PRMT5 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Product dispatch occurs within 1-3 business days of order receipt. Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
72 kDa ICln binding protein antibody; 72 kDa ICln-binding protein antibody; ANM5_HUMAN antibody; Histone synthetic lethal 7; S. cerevisiae; homolog of antibody; Histone-arginine N-methyltransferase PRMT5 antibody; HMT1 hnRNP methyltransferase like 5 antibody; HOMOLOG OF; SKB1 antibody; HRMT1L5 antibody; IBP72 antibody; Jak-binding protein 1 antibody; JBP 1 antibody; JBP1 antibody; PRMT 5 antibody; PRMT5 antibody; Protein arginine methyltransferase 5 antibody; Protein arginine N methyltransferase 5 antibody; Protein arginine N methyltransferase 5 N terminally processed antibody; Protein arginine N-methyltransferase 5 antibody; S. POMBE antibody; S. POMBE HOMOLOG OF; SKB1 antibody; SHK1 KINASE BINDING PROTEIN 1 antibody; Shk1 kinase binding protein 1 homolog antibody; Shk1 kinase-binding protein 1 homolog antibody; Shk1 kinase/binding protein 1; S. pombe; homolog of antibody; SKB 1 antibody; SKB1 antibody; SKB1 homolog antibody; SKB1: SKB1 homolog (S. pombe) antibody; SKB1Hs antibody
Target Names
Uniprot No.

Target Background

Function

PRMT5 is an arginine methyltransferase catalyzing the formation of ω-N-monomethylarginine (MMA) and symmetric dimethylarginine (sDMA), exhibiting a preference for MMA. It specifically mediates the symmetrical dimethylation of arginine residues in the small nuclear ribonucleoproteins Sm D1 (SNRPD1) and Sm D3 (SNRPD3), a process crucial for snRNP core particle assembly and biogenesis. Additional functions include methylation of SUPT5H (potentially regulating transcriptional elongation), myelin basic protein (MBP) in vitro, and involvement in cytokine-activated transduction pathways. PRMT5 negatively regulates cyclin E1 promoter activity and cellular proliferation. It methylates histone H2A and H4 'Arg-3' during germ cell development and histone H3 'Arg-8', potentially repressing transcription. Further, it methylates Piwi proteins (PIWIL1, PIWIL2, and PIWIL4), a methylation required for their interaction with Tudor domain-containing proteins and subsequent meiotic nuage localization. Other substrates include RPS10, EGFR (enhancing its phosphorylation and impacting downstream signaling), RAF1 and BRAF (destabilizing these proteins and reducing their activity), HOXA9, and SRGAP2 (involved in cell migration and differentiation). PRMT5 acts as a transcriptional corepressor in CRY1-mediated repression of PER1 by regulating H4R3 dimethylation at the PER1 promoter and also methylates GM130/GOLGA2, regulating Golgi ribbon formation. It methylates H4R3 in genes involved in glioblastomagenesis in a CHTOP- and/or TET1-dependent manner. Symmetrical methylation of POLR2A facilitates recruitment of proteins like SMN1/SMN2 and SETX, essential for resolving RNA-DNA hybrids (R-loops) during transcription termination. In collaboration with LYAR, it binds the HBG1/HBG2 promoter, repressing gamma-globin expression. Finally, PRMT5 symmetrically methylates NCL and TP53 (potentially influencing TP53 target gene specificity), and plays a role in spliceosome maturation and mRNA splicing in prophase I spermatocytes.

Gene References Into Functions

Numerous studies highlight PRMT5's significant role in various cancers. Its overexpression has been linked to aggressive characteristics in several cancers, including:

  • Gastric cancer: PRMT5 overexpression represses the tumor suppressor IRX1. (PMID: 29802960)
  • Oral squamous cell carcinoma: PRMT5 is implicated in oncogenesis and stromal invasion. (PMID: 29603824)
  • Hepatocellular carcinoma and other cancers: Elevated PRMT5 phosphorylation and H2A/4 dimethylation are observed. (PMID: 28074910)
  • Acute myeloid leukemia: PRMT5 is crucial for leukemic progression. (PMID: 28945229)
  • Hepatocellular carcinoma and colon cancer: PRMT5 contributes to invasiveness. (PMID: 29749478)
  • Breast cancer: PRMT5 regulates breast cancer stem cell proliferation and epigenetically regulates FOXP1. (PMID: 29262329)
  • Various cancers: PRMT5 influences Eno-1 cell-surface localization. (PMID: 29501774)
  • Structural insights: Cryo-EM structure of PRMT5 in complex with MEP50. (PMID: 29518110)
  • Kinetic studies: Understanding the interplay between PRMT5 and PRMT7. (PMID: 28874563)
  • Pancreatic ductal adenocarcinoma and colorectal cancer: PRMT5 is a potential therapeutic target. (PMID: 28591716)
  • Hepatocellular carcinoma: PRMT5 activates the CDK4-RB-E2F pathway. (PMID: 27708221)
  • Various cancers: The PRMT5-E2F1 pathway is a potential target for cancer gene therapy. (PMID: 26990556)
  • Inhibitor development: Studies on PRMT5 inhibitors and mutant residue assays. (PMID: 29154828)
  • Gastric cancer: PRMT5 inhibition downregulates eIF4E. (PMID: 28987382)
  • ZNF326 is an interaction partner and substrate of the PRMT5/WDR77 complex. (PMID: 28977470)
  • Prostate cancer: ERG signaling is driven through PRMT5-dependent methylation of the androgen receptor. (PMID: 27183006)
  • Tumor growth: PRMT5/MEP50 complex is required for tumor growth. (PMID: 28854561)
  • Oropharyngeal squamous cell carcinoma: High nuclear PRMT5 expression is associated with this cancer. (PMID: 28107179)
  • Prostate cancer: High PRMT5 expression is associated with this cancer. (PMID: 27546619)
  • Lung tumorigenesis: PRMT5 and p44 regulate gene expression of growth and anti-growth factors. (PMID: 27480244)
  • Translation: PRMT5 regulates internal ribosome entry site-dependent translation via methylation of hnRNP A1. (PMID: 28115626)
  • Chronic myelogenous leukemia: PRMT5 knockdown reduces growth and survival of CD34+ cells. (PMID: 27643437)
  • DNA repair: Methylation of RUVBL1 by PRMT5 is required for homologous recombination-mediated double-strand break repair. (PMID: 28238654)
  • T cell responses: PRMT5 is involved in the regulation of adaptive memory Th cell responses. (PMID: 28087667)
  • Glioblastoma: PRMT5 differentially regulates self-renewal, survival, and proliferation of glioblastoma neurosphere cells. (PMID: 27292259)
  • Rheumatoid arthritis: PRMT5 regulates inflammatory factors, cell proliferation, migration, and invasion of fibroblast-like synoviocytes. (PMID: 27860244)
  • Plasma membrane association: The C-terminal motif of PRMT5 is required for plasma membrane association. (PMID: 28031468)
  • Hepatocellular carcinoma: PRMT5 may act as a putative oncogene. (PMID: 28101581)
  • Breast cancer: High PRMT5 expression is associated with cell proliferation, cell cycle progression, and apoptosis. (PMID: 28381188)
  • Gastric cancer: PRMT5 enhances the malignant phenotype of cancer cell lines. (PMID: 27315569)
  • ASK1: PRMT5 is an ASK1-binding protein and mediates its arginine methylation. (PMID: 26912789)
  • MTAP deletion: MAT2A and PRMT5 are vulnerable enzymes in cells with MTAP deletion. (PMID: 27068473)
  • Nasopharyngeal carcinoma: PRMT5 promotes radioresistance via targeting FGFR3. (PMID: 26708443)
  • c-Myc: Omomyc co-localizes with c-Myc, PRMT5, and H4R3me2s-enriched chromatin domains. (PMID: 26563484)
  • HTLV-1: PRMT5 expression is upregulated during HTLV-1-mediated T-cell transformation. (PMID: 26729154)
  • Acute myeloid leukemia: Increased PRMT5 activity enhances growth. (PMID: 26536822)
  • Dopamine receptor: Arginine residues within the D2 dopamine receptor are methylated by PRMT5. (PMID: 26554819)
  • Colorectal carcinoma: High nuclear PRMT5 expression is a potential marker for submucosal invasion. (PMID: 26248553)
  • Colorectal cancer: PRMT5 epigenetically regulates arginine methylation of oncogenes like eIF4E and FGFR3. (PMID: 26078354)
  • SREBP1a: PRMT5-induced methylation prevents phosphorylation of SREBP1a. (PMID: 26759235)
  • Hepatocellular carcinoma: PRMT5 regulates growth via beta-catenin downregulation. (PMID: 26541651)
  • Keratinocytes: PRMT5 silences gene expression by histone arginine methylation. (PMID: 26763441)
  • RASSF1A: PRMT5 co-localizes with RASSF1A on microtubules. (PMID: 26482848)
  • Endometrial carcinogenesis: UHRF1 forms a complex with PRMT5. (PMID: 26597461)
  • Multiple cancer lineages: PRMT5 is a potential vulnerability across multiple cancer lineages. (PMID: 26912360)
  • 5-methylthioadenosine phosphorylase-deleted cancers: Hypomorphic PRMT5 state is sensitized toward further PRMT5 inhibition. (PMID: 26912361)
  • Huntington's disease: PRMT5-mediated methylation is attenuated by mutant huntingtin. (PMID: 25927346)
  • Lymphomagenesis: PRMT5 is required for lymphomagenesis. (PMID: 25582697)
  • Novel PRMT5 splice variant: The PRMT5S splice variant contributes to the diverse effects of PRMT5. (PMID: 26151339)
  • MYCN regulation: PRMT5 is implicated in MYCN post-translational regulation. (PMID: 25475372)
Database Links

HGNC: 10894

OMIM: 604045

KEGG: hsa:10419

STRING: 9606.ENSP00000319169

UniGene: Hs.367854

Protein Families
Class I-like SAM-binding methyltransferase superfamily, Protein arginine N-methyltransferase family
Subcellular Location
Cytoplasm. Nucleus. Chromosome. Golgi apparatus.
Tissue Specificity
Ubiquitous.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : WB

Sample type: Human HT-29 cell

Sample dilution: 1:1000

Review: Oxicam drugs at 200 µM concentration downregulated PRMTs.

Q&A

What is PRMT5 and what are its primary functions in cellular biology?

PRMT5 (protein arginine methyltransferase 5) is a key enzyme responsible for symmetric dimethylation of arginine residues on target proteins. It functions as a type II protein arginine methyltransferase with a calculated molecular weight of approximately 73 kDa, though it typically appears around 68 kDa in experimental applications such as Western blotting . PRMT5 plays critical roles in various cellular processes including transcriptional regulation, signal transduction, RNA processing, and DNA damage response. The enzyme contributes to epigenetic regulation through histone modification, particularly H4R3 and H3R8 symmetric dimethylation, which typically results in transcriptional repression. Its expression pattern varies across tissue types, with notable presence in fibroblasts and endothelial cells in dermal tissues . Recent research has uncovered its potential involvement in autoimmune conditions, particularly systemic sclerosis, where PRMT5 may contribute to inflammatory and fibrotic pathways .

How do researchers detect and quantify PRMT5 in experimental settings?

Detection and quantification of PRMT5 in experimental settings typically employ antibody-based methodologies. Western blotting remains the gold standard for protein-level detection, with recommended antibody dilutions ranging from 1:500 to 1:2000 depending on sample characteristics and experimental conditions . Immunoprecipitation followed by mass spectrometry (IP-MS) represents an advanced approach for quantitative proteomics analysis of PRMT5, particularly when studying complex interactions . Enzyme-linked immunosorbent assay (ELISA) provides a sensitive method for quantifying PRMT5 in serum samples, with careful attention to serial dilutions necessary to determine optimal conditions and calculate accurate values of area under the curve (AUC) . Protein microarray technology offers another validation method, especially valuable when confirming findings across independent patient cohorts. For tissue-level detection, immunohistochemistry and immunofluorescence techniques can visualize PRMT5 expression patterns, revealing cell-type specific localization, such as its pronounced presence in fibroblasts and moderate expression in endothelial cells .

What are the critical parameters for selecting an appropriate anti-PRMT5 antibody for specific research applications?

When selecting an anti-PRMT5 antibody, researchers should evaluate several critical parameters to ensure optimal performance in their specific applications. First, consider the antibody's validated reactivity with the species under investigation—commercial options often demonstrate cross-reactivity with human, mouse, and rat samples . The antibody class (monoclonal versus polyclonal) significantly impacts specificity and application versatility; polyclonal antibodies offer broader epitope recognition but potentially increased background, while monoclonals provide high specificity for defined epitopes. Review validated applications thoroughly—some antibodies perform reliably in Western blot and ELISA but may not be optimized for immunohistochemistry or other techniques . The immunogen used for antibody generation provides insight into potential cross-reactivity; for example, antibody 27109-1-AP was generated against a PRMT5 fusion protein (Ag25896) . For quantitative applications, select antibodies with demonstrated linearity across relevant concentration ranges. Finally, consider the detection system compatibility (direct versus indirect) and whether conjugated antibodies might streamline your experimental workflow. Preliminary validation experiments comparing multiple antibodies are recommended for novel research applications.

What optimization strategies improve PRMT5 detection sensitivity in Western blotting applications?

Optimizing PRMT5 detection in Western blotting requires systematic refinement of several key parameters. First, sample preparation is critical—PRMT5's observed molecular weight of 68 kDa necessitates complete protein denaturation and adequate gel resolution in the 60-80 kDa range . Begin with a titration of antibody concentrations, testing the recommended range of 1:500-1:2000 to identify optimal signal-to-noise ratios for your specific cell lines or tissues . Blocking conditions significantly impact background—test both BSA and non-fat dry milk blockers at concentrations between 3-5% to minimize non-specific binding. For challenging samples with low PRMT5 expression, implement signal amplification through enhanced chemiluminescence (ECL) substrates with varied sensitivity levels. The exposure time should be carefully optimized—multiple exposures ranging from 10 seconds to 5 minutes capture the dynamic range of expression across samples. When analyzing post-translational modifications of PRMT5, phosphatase and protease inhibitor cocktails must be incorporated during lysate preparation. For clinical samples, standardize protein loading using multiple housekeeping controls (β-actin, GAPDH) to account for variable expression across patient tissues. Finally, quantitative analysis using densitometry should include appropriate normalization and statistical validation to ensure reproducibility across experimental replicates.

How can researchers establish specificity controls when using anti-PRMT5 antibodies in immunoprecipitation experiments?

Establishing rigorous specificity controls in anti-PRMT5 immunoprecipitation (IP) experiments is essential for result validation. The primary negative control should include an isotype-matched non-specific antibody (typically rabbit IgG for polyclonal antibodies like 27109-1-AP) , processed identically to the anti-PRMT5 IP samples. Pre-adsorption controls, where the antibody is pre-incubated with excess recombinant PRMT5 protein before immunoprecipitation, help verify binding specificity. For advanced validation, researchers should perform parallel IPs using two different anti-PRMT5 antibodies targeting distinct epitopes, which should yield consistent interactome profiles. When conducting MS-based analysis of immunoprecipitated complexes, implement stringent bioinformatic filtering to distinguish true interactors from common contaminants, as demonstrated in the systemic sclerosis studies that identified specific PRMT5-associated proteins . Genetic validation through PRMT5 knockdown or knockout samples provides definitive specificity confirmation—the target band or interacting partners should be significantly diminished or absent. For clinical samples, include healthy control sera alongside patient samples when conducting serum IP-MS studies, as this approach successfully identified anti-PRMT5 antibodies in systemic sclerosis patients . Cross-validation with orthogonal detection methods (Western blotting following IP) further strengthens specificity claims. Finally, quantitative analysis with appropriate statistical thresholds should be applied to distinguish specific interactions from background.

How do anti-PRMT5 antibody levels correlate with disease progression in systemic sclerosis?

Recent research has revealed a significant correlation between anti-PRMT5 antibody levels and disease trajectory in systemic sclerosis (SSc). Patients exhibiting progressive skin fibrosis, defined as a 25% increase in modified Rodnan skin score (mRSS) within a 12-month period, demonstrated elevated serum levels of anti-PRMT5 antibodies compared to non-progressors . Longitudinal analysis revealed a dynamic relationship—anti-PRMT5 antibody titers increased in patients showing disease progression and decreased in those experiencing regression, suggesting these antibodies track with disease activity rather than simply serving as a static diagnostic marker . Quantitatively, anti-PRMT5 antibodies correlated positively with inflammatory markers, including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and tissue inhibitor of metal protease 1 (TIMP-1), linking these antibodies to the broader inflammatory milieu in SSc . Furthermore, patients meeting "active disease" criteria according to the focuSSced study displayed significantly higher anti-PRMT5 antibody levels . The correlation extends to lung involvement—patients with progressive interstitial lung disease (ILD) showed sustained anti-PRMT5 antibody titers, and strikingly, all patients positive for both anti-topoisomerase I antibodies (ATA) and anti-PRMT5 antibodies developed progressive fibrosing ILD within a 24-month follow-up period . These findings collectively demonstrate that anti-PRMT5 antibodies serve as dynamic biomarkers that correlate with both skin and lung disease trajectories in SSc, potentially offering valuable prognostic information beyond traditional serological markers.

What mechanisms might explain the pathogenic role of anti-PRMT5 antibodies in autoimmune conditions?

The pathogenic mechanisms underlying anti-PRMT5 antibodies in autoimmune conditions, particularly systemic sclerosis (SSc), involve complex immunological processes. Experimental evidence from PRMT5-immunized mice demonstrates that these antibodies can directly induce SSc-like manifestations, including significant inflammation and fibrosis in both skin and lungs . At the molecular level, RNA sequencing of affected tissues revealed upregulation of proinflammatory and profibrotic pathways, suggesting these antibodies may trigger or amplify pathological signaling cascades . One potential mechanism involves aberrant exposure of PRMT5 antigen during cell death processes—increased apoptotic PRMT5-positive endothelial cells were observed in the dermis of SSc patients, potentially releasing PRMT5 as a neo-antigen that triggers autoimmune responses . The correlation between anti-PRMT5 antibody levels and inflammatory cytokines, including IL-6, TNF-α, IL-10, and IL-8, further supports their role in promoting inflammatory microenvironments conducive to fibrosis . Additionally, these antibodies may interfere with PRMT5's normal enzymatic functions in arginine methylation, disrupting epigenetic regulation and gene expression patterns in susceptible cell types like fibroblasts, where PRMT5 expression is pronounced in SSc tissues . The strong association with progressive interstitial lung disease, particularly in patients also positive for anti-topoisomerase I antibodies, suggests potential synergistic interactions between different autoantibodies in amplifying tissue damage . These mechanistic insights indicate anti-PRMT5 antibodies may represent more than passive biomarkers—they appear to be active contributors to disease pathogenesis through multiple immunological and fibrotic pathways.

What strategies can resolve discrepancies between different anti-PRMT5 antibody detection methods in research studies?

Resolving discrepancies between different anti-PRMT5 antibody detection methods requires systematic analytical approaches. First, conduct epitope mapping analysis to determine if different antibodies recognize distinct regions of PRMT5—differences may reflect conformation-dependent epitope accessibility rather than assay validity. When comparing ELISA and mass spectrometry results, as seen in SSc studies , implement calibration curves using recombinant PRMT5 standards across both platforms to establish quantitative relationships between signal intensities. For discrepancies between immunohistochemistry and Western blotting, consider that fixation-induced epitope modifications may affect antibody binding in tissue sections; perform parallel analysis of fresh-frozen versus fixed samples from the same source. Cross-validation with orthogonal detection techniques is essential—if microarray and ELISA methods yield conflicting results as observed in some SSc validation cohorts , implement a third independent method like immunoprecipitation-Western blot to arbitrate. Sensitivity differences often explain discrepancies in low-expression contexts; determine detection limits for each method using dilution series of recombinant protein. For clinical samples, pre-absorbing sera with recombinant target protein before testing can confirm specificity across methods. When nuclear versus cytoplasmic fractionation yields different results, validate subcellular localization through immunofluorescence microscopy with appropriate compartment markers. Finally, independent laboratory validation using standardized protocols and blinded sample analysis provides the most rigorous resolution of methodological discrepancies, particularly for novel biomarkers like anti-PRMT5 antibodies in systemic sclerosis .

How should researchers approach validation when developing new assays for detecting anti-PRMT5 antibodies in patient samples?

Developing robust assays for detecting anti-PRMT5 antibodies in patient samples requires comprehensive validation strategies. Initially, establish analytical validation parameters including sensitivity, specificity, precision, and accuracy using reference standards (purified anti-PRMT5 antibodies) and negative controls. Serial dilution experiments are essential to determine optimal conditions for ELISA and calculate reliable area under the curve (AUC) values, as demonstrated in systemic sclerosis studies . Cross-platform validation is crucial—findings from ELISA should be verified using orthogonal methods like protein microarray, as implemented in the independent validation cohorts for anti-PRMT5 antibodies in SSc . When defining positivity thresholds, use the 99th percentile of healthy controls as the upper limit of normal, which yielded 31.11% seropositivity for anti-PRMT5 antibodies in SSc patients with zero false positives in healthy controls . ROC curve analysis should be performed to establish discriminatory power between disease and control groups, with reported AUC values ranging from 0.900 to 0.988 for anti-PRMT5 antibodies distinguishing SSc from healthy controls and other autoimmune conditions . Biological validation requires correlation with established disease phenotypes and trajectories—the association between anti-PRMT5 antibodies and both skin and lung progression in SSc provided compelling biological validation . Longitudinal stability and variation should be assessed through repeated measurements in stable patients to distinguish assay variability from true biological changes. Finally, multi-center validation with standardized protocols is essential before clinical implementation, ensuring reproducibility across different laboratory settings.

What technical challenges exist in differentiating between antibodies targeting PRMT5 and other protein arginine methyltransferases (PRMTs) in research applications?

Differentiating between antibodies targeting PRMT5 and other protein arginine methyltransferases (PRMTs) presents significant technical challenges due to structural and functional homology within this enzyme family. The most fundamental approach involves comprehensive cross-reactivity testing against all PRMT family members (PRMT1-9) using recombinant proteins in controlled assays. Epitope mapping is essential—selecting antibodies targeting less conserved regions of PRMT5 reduces cross-reactivity, while antibodies recognizing highly conserved catalytic domains may detect multiple PRMTs. In the immunoprecipitation-mass spectrometry (IP-MS) studies that identified anti-PRMT5 antibodies in systemic sclerosis, rigorous bioinformatic filtering was necessary to distinguish PRMT5 from other family members . Competitive binding assays, where excess recombinant PRMT1 (the most abundant family member) is used to pre-absorb sera before testing for PRMT5 reactivity, help ensure specificity. Importantly, validation studies for anti-PRMT5 antibodies in SSc demonstrated that anti-PRMT1 antibodies were far less prevalent (only 3.33% positivity compared to 31.11% for PRMT5) , indicating successful differentiation between these related targets. When developing immunohistochemical applications, double-staining with antibodies against different PRMTs helps identify potential cross-reactivity through co-localization analysis. For newly developed antibodies, knockout/knockdown validation is definitive—testing in PRMT5-deficient cells should eliminate specific signal while preserving any cross-reactive binding to other PRMTs. Finally, employing activity-based detection methods that distinguish the symmetric dimethylation catalyzed by PRMT5 from the asymmetric dimethylation by type I PRMTs provides functional differentiation complementary to direct antibody detection.

How might anti-PRMT5 antibodies be leveraged as predictive biomarkers in personalized medicine approaches for systemic sclerosis?

Anti-PRMT5 antibodies show significant potential as predictive biomarkers in personalized medicine approaches for systemic sclerosis (SSc). Their demonstrated correlation with disease trajectory—rising with progression and declining with regression in both skin and lung involvement—positions them as dynamic indicators of disease activity beyond static diagnostic markers . A precision medicine framework could implement serial anti-PRMT5 antibody measurements to stratify patients into risk categories for progressive disease. The finding that patients with double positivity for anti-topoisomerase I and anti-PRMT5 antibodies universally developed progressive fibrosing interstitial lung disease (PF-ILD) within 24 months suggests a powerful prognostic application for treatment intensification in this high-risk subgroup . Quantitative thresholds could be established through larger validation cohorts to define clinically meaningful changes in antibody levels that predict imminent disease flares, allowing for preemptive therapeutic intervention. Integration with other serological and clinical parameters into machine learning algorithms could enhance predictive accuracy—the observed correlations between anti-PRMT5 antibodies and inflammatory markers (ESR, CRP) provide a foundation for such multivariate models . Longitudinal monitoring during therapeutic trials could identify antibody level reductions as early pharmacodynamic markers of treatment response, potentially preceding clinical improvement. Furthermore, the mechanistic link between PRMT5 immunization and SSc-like manifestations in mice suggests that anti-PRMT5 antibody levels might identify patients who would specifically benefit from B-cell targeted therapies or emerging PRMT5 inhibitors. Implementation would require standardized assay development with established reference ranges and clinical decision thresholds, but the groundwork laid by current research indicates significant potential for anti-PRMT5 antibodies in personalizing SSc management.

What research applications could benefit from combining anti-PRMT5 antibodies with CRISPR-Cas9 gene editing technologies?

The integration of anti-PRMT5 antibodies with CRISPR-Cas9 gene editing technologies offers powerful research applications for understanding PRMT5 biology and disease mechanisms. CRISPR-mediated epitope tagging of endogenous PRMT5 creates ideal validation systems for antibody specificity—comparing detection before and after tag insertion provides unambiguous confirmation of antibody performance across applications. For mechanistic studies of SSc, CRISPR-engineered cellular models with modified PRMT5 expression could be exposed to patient-derived anti-PRMT5 antibodies to elucidate downstream signaling effects detected by phospho-specific antibodies. Domain-specific CRISPR editing of PRMT5 could generate variants lacking particular epitopes, helping map the precise binding regions of autoantibodies found in SSc patients . CRISPR screens in fibroblasts or endothelial cells (cell types with increased PRMT5 expression in SSc) could identify genetic modifiers that enhance or diminish cellular responses to anti-PRMT5 antibodies, revealing potential therapeutic targets. For in vivo applications, CRISPR-generated conditional PRMT5 knockout mouse models provide valuable comparison points to the PRMT5-immunized mice that developed SSc-like features , distinguishing effects of protein absence versus antibody-mediated dysfunction. Humanized mouse models with CRISPR-modified immune systems could better recapitulate human autoimmune responses to PRMT5, creating improved platforms for therapeutic testing. Additionally, CRISPR activation (CRISPRa) or interference (CRISPRi) systems targeting PRMT5 regulatory elements, coupled with anti-PRMT5 antibody-based detection methods, could reveal tissue-specific control mechanisms relevant to disease pathophysiology. These combined approaches would substantially advance understanding of PRMT5's role in health and disease beyond what either technology could achieve independently.

How can multi-omics approaches incorporating anti-PRMT5 antibody data enhance our understanding of systemic sclerosis pathogenesis?

Multi-omics approaches integrating anti-PRMT5 antibody data offer transformative potential for understanding systemic sclerosis (SSc) pathogenesis. Combining serum anti-PRMT5 antibody levels with transcriptomics of affected tissues could identify gene expression signatures specifically associated with antibody positivity—the RNA sequencing already performed in PRMT5-immunized mice revealed upregulation of proinflammatory and profibrotic pathways that could serve as a reference for human studies. Epigenomic profiling through techniques like CUT&RUN would be particularly relevant given PRMT5's role in histone arginine methylation, potentially revealing how anti-PRMT5 antibodies disrupt normal epigenetic regulation in disease-relevant cell types. Proteomics of immunoprecipitated complexes from patients stratified by anti-PRMT5 antibody status could identify differential interactome profiles that explain varying clinical trajectories. Metabolomic analysis correlated with antibody levels might uncover altered metabolic pathways in high-antibody states, given PRMT5's known involvement in metabolic regulation. Single-cell multi-omics applied to skin biopsies from patients with varying anti-PRMT5 antibody levels would provide unprecedented resolution of cell-type specific responses, particularly in fibroblasts and endothelial cells where PRMT5 expression is altered in SSc . Spatial transcriptomics and proteomics of tissue sections could map the geographical relationship between anti-PRMT5 antibody deposition, PRMT5 expression, and fibrotic changes. Longitudinal multi-omics sampling during disease progression/regression would create dynamic models of biological changes associated with fluctuating antibody levels, moving beyond static biomarker paradigms. Network analysis integrating these multi-dimensional datasets could identify key regulatory hubs connecting anti-PRMT5 antibodies to downstream pathological processes. This comprehensive systems biology approach would transform understanding of how these newly identified antibodies contribute to the complex pathophysiology of SSc, potentially revealing novel therapeutic targets and patient stratification strategies.

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.