PRMT2, also known as HRMT1L1, is a member of the arginine methyltransferase family that catalyzes arginine methylation and regulates diverse cellular processes including transcription, translation, DNA repair, and RNA processing. It is responsible for the H3R8me2a histone mark, which is associated with active transcription . PRMT2 interacts with many key regulatory proteins including Rb, NF-κB, ER-alpha, and androgen receptor, positioning it as a critical player in various cellular pathways . Recent research has implicated PRMT2 in several disease processes, including its overexpression in cancers such as hepatocellular carcinoma, glioblastoma, and renal cell carcinoma . Additionally, PRMT2 has been identified as a host restriction factor for HIV-1 transcription, promoting viral latency through methylation of the viral Tat protein .
PRMT2 antibodies are versatile tools employed across multiple experimental platforms:
These applications enable researchers to detect PRMT2 expression, localization, and interactions in various experimental systems, including cell lines, tissue samples, and protein extracts .
The choice between monoclonal and polyclonal PRMT2 antibodies depends on your specific research requirements:
Monoclonal antibodies (e.g., 66885-1-Ig) offer:
Superior lot-to-lot consistency
Higher specificity for a single epitope
Reduced background in applications like immunofluorescence
Ideal for quantitative assays and detecting specific epitopes
Polyclonal antibodies offer:
Recognition of multiple epitopes on PRMT2
Often higher sensitivity in applications like Western blotting
More robust detection across different species
PRMT2 typically appears at 45-50 kDa in Western blot applications, which is slightly lower than its calculated molecular weight of 49 kDa . This slight discrepancy between calculated and observed molecular weights is common for many proteins due to post-translational modifications, protein folding, or the specific gel system used. Some splice variants of PRMT2 have been identified in breast cancers, which may appear at different molecular weights . When validating a new PRMT2 antibody, researchers should compare their observed band against both positive controls (e.g., SKOV-3 cells, HeLa cells, Jurkat cells) and negative controls (e.g., PRMT2 knockout or knockdown samples) .
Multiple validated positive controls have been documented for PRMT2 antibody applications:
For Western blotting:
For immunohistochemistry:
Human thyroid cancer tissue (with recommended antigen retrieval using TE buffer pH 9.0 or citrate buffer pH 6.0)
For immunofluorescence:
When establishing a new experimental system, these validated sources provide reliable positive controls for antibody optimization.
Rigorous validation ensures reliable PRMT2 detection across applications:
Knockdown/knockout validation: Compare detection between wild-type samples and those with PRMT2 knockdown using siRNA (e.g., PRMT2-specific siRNA-A from Invitrogen or siRNA-B from Sigma) or CRISPR/Cas9 knockout . Complete signal loss in knockout samples confirms specificity.
Overexpression controls: Compare detection in cells with endogenous PRMT2 versus those overexpressing PRMT2 to confirm proper molecular weight and signal increase .
Cross-reactivity assessment: Test the antibody against samples from multiple species if cross-reactivity is claimed. Many PRMT2 antibodies show reactivity with human, mouse, and rat samples .
Peptide competition: Pre-incubate the antibody with the immunizing peptide before application to confirm that the peptide blocks specific binding .
Multiple antibody validation: Compare results using antibodies recognizing different epitopes of PRMT2 (e.g., N-terminal versus C-terminal) .
Effective sample preparation is crucial for successful PRMT2 detection:
For Western blotting:
Use RIPA buffer or NP-40 buffer with protease inhibitors
Include phosphatase inhibitors if studying PRMT2 phosphorylation
Add deacetylase inhibitors if studying acetylation status
Include 1-5% SDS for membrane protein solubilization
For immunofluorescence:
4% paraformaldehyde fixation (10-15 minutes at room temperature)
Permeabilization with 0.1-0.5% Triton X-100
Blocking with 5% normal serum corresponding to secondary antibody species
For immunohistochemistry:
Antigen retrieval with TE buffer pH 9.0 is recommended
Alternative: citrate buffer pH 6.0
Proper handling ensures maximum antibody performance and longevity:
For optimal performance, allow the antibody to equilibrate to room temperature before opening the vial, and briefly centrifuge before use to collect the solution at the bottom of the tube .
Recent research has identified PRMT2 as a host restriction factor for HIV-1 transcription and proviral reactivation . Researchers studying this mechanism can employ PRMT2 antibodies in several sophisticated approaches:
Detecting PRMT2-Tat interactions: Co-immunoprecipitation using PRMT2 antibodies can pull down HIV-1 Tat protein to study their physical association. Studies have shown that Tat is physically associated with PRMT2 and preferentially methylated at the R52 residue .
Mapping nucleolar localization: Immunofluorescence co-staining with PRMT2 antibodies and nucleolar markers like NPM1 can visualize how PRMT2 enhances Tat association with NPM1, causing its nucleolar sequestration .
Analyzing methylation status: Using custom antibodies that specifically recognize R52ame2 Tat (such as TatR52ame2_2) alongside PRMT2 antibodies helps track the methylation status of Tat in latently infected cells .
Monitoring latency establishment: In dual-color HIV-1 reporter systems, correlating PRMT2 levels with GFP/mKO2 expression can demonstrate how PRMT2 promotes latency establishment during viral infection .
Evaluating latency reversal agents (LRAs): PRMT2 antibodies can assess how PRMT2 inhibition might synergize with existing LRAs to reactivate latent provirus in patient-derived CD4+ T cells .
This approach has revealed that the methylase activity of PRMT2 is critical for inhibiting Tat transactivation and maintaining proviral latency, suggesting potential for PRMT2 inhibitors in HIV cure strategies .
PRMT2 has been implicated in several cancer types, with antibody-based studies revealing key mechanisms:
In renal cell carcinoma (RCC):
PRMT2 is upregulated in primary RCC and RCC cell lines
Overexpression promotes RCC cell proliferation and motility both in vitro and in vivo
PRMT2 mediates H3R8 asymmetric dimethylation (H3R8me2a) in the WNT5A promoter region
This enhances WNT5A transcriptional expression, activating Wnt signaling pathway
In other cancer types:
PRMT2 is overexpressed in hepatocellular carcinoma and glioblastoma
Splice variants of PRMT2 have been described in breast cancers
Antibody-based methodologies to study these mechanisms include:
Chromatin immunoprecipitation (ChIP) using PRMT2 antibodies to identify genomic targets
Immunohistochemistry to correlate PRMT2 expression with clinical outcomes
Co-immunoprecipitation to study interaction partners in cancer contexts
Immunoblotting of tumor samples to quantify expression across cancer stages
These approaches have revealed PRMT2 as a potential therapeutic target in multiple cancer types through its epigenetic regulatory functions.
PRMT2 catalyzes the H3R8me2a (histone H3 asymmetric dimethylarginine 8) mark, which is associated with active transcription . Detecting this specific modification requires a combination of PRMT2 antibodies and modification-specific antibodies:
ChIP-seq approach:
Sequential ChIP (re-ChIP):
First immunoprecipitate with PRMT2 antibodies
Perform a second immunoprecipitation on the eluted material using H3R8me2a antibodies
This identifies genomic regions where PRMT2 directly mediates H3R8me2a modification
Immunofluorescence co-localization:
Co-stain cells with PRMT2 antibodies and H3R8me2a antibodies
Analyze co-localization patterns using confocal microscopy
Quantify co-localization coefficients across different cellular conditions
When conducting these experiments, include appropriate controls such as PRMT2 knockdown/knockout samples to confirm the specificity of the detected modifications .
PRMT2 splice variants have been described particularly in breast cancers , and differentiating between these variants requires careful antibody selection and experimental design:
Epitope-specific antibodies:
Select antibodies targeting regions that are differentially present in splice variants
For N-terminal variants, use antibodies targeting amino acids 1-300 or 22-53
For C-terminal variants, use antibodies targeting amino acids 344-375
Compare detection patterns across multiple antibodies recognizing different regions
Immunoprecipitation followed by mass spectrometry:
Immunoprecipitate PRMT2 using antibodies that recognize all variants
Analyze the precipitated proteins by mass spectrometry to identify peptides unique to specific variants
Quantify the relative abundance of variant-specific peptides
RT-PCR validation:
Complement antibody-based detection with RT-PCR using variant-specific primers
Correlate protein detection with mRNA expression profiles
This dual approach provides stronger evidence for variant expression
Size-based discrimination:
Use high-resolution SDS-PAGE to separate variants by molecular weight
Different variants may show distinct migration patterns due to size differences
Follow with Western blotting using pan-PRMT2 antibodies to detect all variants simultaneously
These approaches can help researchers understand the differential expression and functions of PRMT2 splice variants in normal and disease states.
Optimizing PRMT2 antibodies for ChIP requires careful consideration of several factors:
Antibody selection:
Crosslinking optimization:
Test different formaldehyde concentrations (0.5-1.5%) and crosslinking times (5-15 minutes)
For indirect interactions, consider dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde
Sonication parameters:
Optimize sonication conditions to yield chromatin fragments of 200-500 bp
Verify fragment size by agarose gel electrophoresis before proceeding
Antibody concentration titration:
Perform preliminary ChIP with different antibody amounts (2-10 μg per reaction)
Analyze enrichment by qPCR at known PRMT2 binding sites
The optimal concentration should maximize signal-to-noise ratio
Positive control loci:
Pre-clearing and blocking:
Pre-clear chromatin with protein A/G beads to reduce background
Include sufficient blocking agents (BSA, salmon sperm DNA) to minimize non-specific binding
Washing stringency:
Test different washing buffer stringencies to optimize signal-to-noise ratio
More stringent washes reduce background but may reduce signal from weak interactions
This optimized protocol will enable reliable mapping of PRMT2 genomic binding sites and correlation with its methyltransferase activity.
Co-immunoprecipitation (co-IP) of PRMT2 and its binding partners presents several technical challenges:
Preserving native interactions:
Nuclear protein extraction:
Antibody orientation:
Distinguishing direct vs. indirect interactions:
Include RNase/DNase treatment to eliminate nucleic acid-mediated associations
Consider using crosslinking approaches to stabilize transient interactions
Validate direct interactions using purified recombinant proteins
Detecting methylated partners:
Verifying specificity:
Include appropriate negative controls (IgG, PRMT2 knockout/knockdown)
Confirm interactions using multiple antibodies targeting different epitopes of PRMT2
Validate key interactions with orthogonal methods (proximity ligation assay, FRET)
By addressing these challenges, researchers can reliably identify and characterize the interactome of PRMT2 in various biological contexts.
As potential PRMT2 inhibitors are developed, antibody-based approaches will be crucial for evaluating their efficacy:
Target engagement assays:
Cellular thermal shift assay (CETSA) using PRMT2 antibodies can confirm inhibitor binding to PRMT2 in cells
Drug affinity responsive target stability (DARTS) can assess protective effects of inhibitors against protease digestion
Functional inhibition assessment:
Pathway modulation:
Phenotypic reversal:
Resistance mechanism identification:
Immunoprecipitation followed by mass spectrometry can identify changes in PRMT2 interaction partners upon inhibitor treatment
These analyses may reveal compensatory mechanisms or resistance pathways
Although selective PRMT2 inhibitors are not yet commercially available, the structural information from zebrafish and mouse PRMT2 should facilitate inhibitor development, with antibody-based approaches being essential for their evaluation .
The PRMT family contains nine members with similar catalytic domains, creating potential cross-reactivity challenges:
Epitope selection:
Validation in knockout systems:
Immunodepletion strategy:
Sequentially deplete lysates with antibodies against other PRMT family members before PRMT2 detection
This approach can help isolate PRMT2-specific signals from potential cross-reactive signals
Specific methylation patterns:
Focus on PRMT2-specific methylation targets (H3R8me2a) rather than shared targets
Compare methylation patterns with those of other PRMTs (e.g., PRMT1-mediated H4R3me2a)
This functional readout can complement direct PRMT2 detection
Isoform-specific assays:
These methodological approaches allow researchers to distinguish PRMT2-specific signals from those of other PRMT family members, ensuring reliable data interpretation in complex biological systems.