization: Initiates branched actin filaments via NPFs (e.g., WASL), enabling cell migration and phagocytosis .
DNA Repair: Promotes nuclear actin polymerization, facilitating homologous recombination (HR) repair of double-strand breaks (DSBs) .
Transcriptional Regulation: Modulates chromatin structure and gene expression via nuclear actin dynamics .
ARPC2 overexpression is linked to poor prognosis in multiple cancers:
Proliferation: ARPC2 silencing in HCC cells reduces EdU+ proliferative cells by >50% .
Metastasis: Overexpression enhances invasion via MAPK/WNT pathway activation .
Tumor Microenvironment (TME): Correlates with high tumor mutational burden (TMB) and immune checkpoint gene expression .
Experimental Condition | Cell Line | Effect | Apoptosis % Change |
---|---|---|---|
ARPC2 Silencing | HCC-LM3 | Reduced proliferation | +30% (early/late) |
ARPC2 Overexpression | MHCC97-H | Increased invasion | – |
Control | L-02 (normal) | Baseline activity | – |
ARPC2 interacts with:
Partner | Function |
---|---|
Cortactin | Amplifies actin branching |
WASL | Activates Arp2/3 complex |
ACTR2/3 | Forms ATP-binding core |
ARPC1A | Stabilizes complex structure |
The Human ARPC2 ELISA Kit (HUDL00247) enables precise measurement of ARPC2 in biological fluids:
Parameter | Specification |
---|---|
Sensitivity | 0.068 ng/mL |
Dynamic Range | 0.156–10 ng/mL |
Sample Types | Tissue homogenates, serum, plasma |
Recombinant ARPC2 (ab140555) is available for structural and functional studies, with >90% purity .
ARPC2 encodes one of seven subunits (the p34 subunit) of the human Arp2/3 protein complex. This evolutionarily conserved complex plays a critical role in regulating actin polymerization in cells, which is fundamental to cellular processes involving cytoskeletal remodeling . The Arp2/3 complex initiates the formation of new actin filaments from existing filaments, creating branched actin networks that are essential for cell movement, endocytosis, and intracellular trafficking.
For experimental validation of ARPC2 function:
Use siRNA knockdown to observe effects on actin cytoskeleton organization
Perform immunofluorescence microscopy with anti-ARPC2 antibodies to visualize localization
Conduct in vitro actin polymerization assays with purified components
ARPC2 is widely expressed across different human tissues, though with varying expression levels. Data from the Human Protein Atlas (HPA) indicates relatively higher expression in bone marrow, lymph nodes, and blood/immune cells, with lower expression observed in brain tissues and neuronal cells . This pattern suggests tissue-specific regulation mechanisms.
For studying ARPC2 expression:
RT-qPCR for quantitative mRNA expression analysis across tissues
Western blotting for protein-level expression comparisons
Immunohistochemistry for spatial distribution in tissue sections
Analysis of promoter regions to identify tissue-specific regulatory elements
At least two alternatively spliced variants of ARPC2 have been well-characterized, with additional variants described but not fully characterized regarding their full-length nature . The functional differences between these variants remain an active area of research.
Methodological approach for splice variant analysis:
RT-PCR with variant-specific primers
Northern blotting to validate transcript sizes
Cloning and sequencing of variants
Functional studies comparing activities of different isoforms
ARPC2 has been demonstrated to interact with Cortactin, which is an important regulator of actin assembly and cell migration . As part of the Arp2/3 complex, ARPC2 also interacts with the other six subunits of the complex and with various nucleation-promoting factors.
To study protein interactions:
Co-immunoprecipitation followed by mass spectrometry
Yeast two-hybrid screening
Proximity ligation assays
FRET-based interaction studies in living cells
The following table summarizes key cancer types where ARPC2 overexpression correlates with poor survival outcomes:
Cancer Type | OS Impact | DSS Impact | PFI Impact |
---|---|---|---|
ACC | Negative | Negative | Negative |
KIRC | Negative | Negative | Negative |
KIRP | Negative | Negative | Negative |
LGG | Negative | Negative | Negative |
LIHC (HCC) | Negative | Negative | Not significant |
PAAD | Negative | Negative | Negative |
UCEC | Negative | Negative | Negative |
UVM | Negative | Negative | Negative |
SKCM | Positive | Not significant | Not significant |
THYM | Positive | Not significant | Not significant |
For prognostic analysis:
Kaplan-Meier survival analysis with optimal statistical cutoff values
Univariate and multivariate Cox regression analyses
Integration of multiple survival endpoints (OS, DSS, PFI)
Correlation with clinicopathological parameters
Experimental studies, particularly in hepatocellular carcinoma (HCC), demonstrate that ARPC2 silencing significantly inhibits cell proliferation, migration, and invasion, while ARPC2 overexpression promotes these processes . These findings suggest that ARPC2 may enhance cancer cell motility through its role in actin cytoskeleton remodeling, which is essential for invasive and metastatic behaviors.
To investigate mechanisms:
Perform in vitro migration and invasion assays with ARPC2 knockdown/overexpression
Use live-cell imaging to visualize cytoskeletal dynamics
Analyze downstream signaling pathways affected by ARPC2 modulation
Develop in vivo metastasis models to validate in vitro findings
ARPC2 expression shows significant correlations with tumor microenvironment (TME) characteristics, including stromal and immune cell infiltration in various cancer types . This suggests that ARPC2 may influence or be influenced by the tumor immune microenvironment, potentially affecting immunotherapy responses.
Research approach:
Use ESTIMATE algorithm to calculate stromal and immune scores
Apply CIBERSORT to determine relative proportions of 22 infiltrating immune cell types
Perform correlation analyses between ARPC2 expression and immune cell subsets
Validate findings with multiplexed immunohistochemistry or flow cytometry of tumor samples
Evidence suggests that genetic alterations and DNA methylation patterns in tumor tissues may contribute to aberrant ARPC2 expression . Understanding these regulatory mechanisms could reveal potential therapeutic targets or biomarkers.
Methodological considerations:
Analyze copy number variations using GISTIC algorithm
Perform bisulfite sequencing to profile methylation patterns in promoter regions
Integrate multi-omics data (genomic, epigenomic, transcriptomic)
Use CRISPR-based epigenome editing to validate functional impacts of methylation sites
Researchers have successfully employed multiple complementary techniques to assess ARPC2 expression in clinical samples:
RT-qPCR protocol specifics:
RNA extraction using TRIzol reagent or commercial kits
cDNA synthesis with oligo(dT) primers
Use of reference genes like GAPDH or β-actin for normalization
Optimization of primer design to distinguish splice variants
Immunohistochemistry considerations:
Tissue fixation in 10% formalin, paraffin embedding
Antigen retrieval methods optimization
Primary antibody dilution (1:500 recommended for ab133315, Abcam)
Quantification through average optical density measurement using Image-Pro Plus 6.0
Western blotting parameters:
Sample preparation with RIPA buffer containing protease inhibitors
Protein quantification using BCA assay
SDS-PAGE with 10-12% gels
Transfer optimization and blocking conditions
Pan-cancer analysis has utilized several key databases and tools that are particularly valuable for ARPC2 research:
Database resources:
The Cancer Genome Atlas (TCGA) for cancer expression data
Genotype-Tissue Expression (GTEx) database for normal tissue expression
Human Protein Atlas (HPA) for protein expression patterns
UCSC Xena database for integrated multi-omics data
Analytical tools:
R packages: "limma" for differential expression, "survival" and "survminer" for survival analysis
ESTIMATE algorithm for tumor microenvironment analysis
CIBERSORT for immune cell composition analysis
TIMER2.0 for immune infiltration estimation
Based on published research, the following experimental models have proven effective:
Cell lines:
HCC cell lines (HCC-LM3, MHCC97-H, HepG2, huh-7) for cancer studies
Choose cell lines based on endogenous ARPC2 expression levels
Genetic manipulation approaches:
siRNA transfection using TransIntroTM EL Transfection Reagent
Plasmid-based overexpression using pcDNA 3.1(+) vector and Lipofectamine 3000
Consider stable knockdown using shRNA for long-term experiments
CRISPR-Cas9 for complete knockout studies
Functional assays:
Proliferation assays (CCK-8, EdU incorporation)
Migration assays (wound healing, transwell)
Invasion assays (Matrigel-coated transwell)
3D spheroid formation for more physiologically relevant models
For robust statistical analysis of ARPC2 as a prognostic biomarker:
Statistical methods:
Determine optimal expression cutoffs using statistical methods rather than median splits
Perform both Kaplan-Meier analysis with log-rank tests and Cox regression
Analyze multiple survival endpoints (OS, DSS, PFI) for comprehensive assessment
Include multivariate analysis adjusting for known prognostic factors
Visualization approaches:
Kaplan-Meier curves with hazard ratios and confidence intervals
Forest plots for displaying results across multiple cancer types
Box plots for expression differences between clinical subgroups
Correlation heatmaps for relationships with clinical parameters
Researchers should be aware of several potential pitfalls:
Tissue-specific considerations:
ARPC2 has opposite prognostic implications in different cancers (e.g., negative in most cancers but positive in SKCM and THYM)
Expression patterns may reflect tissue of origin rather than cancer-specific changes
Consider the baseline expression in corresponding normal tissues
Technical considerations:
Account for batch effects when combining data from different sources
Be aware of platform-specific biases in expression quantification
Consider sample purity and heterogeneity in tissue samples
Validate findings using multiple technical approaches
Integrative approaches provide the most comprehensive understanding:
Data integration strategies:
Correlate expression with copy number and methylation data
Perform pathway enrichment analysis using associated genes
Use protein-protein interaction networks to identify functional modules
Apply machine learning for pattern recognition across data types
Validation approaches:
Confirm key findings with independent datasets
Use experimental models to validate computational predictions
Apply single-cell approaches to resolve cellular heterogeneity
Consider longitudinal samples to capture dynamic changes
While ARPC2's role in cancer has been increasingly studied, several aspects of its normal function remain to be fully elucidated:
Research opportunities:
Detailed characterization of tissue-specific splice variants
Role in immune cell function given high expression in bone marrow and lymphoid tissues
Developmental regulation during embryogenesis and tissue differentiation
Potential non-canonical functions beyond actin cytoskeleton regulation
Given ARPC2's role in cancer progression, several therapeutic approaches warrant investigation:
Potential strategies:
Direct inhibition of ARPC2 or the Arp2/3 complex
Targeting upstream regulators of ARPC2 expression
Exploiting synthetic lethality with other cytoskeletal regulators
Combination approaches with immunotherapy based on TME correlations
For preclinical evaluation:
Test cytoskeletal inhibitors in ARPC2-high vs. ARPC2-low cancer models
Evaluate effects on tumor growth and metastasis in vivo
Assess potential toxicities in normal tissues
Identify predictive biomarkers for response
The Actin Related Protein 2/3 Complex, Subunit 2 (ARPC2) is a crucial component of the Arp2/3 complex, a multiprotein assembly that plays a significant role in the regulation of the actin cytoskeleton. This complex is essential for various cellular processes, including cell motility, shape, and intracellular transport.
The Arp2/3 complex consists of seven subunits: Arp2, Arp3, ARPC1A, ARPC1B, ARPC2, ARPC3, ARPC4, and ARPC5 . ARPC2, also known as p34-Arc, is one of these subunits and is integral to the complex’s function. The ARPC2 subunit is approximately 34 kDa in size and is encoded by the ARPC2 gene located on chromosome 2 in humans .
The primary function of the Arp2/3 complex is to initiate the formation of branched actin networks. This is achieved through the nucleation of new actin filaments, a process that is stimulated by nucleation-promoting factors (NPFs) . The complex binds to the sides of existing actin filaments and creates a new branch, thereby generating a dense and dynamic actin network.
ARPC2, as part of the Arp2/3 complex, contributes to various cellular activities:
The Arp2/3 complex, including ARPC2, is vital for maintaining the structural integrity of the cytoskeleton. It is involved in various cellular processes such as:
Mutations or dysregulation of the ARPC2 gene can lead to various diseases. For instance, abnormalities in the Arp2/3 complex have been associated with endometrial type cervical adenomyoma and cervical adenomyoma . Understanding the function and regulation of ARPC2 is crucial for developing therapeutic strategies for these conditions.