ARID4A interacts with the retinoblastoma protein (RB) and recruits histone deacetylases (HDACs) to repress E2F-dependent transcription, regulating cell cycle arrest .
ARID4B collaborates with HDAC1 to modulate chromatin remodeling during embryonic stem cell differentiation, influencing lineage-specific gene expression .
In male fertility, ARID4A/ARID4B double-deficient mice exhibit spermatogenic arrest and impaired blood-testis barrier integrity due to dysregulation of androgen receptor (AR) and RB pathways .
ARID4A acts as a tumor suppressor in colorectal cancer and leukemia. Loss of ARID4A in mice leads to myelodysplastic disorders and acute myeloid leukemia (AML) .
In breast cancer, high ARID4B expression correlates with poor prognosis, particularly in HER2-positive and lymph node-positive patients .
DNA Binding: ARID4A binds DNA without sequence specificity, relying on structural motifs like the Helix 4 tryptophan residue for chromatin interactions .
Protein Interactions:
Mechanistic Insights:
Therapeutic Potential:
ARID4 belongs to the AT-rich interaction domain (ARID) family, which includes 15 members divided into seven subfamilies based on sequence identity. The ARID4 subfamily specifically contains two members: ARID4A and ARID4B. These proteins function as transcription regulators affecting cell growth, differentiation, and development . ARID4B, also known as BRCAA1 or SAP180, is a 1312 amino acid protein containing one ARID domain and localizes in both the nucleus and cytoplasm . ARID4 proteins interact with the retinoblastoma protein (pRB) and the mSIN3-histone deacetylase (mSIN3-HDAC) complex, playing crucial roles in cellular processes including transcriptional regulation and growth control . Their involvement in cancer-related signaling pathways makes them important targets for oncology research.
ARID4A contains an additional LXCXE motif, which functions as an RB binding motif not present in ARID4B
ARID4A represses E2F-dependent transcription by recruiting the mSIN3-HDAC complex to pRB family members, playing a central role in arresting cell growth
ARID4B has been found to localize in both nucleus and cytoplasm, suggesting potentially distinct functions in different cellular compartments
The genes are located on different chromosomes: ARID4A on 14q23.1 and ARID4B on 1q42.1-q43
These differences explain their distinct roles in cellular processes and why researchers might target one over the other depending on the biological question.
ARID4B exhibits tissue-specific expression patterns that researchers should consider when designing experiments. According to available data, ARID4B is:
Highly expressed in testis and in breast, lung, colon, pancreatic and ovarian cancers
Detected in various cell lines including MCF-7 (breast cancer), A549 (lung cancer), and K-562 (leukemia) cells
Understanding these expression patterns is crucial when selecting appropriate experimental models and interpreting results. For instance, using cell lines with known ARID4B expression levels as positive controls can help validate antibody specificity and experimental conditions.
Based on extensive validation data, ARID4B antibodies have been successfully applied in multiple experimental techniques. The polyclonal antibody (24499-1-AP) has been validated for:
| Application | Recommended Dilution | Validated Samples |
|---|---|---|
| Western Blot (WB) | 1:500-1:1000 | MCF-7 cells, A549 cells, K-562 cells |
| Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate | MCF-7 cells |
| Immunohistochemistry (IHC) | 1:500-1:2000 | Human pancreas cancer tissue, breast cancer tissue, cervical cancer tissue, testis tissue |
| Immunofluorescence (IF)/ICC | 1:200-1:800 | MCF-7 cells |
| ELISA | Validated | Human samples |
For the monoclonal antibody (67384-1-PBS), validated applications include:
Researchers should always optimize these conditions for their specific experimental systems to obtain optimal results.
Antigen retrieval is a critical step for successful ARID4B immunohistochemistry. Based on experimental validation:
The recommended primary protocol involves using TE buffer at pH 9.0 for antigen retrieval . This alkaline pH has been shown to effectively unmask ARID4B epitopes in formalin-fixed, paraffin-embedded tissues including pancreatic cancer, breast cancer, cervical cancer, and testis tissues.
As an alternative method, researchers can perform antigen retrieval using citrate buffer at pH 6.0 . When optimizing antigen retrieval:
Compare both TE and citrate buffer methods side-by-side to determine which provides better signal-to-noise ratio
Ensure appropriate incubation time (typically 15-20 minutes) at optimal temperature
Include positive control tissues (such as testis or breast cancer samples) known to express ARID4B
Run parallel negative controls (omitting primary antibody) to assess background staining
Optimization of antigen retrieval conditions is particularly important for tissues with high fixation variability or when working with archived samples.
Ensuring ARID4B antibody specificity is crucial for generating reliable research data. Recommended validation approaches include:
Molecular weight verification: ARID4B has a calculated molecular weight of 148 kDa (1312 amino acids), while observed bands typically appear at 100 kDa and 200 kDa . Discrepancies between calculated and observed weights may be due to post-translational modifications or protein processing.
Knockdown/knockout controls: Utilizing ARID4B knockdown or knockout samples as negative controls. Published applications have validated antibody specificity using KD/KO systems .
Multiple antibody approach: Compare results using different antibodies targeting distinct epitopes of ARID4B (such as comparing monoclonal and polyclonal antibodies).
Recombinant protein controls: Use purified recombinant ARID4B protein as a positive control in Western blot experiments.
Tissue/cell type controls: Include samples with known ARID4B expression levels. MCF-7 cells, A549 cells, and K-562 cells have been validated as positive controls for ARID4B expression .
Cross-reactivity assessment: Test antibody reactivity against related proteins, particularly ARID4A which shares sequence similarity with ARID4B.
These validation steps should be performed before conducting critical experiments to ensure reliability of results and accurate interpretation of data.
Investigating ARID4B's role in cancer requires a multifaceted approach:
Expression analysis in cancer tissues: Compare ARID4B expression between tumor and matched normal tissues. ARID4B is highly expressed in breast, lung, colon, pancreatic, and ovarian cancers , suggesting potential oncogenic functions in these tissues.
Mechanistic studies: Analyze ARID4B's interactions with key cancer-related pathways:
Examine its role in the Sin3A corepressor complex, where it may function in transcriptional repression
Investigate its involvement in cell cycle regulation and proliferation
Study its interaction with other transcription factors
Functional studies using genetic manipulation:
Use RNA interference (siRNA/shRNA) to knock down ARID4B expression
Employ CRISPR-Cas9 to create ARID4B knockout cell lines
Perform rescue experiments with wild-type or mutant ARID4B
Animal models: Develop ARID4B knockout or transgenic mouse models to study its role in tumorigenesis in vivo.
Clinical correlation: Analyze ARID4B expression in patient samples and correlate with clinical outcomes, response to therapy, and other clinical parameters.
By integrating these approaches, researchers can comprehensively investigate ARID4B's functional roles in cancer initiation, progression, and potential as a therapeutic target.
Studying ARID4B protein-protein interactions presents several challenges:
Nuclear localization: ARID4B localizes to both nucleus and cytoplasm, with nuclear ARID4B being less stable due to rapid degradation by the ubiquitin-proteasome system . This differential stability complicates interaction studies.
Technical challenges:
The large size of ARID4B (1312 amino acids) can make recombinant protein production difficult
Multiple protein isoforms may exhibit different interaction profiles
Transient or weak interactions may be missed by standard approaches
Solutions and methodologies:
Optimized immunoprecipitation: Use crosslinking agents to stabilize transient interactions. The validated protocol recommends 0.5-4.0 μg of antibody for 1.0-3.0 mg of total protein lysate .
Proximity labeling approaches: BioID or APEX2 fusion proteins can identify proximal proteins regardless of interaction strength.
Mass spectrometry-based approaches: Analyze ARID4B interactome using quantitative proteomics after immunoprecipitation.
Subcellular fractionation: Separate nuclear and cytoplasmic fractions before performing interaction studies to account for compartment-specific interactions.
Domain-specific interaction mapping: Generate constructs expressing specific ARID4B domains to map domain-specific interactions.
Verification approaches:
Confirm interactions using multiple techniques (co-IP, proximity ligation assay, FRET)
Validate physiological relevance through functional assays
Perform reciprocal IPs to strengthen evidence for interactions
These strategies can help overcome the inherent challenges of studying ARID4B's protein interaction network.
ARID4B possesses distinct characteristics compared to other ARID family members:
Structural differences:
Functional roles:
While many ARID family members (like ARID1A) act as tumor suppressors, ARID4B shows context-dependent functions
ARID4B may function in the assembly and/or enzymatic activity of the Sin3A corepressor complex
Unlike some ARID family members that are strictly nuclear, ARID4B localizes to both nucleus and cytoplasm
Expression patterns:
Cancer relevance:
Understanding these differences is crucial for researchers targeting specific ARID family members and interpreting the results of ARID4B-focused experiments in the broader context of ARID family functions.
Researchers frequently encounter several challenges when working with ARID4B antibodies:
Multiple bands in Western blot:
Weak or absent signal:
High background:
Potential causes: Non-specific binding, excessive antibody concentration, inadequate blocking
Resolution: Increase blocking time, optimize antibody dilution, use additional washing steps, and consider alternative blocking agents
Variability between experiments:
Resolution: Standardize protocols, prepare fresh working solutions, aliquot antibodies to avoid freeze-thaw cycles, and use consistent positive controls
Storage and stability issues:
Following these troubleshooting approaches can significantly improve experimental outcomes when working with ARID4B antibodies.
Successful immunofluorescence experiments with ARID4B antibodies require careful optimization:
Cell line selection:
Fixation and permeabilization:
ARID4B localizes to both nucleus and cytoplasm, requiring balanced fixation and permeabilization
Test both cross-linking (paraformaldehyde) and precipitating (methanol) fixatives
Optimize permeabilization conditions to ensure antibody accessibility to nuclear ARID4B
Antibody concentration and incubation:
Controls and validation:
Include positive and negative controls in each experiment
Consider co-staining with nuclear markers to verify subcellular localization
Perform ARID4B knockdown to confirm specificity of the signal
Image acquisition and analysis:
Capture both nuclear and cytoplasmic signals due to ARID4B's dual localization
Consider z-stack imaging to fully capture the 3D distribution
Quantify nuclear vs. cytoplasmic distribution as this may have functional relevance
These optimization steps will enhance the quality and reliability of ARID4B immunofluorescence experiments, enabling better visualization of this protein's subcellular distribution.
The choice between monoclonal (67384-1-PBS) and polyclonal (24499-1-AP) ARID4B antibodies depends on specific experimental needs:
Application compatibility:
Epitope recognition:
Polyclonal antibodies recognize multiple epitopes, potentially providing stronger signals
Monoclonal antibodies offer higher specificity for a single epitope, reducing background
Experimental reproducibility:
Monoclonal antibodies provide consistent lot-to-lot reproducibility
Polyclonal antibodies may show batch variation but can be more robust against epitope changes
Storage requirements:
Buffer compatibility:
Research question considerations:
For detecting specific ARID4B isoforms or phosphorylation states, monoclonal antibodies may be preferred
For maximum sensitivity in detecting low abundance protein, polyclonal antibodies may provide advantages
Researchers should select the appropriate antibody based on these considerations and their specific experimental requirements.
ARID4B's involvement in epigenetic regulation offers promising research directions:
Chromatin immunoprecipitation (ChIP) approaches:
Study ARID4B in the context of the Sin3A corepressor complex:
Transcriptional repression assays:
Use reporter assays to measure ARID4B's impact on gene expression
Analyze changes in the transcriptome after ARID4B depletion or overexpression
Investigate how ARID4B interacts with transcription factors and epigenetic machinery
Cancer epigenetics:
Explore how ARID4B contributes to altered epigenetic landscapes in cancer
Investigate potential interactions with DNA methylation machinery
Examine ARID4B's role in maintaining cancer-specific gene expression patterns
Single-cell approaches:
Apply single-cell technologies to understand cell-to-cell variation in ARID4B-mediated epigenetic regulation
Investigate how ARID4B contributes to epigenetic heterogeneity in tumors
These approaches will advance our understanding of ARID4B's role in epigenetic processes and potentially identify new therapeutic targets in diseases with dysregulated epigenetic pathways.
Emerging research suggests several avenues for targeting ARID4B in cancer therapy:
Expression patterns relevant to therapeutic development:
Potential therapeutic approaches:
Small molecule inhibitors: Design compounds that disrupt ARID4B's interaction with the Sin3A corepressor complex
Degradation-based approaches: Develop proteolysis-targeting chimeras (PROTACs) specific for ARID4B
Gene silencing strategies: Utilize siRNA or antisense oligonucleotides to reduce ARID4B expression
Functional antibodies: Develop antibodies that can modulate ARID4B function in vivo
Biomarker potential:
Combination therapy approaches:
Investigate synergistic effects of ARID4B targeting with existing epigenetic drugs (HDAC inhibitors, DNA methyltransferase inhibitors)
Study how ARID4B inhibition affects sensitivity to conventional chemotherapeutics
Target validation:
Use ARID4B antibodies to evaluate on-target effects of experimental therapeutics
Employ genetic approaches (CRISPR-Cas9) to validate ARID4B as a therapeutic target
Study compensatory mechanisms that may arise upon ARID4B inhibition
These approaches offer promising directions for translating ARID4B research into clinical applications, potentially expanding the repertoire of targeted therapies for cancers with ARID4B dysregulation.
Despite significant advances in understanding ARID4B function, several limitations and knowledge gaps remain:
Technical limitations:
Current antibodies may not distinguish between ARID4B splice variants or post-translationally modified forms
Limited availability of highly specific inhibitors or activators for functional studies
Challenges in studying ARID4B in complex with other proteins due to its size and multi-domain structure
Knowledge gaps:
Incomplete characterization of ARID4B genomic binding sites across different cell types
Limited understanding of how ARID4B contributes to tissue-specific gene regulation
Unclear mechanistic details of how ARID4B mediates interactions between different regulatory complexes
Insufficient data on ARID4B mutations and their functional consequences in different cancer types
Future research priorities:
Develop more selective antibodies capable of distinguishing ARID4B isoforms and modifications
Expand ChIP-seq and other genomic approaches to map ARID4B binding sites across diverse cell types
Investigate ARID4B's role in normal development and tissue homeostasis
Explore therapeutic potential through development of specific inhibitors
Elucidate the structural basis of ARID4B interactions with DNA and protein partners
Methodological advancements needed:
Improved protocols for studying chromatin-associated proteins in native complexes
Better animal models to study ARID4B function in development and disease
Advanced imaging techniques to visualize ARID4B dynamics in living cells
Addressing these limitations and priorities will advance our understanding of ARID4B biology and its potential therapeutic applications in cancer and other diseases.