PRMT10 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview

PRMT10 Antibody is a polyclonal or monoclonal immunoglobulin designed to specifically recognize and bind to Protein Arginine Methyltransferase 10 (PRMT10), an enzyme involved in post-translational modification of proteins through arginine methylation. PRMT10 belongs to the class I-like SAM-binding methyltransferase superfamily and is implicated in alternative splicing regulation, flagellar assembly, and epigenetic control .

Critical Notes:

  • Antibody Specificity: Cross-reactivity with PRMT9 is common due to conserved domains .

  • Control Experiments: Use recombinant PRMT10 protein or siRNA knockdown to confirm specificity .

Research Findings and Functional Insights

PRMT10 antibodies have elucidated critical roles of PRMT10 in diverse biological processes:

**4.1 Methylation-Driven Processes

ProcessSubstrateFunctional Impact
Alternative SplicingSF3B2Symmetric dimethylation (sDMA) of SF3B2 regulates pre-mRNA splicing .
Flagellar AssemblyAxonemal ProteinsTransport via IFT particles; localization at flagellar tips/bases .
Epigenetic RegulationHistone H4 (Arabidopsis)PRMT10 methylates H4R3, influencing flowering time .

**4.2 Disease Associations

  • Cancer: Overexpression of type I PRMTs (including PRMT10) correlates with colorectal cancer progression. Substrates like CACYBP and MAPRE1 show elevated methylation in CRC .

  • Neurological Disorders: PRMT10’s interaction with hnRNPA1 may influence RNA metabolism, though direct links remain under investigation .

Technical Considerations

  • Optimal Dilutions:

    ApplicationDilution
    IHC-P1–4 µg/ml
    ICC/IF1:200
    WB1:1,000
  • Blocking Buffers: Use 5% BSA or milk to reduce non-specific binding.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
PRMT10 antibody; PRMT4.2 antibody; At1g04870 antibody; F13M7.14 antibody; F13M7_12Protein arginine N-methyltransferase PRMT10 antibody; EC 2.1.1.319 antibody
Target Names
PRMT10
Uniprot No.

Target Background

Function
This antibody targets PRMT10, an enzyme that catalyzes the mono- and asymmetric dimethylation of arginine residues in certain proteins. PRMT10 plays a crucial role in regulating flowering time.
Gene References Into Functions
  1. The methyltransferase activity of AtPRMT10 is essential for suppressing FLC expression and promoting flowering in Arabidopsis. PMID: 22729397
  2. Studies have demonstrated that genetic disruption of AtPRMT10 leads to delayed flowering due to increased transcript levels of FLOWERING LOCUS C (FLC). [AtPRMT10] PMID: 18007657
Database Links

KEGG: ath:AT1G04870

STRING: 3702.AT1G04870.2

UniGene: At.12003

Protein Families
Class I-like SAM-binding methyltransferase superfamily, Protein arginine N-methyltransferase family

Q&A

What is PRMT10 and why is it significant in research?

PRMT10 (Protein Arginine Methyltransferase 10) is a member of the protein arginine methyltransferase family that catalyzes the methylation of arginine residues on target proteins. Its significance lies in its role as an androgen receptor (AR) interacting protein that is highly expressed in reproductive tissues, particularly the prostate. PRMT10 has been identified as playing a crucial role in androgen-dependent proliferation of prostate cancer cells through its interaction with AR signaling pathways. In research contexts, PRMT10 is studied for its potential implications in prostate cancer development and progression, as knockdown of PRMT10 has been shown to decrease dihydrotestosterone (DHT)-dependent cell growth in prostate cancer models . Additionally, some research has implicated PRMT10 homologs in plant systems (AtPRMT10) in the regulation of flowering time and growth, suggesting evolutionary conservation of certain methyltransferase functions across kingdoms .

What applications are PRMT10 antibodies validated for?

PRMT10 antibodies have been validated for several common laboratory applications. The most frequently validated applications include immunocytochemistry/immunofluorescence at concentrations of 1-4 μg/mL, which allows for subcellular localization studies of PRMT10 . Additionally, many commercially available PRMT10 antibodies are validated for Western blot (WB) analysis, which enables detection of PRMT10 protein in cell or tissue lysates . Some antibodies are also suitable for immunoprecipitation (IP), which can be useful for studying protein-protein interactions involving PRMT10, and enzyme-linked immunosorbent assay (ELISA) for quantitative detection . Immunohistochemistry (IHC) applications, both standard and paraffin-embedded (IHC-p), allow for detection of PRMT10 in tissue sections, which is particularly valuable for studying its expression patterns in normal and diseased tissues like prostate cancer samples .

How should PRMT10 antibodies be stored and handled to maintain reactivity?

For optimal preservation of PRMT10 antibody reactivity and specificity, proper storage and handling procedures must be followed. Most PRMT10 antibodies should be stored at 4°C for short-term use (days to weeks) . For long-term storage, aliquoting the antibody and storing at -20°C is recommended to prevent repeated freeze-thaw cycles that can degrade protein structure and reduce antibody performance . When working with PRMT10 antibodies, it's advisable to prepare small working aliquots to avoid repeated freeze-thaw cycles of the stock solution. PRMT10 antibodies are typically supplied in buffer solutions containing preservatives such as sodium azide (commonly at 0.02%) and stabilizers like glycerol (often at 40%) in PBS at pH 7.2 . When diluting these antibodies for experimental use, researchers should use fresh, high-quality buffer solutions that maintain protein stability. Additionally, following the manufacturer's specific recommendations for each antibody is crucial, as storage and handling requirements may vary slightly between different antibody preparations .

How can I optimize immunostaining protocols for PRMT10 detection in subcellular localization studies?

Optimizing immunostaining protocols for PRMT10 subcellular localization requires careful consideration of fixation, permeabilization, and antibody incubation conditions. Based on published research, PRMT10 is expressed in the nucleus of both epithelial and stromal cells in prostate tissue . For effective nuclear staining, begin with paraformaldehyde fixation (4%) for 15-20 minutes at room temperature, followed by permeabilization with 0.1-0.5% Triton X-100 for 10 minutes. Blocking should be performed with 5% normal serum (matching the species of the secondary antibody) with 1% BSA in PBS for 1 hour at room temperature.

For PRMT10 antibody incubation, use concentrations of 1-4 μg/mL in blocking buffer overnight at 4°C . When performing co-localization studies with androgen receptor, which PRMT10 is known to interact with, ensure sequential staining with appropriate controls to avoid cross-reactivity. To enhance signal-to-noise ratio, increase washing steps (at least 3 × 10 minutes with 0.1% Tween-20 in PBS) and optimize secondary antibody concentration. For nuclear PRMT10 detection, counterstaining with DAPI helps confirm nuclear localization, but use lower concentrations to avoid overwhelming the PRMT10 signal. Implementation of Z-stack imaging with confocal microscopy is particularly valuable for determining the precise subcellular localization of PRMT10 within nuclear subcompartments.

What are the best experimental approaches to study PRMT10-androgen receptor interactions?

To effectively study PRMT10-androgen receptor (AR) interactions, a multi-method approach yields the most comprehensive and reliable results. Co-immunoprecipitation (Co-IP) has been successfully used to demonstrate that PRMT10 co-immunoprecipitates with AR in prostate cancer LNCaP cells both in the presence and absence of dihydrotestosterone (DHT) . For Co-IP, use lysis buffers containing 150 mM NaCl, 50 mM Tris-HCl (pH 7.4), 1% NP-40, and protease inhibitors, with antibody coupling to protein A/G magnetic beads for efficient precipitation.

For confirming physiological relevance, proximity ligation assays (PLA) offer high sensitivity for detecting protein-protein interactions in situ. Additionally, chromatin immunoprecipitation (ChIP) followed by sequencing (ChIP-seq) can determine if PRMT10 co-localizes with AR at androgen response elements on DNA. For functional validation, siRNA knockdown of PRMT10 has demonstrated decreased DHT-dependent growth of LNCaP cells and reduced expression of AR-target genes like prostate-specific antigen . In designing knockdown experiments, at least two non-overlapping siRNAs should be used to control for off-target effects, and rescue experiments with siRNA-resistant PRMT10 constructs provide strong validation.

FRET (Fluorescence Resonance Energy Transfer) or BiFC (Bimolecular Fluorescence Complementation) assays can also visualize direct interactions in living cells. When performing these interaction studies, it's crucial to include both ligand-treated (DHT) and untreated conditions, as research has shown PRMT10-AR interactions occur in both contexts but may serve different regulatory functions .

How can PRMT10 enzymatic activity be accurately measured in vitro and in cellular contexts?

Measuring PRMT10 enzymatic activity requires specialized assays that detect arginine methylation of target substrates. For in vitro methyltransferase assays, purified recombinant PRMT10 (which can be produced as a GST-fusion protein) is incubated with potential substrates such as histone H4 or synthetic peptides containing target arginine residues in the presence of S-adenosyl-L-methionine (SAM) as the methyl donor . Radioactive assays using [³H]SAM allow direct visualization of methylated products through thin-layer chromatography (TLC) or autoradiography after SDS-PAGE separation .

For non-radioactive alternatives, antibodies specific to asymmetrically dimethylated histone H4 at arginine 3 (H4R3me2a) can be used in western blot analysis to detect PRMT10 activity, as evidence suggests PRMT10 functions as a type I PRMT that catalyzes asymmetric dimethylation . Mass spectrometry approaches provide the most comprehensive analysis of methylation sites and can distinguish between monomethylation and symmetric or asymmetric dimethylation.

In cellular contexts, methyltransferase activity can be measured by transfecting cells with PRMT10 expression constructs or siRNA for knockdown studies, followed by immunoblotting with methylation-specific antibodies. When designing these experiments, include appropriate controls such as catalytically inactive PRMT10 mutants (mutations in the SAM-binding domain) and type-specific PRMT inhibitors. The automethylation activity of PRMT10 observed when no substrate is present can serve as an internal control for enzyme activity . For quantitative measurements, enzyme kinetics should be determined using varying concentrations of both substrate and SAM to establish Km and Vmax values.

What are the technical considerations for validating PRMT10 antibody specificity in experimental systems?

Validating PRMT10 antibody specificity is crucial for obtaining reliable experimental results. A multi-tiered validation approach should include several complementary techniques. First, western blot analysis should demonstrate a single band (or closely migrating bands) at the expected molecular weight of approximately 42-43 kDa . Knockdown validation through siRNA or CRISPR methods provides strong evidence of specificity - the antibody signal should decrease proportionally to the reduction in PRMT10 protein levels .

For immunohistochemistry and immunocytochemistry applications, comparison of staining patterns with multiple antibodies recognizing different epitopes of PRMT10 helps confirm specificity. Pre-absorption tests, where the antibody is pre-incubated with the immunizing peptide, should abolish or significantly reduce the signal if the antibody is specific .

When working with novel tissue types or species, researchers should include positive controls (tissues known to express high levels of PRMT10, such as prostate) and negative controls (tissues with minimal PRMT10 expression or PRMT10-knockout samples) . Cross-reactivity with other PRMT family members, particularly the closely related PRMT9, should be evaluated through overexpression systems or with recombinant protein panels .

It's important to note that the reactivity profile varies between antibodies - some PRMT10 antibodies have been tested for cross-reactivity with unrelated antigens in ELISA assays and showed no cross-reactivity, while others may have different specificity profiles . For experiments requiring absolute specificity, validation using immune-mass spectrometry to identify the exact proteins immunoprecipitated by the antibody provides the highest level of confidence.

How does PRMT10 expression and function differ between normal and cancer tissues?

PRMT10 expression and function exhibit significant differences between normal and cancer tissues, particularly in the context of prostate biology. In normal tissues, PRMT10 is highly expressed in reproductive tissues, including the prostate, where immunostaining has revealed expression in the nucleus of both epithelial and stromal cells . This nuclear localization aligns with its function as a methyltransferase that may regulate gene expression.

In prostate cancer, PRMT10 appears to play a crucial role in androgen-dependent cell proliferation. Studies in LNCaP human prostate cancer cells have demonstrated that PRMT10 co-immunoprecipitates with androgen receptor (AR) in both the presence and absence of dihydrotestosterone (DHT) . This interaction suggests that PRMT10 may function as a co-regulator of AR-mediated transcription, potentially contributing to prostate cancer progression.

Functionally, knockdown of PRMT10 by siRNA in LNCaP cells resulted in decreased DHT-dependent cell growth and reduced induction of prostate-specific antigen (PSA), an AR-target gene . This suggests that PRMT10 is necessary for optimal androgen signaling in prostate cancer cells. Interestingly, DHT treatment was found to decrease PRMT10 at both mRNA and protein levels, and this decrease was prevented by AR knockdown or treatment with an AR antagonist . This indicates a potential negative feedback mechanism where AR signaling downregulates PRMT10 expression, possibly as a regulatory circuit to modulate androgen response.

When designing studies to compare PRMT10 in normal versus cancer tissues, researchers should consider tissue microarrays with matched normal and tumor samples from the same patients, quantitative assessment of both expression levels and subcellular localization, and correlation with clinical parameters such as Gleason score, tumor stage, and patient outcomes in prostate cancer studies.

What are the recommended dilutions and incubation conditions for PRMT10 antibodies in different applications?

Optimal working conditions for PRMT10 antibodies vary by application and specific antibody product. For immunocytochemistry and immunofluorescence applications, PRMT10 antibodies are typically used at concentrations of 1-4 μg/mL . This concentration range has been validated to provide specific staining with minimal background. For these applications, overnight incubation at 4°C in appropriate blocking buffer (typically PBS with 1-5% BSA or normal serum) yields optimal results.

For Western blot applications, dilutions typically range from 1:500 to 1:2000 depending on the specific antibody concentration and sensitivity requirements . Incubation is generally performed overnight at 4°C in 5% non-fat dry milk or BSA in TBST buffer. For immunoprecipitation, approximately 2-5 μg of antibody per 500 μg of total protein lysate is recommended, with incubation performed overnight at 4°C with gentle rotation .

ELISA applications typically employ antibody dilutions ranging from 1:1000 to 1:5000, with specific optimization required for each assay format . For all applications, it's advisable to perform a dilution series during initial optimization to determine the optimal antibody concentration that provides maximum specific signal with minimal background. Temperature and duration of incubation should also be optimized, with longer incubations at 4°C generally providing better signal-to-noise ratios compared to shorter incubations at room temperature.

What controls should be included when using PRMT10 antibodies in research applications?

A comprehensive control strategy is essential when using PRMT10 antibodies to ensure experimental validity and interpretability. Positive controls should include samples known to express PRMT10, such as reproductive tissues (particularly prostate tissue) or cell lines with confirmed PRMT10 expression like LNCaP prostate cancer cells . Negative controls should include samples with PRMT10 knockdown or knockout, achieved through siRNA, shRNA, or CRISPR-Cas9 technology .

For immunostaining applications, isotype controls using non-specific antibodies of the same isotype (IgG) and concentration as the PRMT10 antibody help distinguish specific from non-specific binding . Secondary antibody-only controls (omitting primary antibody) are essential to assess potential background from the detection system.

When performing functional studies, include both wild-type and catalytically inactive PRMT10 mutants to distinguish between enzymatic and non-enzymatic roles. For interaction studies, such as co-immunoprecipitation with androgen receptor, include both ligand-treated (DHT) and untreated conditions, as research shows PRMT10-AR interactions occur in both contexts .

Technical controls should address potential cross-reactivity with other PRMT family members, particularly PRMT9 which may share structural similarities . For quantitative applications, standard curves using recombinant PRMT10 protein at known concentrations provide critical calibration references. When interpreting results, be aware that DHT treatment has been shown to decrease PRMT10 at both mRNA and protein levels, which can serve as an internal control for hormone-responsive systems .

How can PRMT10 antibodies be effectively used in multiplex immunofluorescence with other prostate cancer markers?

Multiplex immunofluorescence combining PRMT10 detection with other prostate cancer markers requires careful optimization of antibody combinations, fluorophore selection, and staining protocols. When designing a multiplex panel, pair PRMT10 antibodies with markers that provide complementary biological information, such as androgen receptor (AR), prostate-specific antigen (PSA), and other PRMT family members or epigenetic regulators .

For successful multiplexing, select primary antibodies raised in different host species to avoid cross-reactivity; for example, pair rabbit polyclonal PRMT10 antibodies with mouse monoclonal AR antibodies . When antibodies from the same species must be used, sequential staining with complete blocking steps between antibodies can minimize cross-reactivity.

Fluorophore selection should account for spectral overlap and tissue autofluorescence. Tyramide signal amplification (TSA) can enhance detection sensitivity for low-abundance proteins while allowing antibody stripping and re-probing of the same tissue section. To minimize bleed-through, select fluorophores with well-separated excitation and emission spectra, and include single-stain controls for spectral unmixing during analysis.

For co-localization studies of PRMT10 with AR, which are known to interact in prostate cancer cells , high-resolution confocal microscopy with Z-stack acquisition is recommended. Quantitative analysis should employ automated image analysis algorithms that can distinguish nuclear from cytoplasmic staining, measure co-localization coefficients, and correlate expression patterns with clinical parameters in tissue microarrays.

When interpreting results, consider that PRMT10 and AR have a complex regulatory relationship, with DHT treatment decreasing PRMT10 levels while PRMT10 is required for optimal AR signaling . This dynamic relationship may manifest as varying degrees of co-localization dependent on hormonal status and disease progression.

What are the challenges in quantifying PRMT10 protein levels in clinical samples?

Quantifying PRMT10 protein levels in clinical samples presents several methodological challenges that researchers must address to obtain reliable and reproducible results. Pre-analytical variables significantly impact PRMT10 detection, including tissue fixation time, fixative type, and storage conditions. Standard formalin-fixed paraffin-embedded (FFPE) processing can mask PRMT10 epitopes, necessitating optimized antigen retrieval methods (typically heat-induced epitope retrieval in citrate buffer pH 6.0 or EDTA buffer pH 9.0) .

Normalization strategies are critical for meaningful quantification. For immunohistochemistry, digital pathology approaches using whole slide imaging and automated scoring algorithms help standardize interpretation. When quantifying by Western blot, selecting appropriate loading controls is essential; traditional housekeeping proteins may vary across different prostate cancer stages, making total protein staining methods such as Ponceau S or Stain-Free technology preferable .

Technical limitations include antibody lot-to-lot variability, which can be addressed by maintaining reference standards across experiments. The dynamic range of detection methods may also be limiting, particularly for samples with extreme expression levels. Mass spectrometry-based approaches offer an antibody-independent alternative for absolute quantification but require specialized equipment and expertise.

Biological variables further complicate quantification. PRMT10 levels are regulated by androgens, with DHT decreasing PRMT10 at both mRNA and protein levels . This hormonal dependency means patient treatment history (particularly androgen deprivation therapy) must be considered when interpreting results. Additionally, PRMT10's subcellular localization (nuclear vs. cytoplasmic) may have functional significance, requiring separate quantification of these compartments rather than total protein levels .

For longitudinal studies, researchers should establish standardized protocols, use automated staining platforms, and implement digital pathology with artificial intelligence-assisted quantification to minimize variability and enhance reproducibility across different clinical sample cohorts.

How might PRMT10 function as a potential therapeutic target in prostate cancer?

PRMT10's role as a potential therapeutic target in prostate cancer is supported by several lines of evidence from molecular studies. PRMT10 has been identified as an androgen receptor (AR) interacting protein that plays a critical role in prostate cancer cell proliferation . Knockdown studies have demonstrated that reducing PRMT10 expression decreases dihydrotestosterone (DHT)-dependent growth of LNCaP prostate cancer cells and reduces the expression of prostate-specific antigen (PSA), an important AR-target gene . These findings suggest that inhibiting PRMT10 could potentially suppress AR signaling and prostate cancer growth.

Several potential therapeutic strategies could be developed targeting PRMT10. Small molecule inhibitors targeting the methyltransferase activity of PRMT10 could disrupt its enzymatic function. Since PRMT10 appears to be a type I PRMT that catalyzes asymmetric dimethylation of arginine residues , designing selective inhibitors that distinguish between type I and type II PRMTs would be crucial for specificity. Structure-based drug design approaches could exploit the unique features of PRMT10's catalytic domain to develop selective inhibitors.

Alternatively, disrupting the protein-protein interaction between PRMT10 and AR could be an effective strategy. This would require detailed mapping of the interaction interface between these proteins, followed by development of peptide mimetics or small molecules that competitively inhibit this interaction. Another approach could involve targeted protein degradation strategies such as PROTACs (Proteolysis Targeting Chimeras) directed at PRMT10.

The complex regulatory relationship between PRMT10 and AR signaling must be considered when developing therapeutic strategies. DHT treatment decreases PRMT10 at both mRNA and protein levels, suggesting a negative feedback loop in AR signaling . Therefore, combination therapies targeting both AR and PRMT10 might be more effective than single-agent approaches, particularly in castration-resistant prostate cancer where AR signaling remains active despite androgen deprivation therapy.

What is the relationship between PRMT10 expression and clinical outcomes in prostate cancer patients?

The relationship between PRMT10 expression and clinical outcomes in prostate cancer patients represents an emerging area of research with significant translational potential. While comprehensive clinical studies correlating PRMT10 expression with patient outcomes are still developing, molecular evidence suggests important prognostic implications. PRMT10 plays a crucial role in androgen receptor (AR) signaling, which is the primary driver of prostate cancer progression . The interaction between PRMT10 and AR, coupled with PRMT10's requirement for optimal androgen-dependent cell growth, suggests that its expression levels may correlate with disease aggressiveness and treatment response.

In designing clinical correlation studies, researchers should consider several methodological approaches. Immunohistochemical analysis of PRMT10 in prostate cancer tissue microarrays, with quantification of both expression intensity and subcellular localization, provides spatial context that may reveal heterogeneity within tumors. Correlation analyses should examine relationships between PRMT10 expression and established prognostic factors including Gleason score, tumor stage, PSA levels, and proliferation markers like Ki-67.

The complex regulatory relationship between PRMT10 and androgen signaling must be considered in these analyses. Since DHT treatment decreases PRMT10 levels , PRMT10 expression patterns may differ between hormone-naïve and castration-resistant prostate cancers. Therefore, subgroup analyses based on prior hormonal therapies are essential. Additionally, evaluating the ratio of PRMT10 to AR expression, rather than absolute PRMT10 levels alone, may provide better prognostic information given their functional interaction.

Longitudinal studies examining changes in PRMT10 expression during disease progression, particularly during the transition to castration resistance, could reveal its role in treatment resistance mechanisms and inform therapeutic strategies targeting this protein.

How does PRMT10 interact with other epigenetic regulators in controlling gene expression?

PRMT10's role within the broader epigenetic regulatory network involves complex interactions with various chromatin modifiers and transcription factors. As a protein arginine methyltransferase, PRMT10 catalyzes the methylation of arginine residues on target proteins, including histones. Evidence suggests that PRMT10 functions as a type I PRMT, catalyzing asymmetric dimethylation of substrates, which has been confirmed using antibodies against asymmetrically dimethylated histone H4 at arginine 3 (H4R3me2a) . This specific histone modification is generally associated with transcriptional activation.

The interaction between PRMT10 and androgen receptor (AR) in prostate cells suggests that PRMT10 functions within transcriptional regulatory complexes . AR is known to recruit various coregulators, including histone-modifying enzymes, to regulate gene expression. PRMT10 likely cooperates with these other epigenetic regulators to fine-tune the expression of AR target genes. The decrease in PRMT10 levels following DHT treatment, coupled with reduced PSA expression (an AR target gene) after PRMT10 knockdown, indicates a complex regulatory circuit involving feedback mechanisms .

To study these interactions comprehensively, ChIP-seq (Chromatin Immunoprecipitation followed by sequencing) experiments using antibodies against PRMT10, AR, and various histone modifications (H4R3me2a, H3K4me3, H3K27ac) would reveal genome-wide co-occupancy patterns. Sequential ChIP (re-ChIP) could confirm co-occupancy of PRMT10 with other epigenetic regulators at specific genomic loci.

Protein interaction studies using techniques such as BioID or proximity labeling coupled with mass spectrometry would identify PRMT10's protein interaction network within chromatin regulatory complexes. RNA-seq following PRMT10 knockdown or overexpression, compared with manipulation of other epigenetic regulators, could reveal cooperative or antagonistic relationships in gene expression control.

The integration of these multi-omics approaches would provide a comprehensive understanding of how PRMT10 functions within the broader epigenetic landscape to control gene expression programs relevant to cellular proliferation and differentiation in both normal and cancer contexts.

What are common technical issues when using PRMT10 antibodies and how can they be resolved?

Researchers frequently encounter technical challenges when working with PRMT10 antibodies across various applications. Understanding these issues and implementing appropriate solutions ensures more reliable and reproducible results. One common problem is high background or non-specific staining in immunohistochemistry and immunofluorescence applications. This can be addressed by optimizing blocking conditions (using 5% normal serum from the same species as the secondary antibody, combined with 1% BSA), increasing washing steps (at least 3-5 washes of 5-10 minutes each), and titrating antibody concentration to find the optimal 1-4 μg/mL range that balances specific signal with minimal background .

For Western blot applications, multiple bands or unexpected molecular weight bands may appear. This could indicate protein degradation, which can be minimized by using fresh samples and complete protease inhibitor cocktails during extraction. Additionally, post-translational modifications of PRMT10 or detection of splice variants may result in bands at unexpected sizes. To resolve this, sample denaturing conditions can be optimized, and comparison with recombinant PRMT10 protein as a positive control helps identify the specific band .

Weak or absent signals despite expected PRMT10 expression may result from insufficient antigen retrieval in fixed tissues. For FFPE samples, optimizing antigen retrieval methods is crucial - try both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) with different heating times to determine optimal conditions. Additionally, some epitopes may be masked by protein interactions or conformational issues; using denaturing conditions or multiple antibodies targeting different PRMT10 epitopes can help overcome this limitation .

Inconsistent results between experiments often stem from antibody lot-to-lot variability. This can be mitigated by purchasing larger lots for long-term studies, including consistent positive controls across experiments, and validating each new antibody lot against previous lots. If inconsistencies persist, consider using alternative detection methods like RT-qPCR to verify PRMT10 expression patterns at the mRNA level as a complementary approach.

How can researchers distinguish between PRMT10 and other PRMT family members in their experiments?

Distinguishing PRMT10 from other PRMT family members requires careful experimental design and validation strategies. Sequence analysis reveals that PRMT9 shares the highest similarity with PRMT10 among the PRMT family , making specificity particularly important. When selecting antibodies, researchers should prioritize those that have been explicitly tested for cross-reactivity with other PRMT family members, especially PRMT9. Antibodies targeting unique regions of PRMT10, particularly those directed against the N-terminal domain (amino acids 1-50 or 39-68) rather than the more conserved methyltransferase domain, generally offer better specificity .

For definitive validation of antibody specificity, expression systems provide powerful tools. Overexpression of PRMT10 in cell lines with low endogenous expression, alongside overexpression of other PRMT family members in separate samples, allows direct comparison of antibody reactivity. Similarly, knockdown or knockout approaches using siRNA, shRNA, or CRISPR-Cas9 targeting PRMT10 specifically should reduce or eliminate the signal if the antibody is truly specific .

At the functional level, substrate specificity can help distinguish PRMT10 activity. Evidence suggests that PRMT10 functions as a type I PRMT with high specificity for histone H4 at arginine 3 (H4R3), catalyzing asymmetric dimethylation (H4R3me2a) . In methyltransferase activity assays, comparing the substrate preferences and product specificity (asymmetric versus symmetric dimethylation) between PRMT10 and other family members can provide functional discrimination.

For mRNA-level studies, qRT-PCR using primers designed to unique regions of PRMT10 transcript offers an antibody-independent method for specific detection. When designing primers, target regions with minimal sequence homology to other PRMT family members, particularly PRMT9, and validate specificity using positive and negative control samples.

Mass spectrometry-based approaches provide the highest specificity for protein identification. Immunoprecipitation followed by mass spectrometry can definitively identify PRMT10 and distinguish it from other PRMT family members based on unique peptide sequences, regardless of structural similarities.

What are the best approaches for developing and validating novel PRMT10 antibodies for research use?

Developing and validating novel PRMT10 antibodies requires a systematic approach to ensure specificity, sensitivity, and reliability across different applications. The initial step involves careful antigen design, selecting unique epitopes that distinguish PRMT10 from other PRMT family members, particularly PRMT9 . Bioinformatic analysis should identify regions with minimal sequence homology to other proteins, with the N-terminal domain (amino acids 1-50 or 39-68) being particularly useful for generating specific antibodies . Both synthetic peptides and recombinant protein fragments can serve as immunogens, with the latter potentially preserving some structural epitopes.

For antibody production, both polyclonal and monoclonal approaches have merits. Polyclonal antibodies recognize multiple epitopes, potentially increasing sensitivity, while monoclonal antibodies offer consistency and specificity for a single epitope. Rabbit host animals are commonly used for PRMT10 antibody production and have demonstrated good results in available commercial antibodies .

Comprehensive validation should include multiple complementary techniques. Western blot analysis should demonstrate a single band (or closely migrating bands) at the expected molecular weight of approximately 42-43 kDa . Testing against recombinant PRMT10 protein provides a positive control, while testing against other recombinant PRMT family members confirms specificity. Immunoprecipitation followed by mass spectrometry offers the highest standard for specificity validation, definitively identifying immunoprecipitated proteins.

Cell-based validation using overexpression and knockdown/knockout systems is essential. Transfection of expression constructs for PRMT10 should increase antibody signal, while siRNA, shRNA, or CRISPR-Cas9 targeting PRMT10 should reduce or eliminate the signal . Testing across multiple cell types with varying endogenous PRMT10 expression levels evaluates sensitivity and dynamic range.

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.