ARPC1A Antibody

Shipped with Ice Packs
In Stock

Description

Research Findings in Cancer Biology

ARPC1A is overexpressed in multiple cancers and correlates with aggressive phenotypes:

Table 1: ARPC1A in Cancer Progression

Cancer TypeRole in MetastasisKey MechanismsClinical ImpactSource
Prostate CancerPromotes migration/invasion via actin cytoskeleton remodelingSilencing reduces cell motility (in vitro); linked to 1.58x higher risk of biochemical recurrencePoor prognosis biomarker
Non-Small Cell Lung Cancer (NSCLC)Enhances proliferation via c-Myc regulationKnockdown reduces colony formation, migration, and invasion (in vitro)Potential therapeutic target
Glioblastoma (GBM)Drives temozolomide (TMZ) resistanceARPC1A suppression sensitizes cells to TMZPredictive biomarker for therapy resistance
  • Immune Modulation: High ARPC1A expression correlates with suppressed interferon pathways and reduced immune infiltration in tumors .

  • Therapeutic Potential: Preclinical studies suggest ARPC1A inhibition could reduce metastatic spread .

Applications in Biomedical Research

ARPC1A antibodies are widely used to study:

  1. Actin Dynamics: Visualizing Arp2/3-mediated actin branching in cell migration assays .

  2. Cancer Biomarker Validation: Detecting ARPC1A overexpression in tumor tissues (e.g., prostate, lung, and glioma) .

  3. Drug Development: Screening inhibitors targeting Arp2/3 complex activity .

Common Protocols:

ApplicationDilution RangeRecommended Cell Lines
Western Blot0.04–0.4 µg/mL293T (transfected lysate)
Immunofluorescence0.25–2 µg/mLPC-3, DU-145 (prostate cancer)
IHC (FFPE)1:200–1:500Human tumor tissue microarrays

Future Directions

  1. Clinical Translation: ARPC1A’s role in immune evasion highlights its potential as a biomarker for immunotherapy response .

  2. Therapeutic Targeting: Small-molecule inhibitors or monoclonal antibodies against ARPC1A could disrupt metastatic pathways .

  3. Multi-Omics Integration: Combining ARPC1A expression data with genomic and proteomic profiles may refine prognostic models .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please contact your local distributor for specific delivery details.
Synonyms
Actin binding protein (Schizosaccharomyces pombe sop2 like) antibody; Actin related protein 2/3 complex subunit 1A 41kDa antibody; Actin-related protein 2/3 complex subunit 1A antibody; ARC1A_HUMAN antibody; Arc40 antibody; ARPC1A antibody; Epididymis luminal protein 68 antibody; Epididymis secretory protein Li 307 antibody; HEL 68 antibody; HEL S 307 antibody; SOP2-like protein antibody; SOP2Hs antibody; SOP2L antibody
Target Names
Uniprot No.

Target Background

Function
ARPC1A likely functions as a component of the Arp2/3 complex. This complex plays a crucial role in regulating actin polymerization. In conjunction with an activating nucleation-promoting factor (NPF), the Arp2/3 complex facilitates the formation of branched actin networks.
Gene References Into Functions
  1. Research suggests that ARPC1A is a novel target for the 7q21-q22 amplification and acts as a regulator of cell migration and invasion in pancreatic cancer. PMID: 19145645
Database Links

HGNC: 703

OMIM: 604220

KEGG: hsa:10552

STRING: 9606.ENSP00000262942

UniGene: Hs.124126

Protein Families
WD repeat ARPC1 family
Subcellular Location
Cytoplasm, cytoskeleton. Nucleus.

Q&A

What criteria should be considered when selecting an ARPC1A antibody for experimental use?

When selecting an ARPC1A antibody, researchers should consider several critical factors: (1) Target specificity - verify the immunogen sequence to ensure it targets your region of interest within ARPC1A; (2) Validated applications - confirm the antibody has been validated for your specific application (WB, IHC, IF/ICC); (3) Species reactivity - ensure compatibility with your experimental model (human, mouse, rat, etc.); (4) Clonality - polyclonal antibodies offer broader epitope recognition while monoclonals provide greater specificity; (5) Validation data - review existing validation including western blot bands (37-42 kDa expected) and positive control recommendations like HEK-293 or PC-3 cells . The comprehensive validation data should ideally include knockout/knockdown controls to confirm specificity.

How can I validate the specificity of an ARPC1A antibody in my experimental system?

Validation should employ multiple complementary approaches: (1) Positive controls - test the antibody in cell lines with known ARPC1A expression (HEK-293, PC-3 cells are recommended) ; (2) Negative controls - include samples where the primary antibody is omitted; (3) Molecular weight verification - confirm detection at the expected 37-42 kDa range ; (4) RNAi knockdown validation - compare antibody signal between ARPC1A-expressing and knockdown samples to confirm specificity ; (5) Peptide competition assay - pre-incubate the antibody with immunizing peptide to block specific binding; (6) Cross-reactivity assessment - test against related proteins, particularly ARPC1B which shares structural similarity . Document all validation steps methodically for publication requirements.

What is the optimal sample preparation protocol for detecting ARPC1A in Western blotting?

For optimal Western blot detection of ARPC1A: (1) Cell lysis - use RIPA buffer with protease inhibitors to effectively extract ARPC1A while preserving its integrity ; (2) Protein quantification - standardize loading (35 μg recommended based on published protocols) ; (3) Denaturation - heat samples at 95°C for 5 minutes in reducing buffer; (4) Gel percentage - use 10-12% polyacrylamide gels to properly resolve the 37-42 kDa ARPC1A protein; (5) Transfer conditions - optimize transfer time for proteins in this molecular weight range (typically 60-90 minutes at 100V); (6) Blocking - use 5% non-fat milk or BSA in TBST; (7) Primary antibody incubation - dilute according to manufacturer recommendations (typically 1:500-1:3000) and incubate at 4°C overnight; (8) Detection - ECL method has been successfully used in published work . This protocol has been validated for detecting ARPC1A in various human cell lines including HEK-293 and PC-3.

What are the optimal conditions for immunohistochemical detection of ARPC1A in prostate cancer tissues?

For IHC detection of ARPC1A in prostate cancer tissues: (1) Fixation - use 10% neutral buffered formalin followed by paraffin embedding; (2) Sectioning - prepare 4-5 μm thick sections; (3) Antigen retrieval - heat-induced epitope retrieval in citrate buffer (pH 6.0) is generally effective; (4) Blocking - block endogenous peroxidase activity with 3% H₂O₂ and non-specific binding with serum; (5) Primary antibody - dilute according to manufacturer recommendations (typically 1:50-1:200) and incubate overnight at 4°C; (6) Detection system - use appropriate secondary antibody and visualization system (DAB is commonly used); (7) Counterstaining - lightly counterstain with hematoxylin; (8) Controls - include positive controls (prostate cancer tissues with known ARPC1A expression) and negative controls (omitting primary antibody) . This protocol has been validated in tissue microarray (TMA) studies of prostate cancer samples containing 301 cases .

How can I optimize immunofluorescence staining to visualize ARPC1A in cytoskeletal structures?

To optimize IF staining for ARPC1A in cytoskeletal structures: (1) Cell preparation - grow cells on glass coverslips to 70-80% confluence; (2) Fixation - use 4% paraformaldehyde for 15 minutes at room temperature to preserve cytoskeletal structure; (3) Permeabilization - 0.1% Triton X-100 for 10 minutes to allow antibody access to intracellular targets; (4) Blocking - use 3-5% BSA or normal serum for 1 hour at room temperature; (5) Primary antibody - dilute to manufacturer's recommendations (1:10-1:50 has been successful) ; (6) Co-staining - include phalloidin for F-actin visualization to examine colocalization with ARPC1A; (7) Secondary antibody - use fluorescently-labeled secondary antibodies specific to the primary antibody host species; (8) Counterstaining - include DAPI for nuclear visualization; (9) Mounting - use anti-fade mounting medium to prevent photobleaching . This approach has been successfully used to demonstrate cytoskeletal changes in ARPC1A-knockdown prostate cancer cells .

What controls should be included when examining ARPC1A expression in comparative studies between normal and cancer tissues?

For rigorous comparative studies: (1) Tissue controls - include matched normal and tumor tissues from the same patients when possible; (2) Cellular controls - include cell lines with known ARPC1A expression levels; (3) Technical controls - process all samples simultaneously with identical protocols; (4) Antibody controls - include primary antibody omission and isotype controls; (5) Quantitative controls - standardize protein loading for Western blots (35 μg recommended) ; (6) Expression validation - confirm expression patterns using multiple techniques (e.g., IHC, WB, IF); (7) Internal reference controls - include housekeeping proteins as loading controls for normalization; (8) Biological replicates - analyze multiple independent samples (the TCGA cohort used 434 PCa tissues and 51 normal prostate tissues) . This comprehensive approach enables robust statistical analysis of differential expression patterns.

How does ARPC1A contribute to the function of the Arp2/3 complex in actin polymerization?

ARPC1A functions as a critical structural component of the seven-subunit Arp2/3 complex: (1) Structural role - ARPC1A contains WD repeats that form a β-propeller structure, serving as a scaffold within the complex; (2) Complex assembly - evidence suggests ARPC1A is involved in assembling and maintaining Arp2/3 complex structure; (3) Nucleation promotion - ARPC1A participates in complex activation by nucleation-promoting factors (NPFs); (4) Branched network formation - the intact complex with ARPC1A mediates formation of branched actin networks by initiating daughter filaments at 70° angles from mother filaments; (5) Isoform specificity - ARPC1A may provide tissue-specific or developmental stage-specific functions compared to its homolog ARPC1B; (6) Regulatory interactions - ARPC1A likely participates in protein-protein interactions that regulate Arp2/3 complex activity . These mechanisms collectively contribute to cytoskeletal reorganization necessary for cellular processes like migration and invasion.

What experimental approaches can distinguish between ARPC1A and ARPC1B functions in cell models?

To distinguish ARPC1A from ARPC1B functions: (1) Isoform-specific knockdown - use siRNA or shRNA targeting unique regions of each isoform with verification of specificity by qRT-PCR and Western blot; (2) CRISPR/Cas9 gene editing - generate single and double knockout cell lines for comparative functional assays; (3) Rescue experiments - re-express one isoform in double knockout cells to identify specific functions; (4) Isoform-specific antibodies - use highly validated antibodies that distinguish between the similar proteins; (5) Domain swapping - create chimeric proteins to identify functionally important regions; (6) Tissue-specific expression analysis - examine differential expression patterns across tissues to identify contexts where one isoform predominates; (7) Interactome analysis - perform co-immunoprecipitation followed by mass spectrometry to identify isoform-specific binding partners . These approaches have demonstrated that while both proteins may function as p41 subunits of Arp2/3, they likely have distinct roles in different cellular contexts.

What experimental evidence supports the role of ARPC1A in cancer cell migration and invasion?

Multiple lines of evidence support ARPC1A's role in cancer metastasis: (1) Knockdown studies - ARPC1A siRNA/shRNA significantly reduced migration and invasion of prostate cancer cells in transwell assays without affecting proliferation; (2) Cytoskeletal analysis - immunofluorescence imaging revealed that ARPC1A-knockdown cells exhibit reduced actin filament formation and altered cytoskeletal architecture; (3) In vivo metastasis models - ARPC1A overexpression promoted lung metastasis in mouse models without affecting primary tumor growth; (4) Clinical correlations - high ARPC1A expression correlates with lymph node metastasis and poor prognosis in prostate cancer patients; (5) Mechanistic studies - glutamine metabolism was identified as an upstream regulator of ARPC1A, promoting migration and invasion through cytoskeletal remodeling; (6) Multivariate analysis - ARPC1A was confirmed as an independent prognostic factor for biochemical recurrence after radical prostatectomy (hazard ratio: 1.581) . These findings collectively establish ARPC1A as a key regulator of the metastatic phenotype in cancer cells.

How can ARPC1A expression be quantified for prognostic studies in cancer research?

For rigorous quantification in prognostic studies: (1) Tissue microarray (TMA) approach - standardize analysis using TMA containing hundreds of patient samples (301 cases were analyzed in published work) ; (2) Immunohistochemical scoring - implement a standardized scoring system (e.g., H-score combining intensity and percentage of positive cells); (3) Digital pathology - use automated image analysis software for objective quantification; (4) Multiple observers - involve at least two pathologists for independent scoring; (5) Statistical analysis - employ receiver operating characteristic (ROC) curve analysis to determine optimal cutoff values for stratifying patients (AUC of 0.775 was reported for ARPC1A in predicting biochemical recurrence) ; (6) Multivariate models - incorporate ARPC1A expression with established clinicopathological parameters in Cox proportional hazard regression models; (7) Independent validation - confirm findings in separate patient cohorts (both TMA and TCGA cohorts yielded consistent results) . This comprehensive approach has established ARPC1A as an independent prognostic factor in prostate cancer.

What methodological considerations are important when investigating ARPC1A as a therapeutic target in cancer?

When investigating ARPC1A as a therapeutic target: (1) Target validation - confirm ARPC1A overexpression in patient samples and correlation with aggressive phenotypes; (2) Functional dependency - demonstrate that cancer cells depend on ARPC1A for migration/invasion using knockdown/knockout approaches; (3) Rescue experiments - confirm phenotypic effects are specifically due to ARPC1A loss; (4) Combination strategies - test ARPC1A targeting alongside standard therapies to identify synergistic effects; (5) Selective inhibition - develop approaches that target ARPC1A without affecting ARPC1B to minimize toxicity; (6) Delivery strategies - for RNA-based therapeutics, optimize delivery vehicles for tumor-specific targeting; (7) Pharmacodynamic markers - identify downstream effects that can serve as markers of successful target engagement; (8) Resistance mechanisms - investigate potential compensatory pathways that might emerge following ARPC1A inhibition . Research has demonstrated that glutamine metabolism regulates ARPC1A, suggesting metabolic interventions might indirectly modulate ARPC1A function.

How should researchers approach the development of ARPC1A-based biomarker assays for clinical applications?

For biomarker assay development: (1) Analytical validation - establish assay sensitivity, specificity, reproducibility, and dynamic range using standardized protocols; (2) Pre-analytical variables - determine impact of tissue fixation, processing time, and storage conditions on ARPC1A detection; (3) Reference standards - develop calibrated reference materials for consistent quantification; (4) Threshold determination - define clinically relevant cutoff values through ROC analysis (published data showed specificity of 74.0% and sensitivity of 77.4% for biochemical recurrence prediction) ; (5) Complementary markers - evaluate ARPC1A in conjunction with established biomarkers to improve predictive accuracy; (6) Multicenter validation - test assay performance across different laboratories; (7) Standardized reporting - develop clear reporting guidelines for interpreting results; (8) Clinical utility validation - demonstrate that ARPC1A testing improves clinical decision-making in prospective studies . This approach would build on findings that ARPC1A is an independent prognostic factor for biochemical recurrence after radical prostatectomy.

What are the methodological approaches to study post-translational modifications of ARPC1A?

To investigate ARPC1A post-translational modifications: (1) Phospho-specific antibodies - develop or utilize antibodies that recognize specific phosphorylation sites on ARPC1A; (2) Mass spectrometry analysis - perform liquid chromatography-tandem mass spectrometry (LC-MS/MS) on immunoprecipitated ARPC1A to identify modification sites; (3) In vitro kinase assays - identify kinases that modify ARPC1A; (4) Phosphomimetic mutations - generate point mutations that mimic constitutive phosphorylation (e.g., S→D) or prevent phosphorylation (e.g., S→A); (5) Functional correlation - correlate modifications with ARPC1A activity in actin polymerization assays; (6) Temporal dynamics - study how modifications change during cell migration or cancer progression; (7) Inhibitor studies - use specific inhibitors of post-translational modifying enzymes to determine functional consequences . These approaches would extend current knowledge beyond the basic characterization of ARPC1A to understand its regulation in normal and disease states.

How can researchers investigate the role of ARPC1A in three-dimensional invasion models that better recapitulate the tumor microenvironment?

For studying ARPC1A in 3D invasion models: (1) Spheroid formation - establish spheroids from cancer cells with modulated ARPC1A expression; (2) Organoid cultures - develop patient-derived organoids that maintain tissue architecture; (3) Matrix composition - test invasion through different ECM components (Matrigel, collagen) to evaluate matrix-specific effects; (4) Live-cell imaging - perform time-lapse microscopy with fluorescently-tagged ARPC1A to visualize dynamics during invasion; (5) Second harmonic generation imaging - visualize collagen remodeling during ARPC1A-dependent invasion; (6) Co-culture systems - incorporate stromal cells to study heterotypic interactions; (7) Quantitative analysis - implement automated image analysis for objective quantification of invasion parameters; (8) Drug response - test how targeting ARPC1A affects response to standard therapies in 3D models . This approach would extend the 2D migration/invasion findings that demonstrated ARPC1A knockdown inhibits these processes in prostate cancer cells.

What strategies can be employed to study the interactome of ARPC1A in the context of cancer progression?

To characterize the ARPC1A interactome: (1) Co-immunoprecipitation coupled with mass spectrometry - identify proteins that physically interact with ARPC1A in normal vs. cancer cells; (2) Proximity labeling - use BioID or APEX approaches to identify proteins in close proximity to ARPC1A in living cells; (3) Yeast two-hybrid screening - identify direct protein-protein interactions; (4) Protein microarrays - screen for interactions with specific protein families; (5) Fluorescence resonance energy transfer (FRET) - visualize interactions in living cells; (6) Cross-linking mass spectrometry - identify interaction interfaces; (7) Comparative interactomics - analyze how the interactome changes during cancer progression or in response to treatments; (8) Bioinformatic analysis - employ network analysis to identify critical nodes and pathways . Research has already identified glutamine metabolism as an upstream regulator of ARPC1A in prostate cancer, and further interactome studies could reveal additional regulatory mechanisms and therapeutic opportunities.

What are the common challenges in detecting ARPC1A by Western blot and how can they be addressed?

Common challenges and solutions include: (1) Non-specific bands - use higher antibody dilutions (1:1000-1:3000) and more stringent washing conditions; verify specificity with knockdown controls; (2) Weak signal - increase protein loading to 35-50 μg; optimize antibody concentration; extend primary antibody incubation time to overnight at 4°C; (3) High background - increase blocking time or concentration; use fresh blocking reagents; ensure thorough washing between steps; (4) Variable band size - ARPC1A can appear between 37-42 kDa; use appropriate molecular weight markers and positive controls (HEK-293 or PC-3 cells); (5) Degradation - include protease inhibitors in lysis buffer; keep samples cold; avoid repeated freeze-thaw cycles; (6) Membrane optimization - PVDF membranes are generally preferable for ARPC1A detection; (7) Detection method - ECL has been successfully used but more sensitive detection methods may be required for low expression samples . These approaches have been validated in multiple studies detecting ARPC1A in various cell types.

How can researchers address challenges when comparing ARPC1A expression across different experimental models?

For consistent cross-model comparisons: (1) Reference standards - include common positive control samples across all experiments; (2) Normalization strategy - use multiple housekeeping controls appropriate for each experimental system; (3) Batch effects - process samples simultaneously when possible or include inter-batch controls; (4) Protocol standardization - maintain identical protocols for sample preparation, antibody dilutions, and detection methods; (5) Antibody lot consistency - use the same antibody lot when possible or validate new lots against previous ones; (6) Species considerations - when comparing across species, confirm antibody cross-reactivity experimentally; (7) Quantification methods - standardize image acquisition settings and quantification algorithms; (8) Statistical approach - employ appropriate statistical tests that account for inter-experimental variation . This systematic approach enables reliable comparisons between different cell lines, tissues, or animal models.

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.