Phospho-POLR2A (S2) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Phospho-POLR2A (S2) Recombinant Monoclonal Antibody

Phospho-POLR2A (S2) Recombinant Monoclonal Antibody is a highly specific tool designed to detect RNA polymerase II subunit A (POLR2A) phosphorylated at serine 2 (S2) within its C-terminal domain (CTD). This antibody is critical for studying transcriptional regulation, as phosphorylation at S2 is a hallmark of transcription elongation and pre-mRNA processing . Produced via recombinant technology, it ensures batch-to-batch consistency and high affinity for its target epitope .

Mechanism of Action

The CTD of POLR2A contains tandem heptapeptide repeats (YSPTSPS), where phosphorylation at S2 facilitates:

  • Transcription elongation by releasing paused RNA polymerase II (Pol II) .

  • Recruitment of mRNA processing factors (e.g., capping enzymes, spliceosomes) .

  • Coordination of chromatin remodeling via interactions with histone modifiers .

This antibody selectively binds the phosphorylated S2 epitope, enabling precise detection in cellular contexts .

Applications and Validation Data

Validated across multiple platforms, this antibody is widely used in:

ApplicationRecommended DilutionKey Findings
Western Blot (WB)1:500–1:5000 Detects ~270 kDa band in HeLa, MCF7, and 293T lysates .
Immunohistochemistry (IHC)1:50–1:200 Strong nuclear staining in human ovarian carcinoma and spleen tissues .
Immunofluorescence (IF)1:20–1:200 Localizes phosphorylated Pol II to transcriptionally active nuclear foci .
Chromatin Immunoprecipitation (ChIP)1:50–1:100 Enriches POLR2A-bound DNA at active gene loci (e.g., GAPDH) .
Flow Cytometry (FCM)1:20–1:100 Quantifies S2 phosphorylation in cell cycle studies .

Product Attributes

ParameterDetails
Host SpeciesRabbit
ClonalityMonoclonal
ReactivityHuman, Mouse, Rat
ImmunogenSynthetic peptide spanning POLR2A CTD phosphorylated at S2
Storage-20°C long-term; 4°C for short-term use

Cross-Reactivity and Selectivity

  • No cross-reactivity with non-phosphorylated CTD or phosphorylated S5/S7/Y1 peptides .

  • Specificity confirmed via lambda phosphatase treatment and peptide competition assays .

Transcriptional Regulation Studies

  • S2 phosphorylation peaks during transcriptional elongation, correlating with histone H3K36me3 marks .

  • Inhibition of S2 phosphorylation disrupts mRNA splicing and export .

Disease Associations

  • Cancer: Overexpression linked to dysregulated Pol II activity in ovarian and breast cancers .

  • Neurological disorders: Altered S2 phosphorylation observed in Alzheimer’s disease models .

Product Specs

Buffer
Rabbit IgG in phosphate-buffered saline (PBS), pH 7.4, containing 150 mM NaCl, 0.02% sodium azide, and 50% glycerol.
Description

This recombinant POLR2A antibody was generated using a novel approach. A synthesized peptide derived from human Phospho-POLR2A (S2) was used to immunize an animal. Positive splenocytes were isolated, and RNA was extracted and reverse-transcribed into cDNA. The POLR2A antibody gene was sequenced and screened, and the heavy and light chain sequences were amplified via PCR and cloned into mammalian expression vectors. Following transfection into mammalian cells, the recombinant POLR2A antibody was purified from the culture medium using affinity chromatography. This antibody exhibits reactivity with human POLR2A protein and is suitable for use in ELISA, Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF), and immunoprecipitation (IP).

POLR2A encodes the largest subunit of RNA polymerase II, responsible for the synthesis of eukaryotic messenger RNA. The POLR2A protein contains a carboxy-terminal domain (CTD) composed of heptapeptide repeats, crucial for polymerase activity. Research suggests several key roles for POLR2A, including:

  • Potential applications in pH-responsive nanoparticles for precise targeting of POLR2A in triple-negative breast cancer (TNBC) with common TP53 genomic alterations.
  • Clinical significance of POLR2A variants dependent on their impact on Pol II-mediated transcription, with loss-of-function variants generally more tolerable than missense mutations.
  • Involvement in cell proliferation and growth promotion, potentially through upregulation of POLR2A and subsequent enhancement of catalytic and transferase activity, as indicated by its interaction with BCAR1.
  • Regulation of tumor cell growth through humanized monoclonal antibody-induced nuclear localization of CD26 and subsequent modulation of POLR2A transcription.
Form
Liquid
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Synonyms
DNA directed RNA polymerase II A antibody; DNA-directed RNA polymerase II largest subunit RNA polymerase II 220 kd subunit antibody; DNA-directed RNA polymerase II subunit A antibody; DNA-directed RNA polymerase II subunit RPB1 antibody; DNA-directed RNA polymerase III largest subunit antibody; hRPB220 antibody; hsRPB1 antibody; POLR2 antibody; Polr2a antibody; POLRA antibody; Polymerase (RNA) II (DNA directed) polypeptide A 220kDa antibody; Polymerase (RNA) II (DNA directed) polypeptide A antibody; RNA polymerase II subunit B1 antibody; RNA-directed RNA polymerase II subunit RPB1 antibody; RPB1 antibody; RPB1_HUMAN antibody; RPBh1 antibody; RpIILS antibody; RPO2 antibody; RPOL2 antibody
Target Names
POLR2A
Uniprot No.

Target Background

Function

DNA-dependent RNA polymerase II (Pol II) catalyzes the transcription of DNA into RNA using ribonucleoside triphosphates as substrates. It is the largest and catalytic component of Pol II, responsible for synthesizing mRNA precursors and various functional non-coding RNAs. The POLR2A subunit forms the polymerase active center in conjunction with the second largest subunit. Pol II is a central component of the basal RNA polymerase II transcription machinery, comprised of mobile elements that exhibit relative movement. RPB1, part of the core element, features a central large cleft, a clamp element regulating cleft opening and closing, and jaws that grasp the incoming DNA template. Transcription initiation involves positioning a single-stranded DNA template strand within the central active site cleft. A bridging helix, emanating from RPB1 and crossing the cleft near the catalytic site, is believed to facilitate Pol II translocation by acting as a ratchet mechanism, moving the RNA-DNA hybrid through the active site. During transcription elongation, Pol II moves along the template as the transcript extends. Elongation is modulated by the phosphorylation state of the CTD of the largest Pol II subunit (RPB1), which serves as a platform for assembling factors regulating transcription initiation, elongation, termination, and mRNA processing. Regulation of gene expression levels depends on the balance between methylation and acetylation of CTD lysines. Acetylation status of the CTD influences initiation or early elongation steps of transcription of growth factor-induced immediate early genes, while methylation and dimethylation exhibit a repressive effect on target gene expression. In the context of microbial infection, POLR2A acts as an RNA-dependent RNA polymerase when associated with the Hepatitis delta virus small delta antigen, functioning as both a replicase and transcriptase for the viral RNA circular genome.

Gene References Into Functions
  1. XPC, an RNA polymerase II cofactor, recruits the ATAC coactivator complex to promoters through interaction with E2F1. PMID: 29973595
  2. Weak, multivalent interactions between TAF15 fibrils and heptads throughout the RNA Pol II CTD collectively mediate complex formation. PMID: 28945358
  3. CDK9 stimulates the release of paused polymerase and activates transcription by increasing the number of transcribing polymerases and thus mRNA synthesis rate. PMID: 28994650
  4. The single nucleotide polymorphism rs2071504 in the POLR2A gene is associated with poor overall and disease-free survival in early-stage non-small cell lung cancer patients. PMID: 28922562
  5. Hydrogen peroxide alters RNA polymerase II (Pol II) occupancy at promoters and enhancers genome-wide. PMID: 28977633
  6. Rpb1/2 dynamics influence the choice between sense and divergent antisense transcription. PMID: 28506463
  7. In Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation, episomes aggregate within large domains, functioning as viral transcription factories where a significant portion of cellular RNA polymerase II is involved in viral genome transcription. PMID: 28331082
  8. Inhibition of VCP/p97 or siRNA-mediated ablation impairs ultraviolet radiation (UVR)-induced RNA polymerase II (RNAPII) degradation. PMID: 28036256
  9. Chromatin-bound EGFR and ERK kinases play a role in RNA polymerase 2 transcription. PMID: 27587583
  10. Recurrent somatic mutations in POLR2A drive meningioma neoplasia. PMID: 27548314
  11. The Elongin A ubiquitin ligase and the CSB protein function together in a common pathway responding to Pol II stalling and DNA damage. PMID: 28292928
  12. CGGBP1 affects the expression of RNA Pol II-transcribed genes through a mechanism dependent on Alu RNA. PMID: 25483050
  13. POLR2A mutations confer elevated sensitivity to pharmacological suppression, impacting interacting proteins like INTS10 and genes involved in mRNA splicing and nonsense-mediated mRNA decay. PMID: 28027311
  14. HIV Tat precisely controls RNA polymerase II recruitment and pause release to fine-tune transcription initiation and elongation. PMID: 26488441
  15. TOP1 becomes fully active after pause-release, coupling carboxyl-terminal-domain (CTD) phosphorylation of RNA polymerase II (RNAPII) with TOP1 stimulation, enhancing its processivity. PMID: 27058666
  16. POLR2A variants are not associated with sporadic Parkinson's disease in the Chinese Han population. PMID: 26432391
  17. POLR2A's CTD is extensively modified by O-GlcNAc transferase (OGT), requiring a minimum of 20 heptad CTD repeats and more than half of OGT's NTD for efficient O-GlcNAcylation. PMID: 26807597
  18. Serine phosphorylation stimulates, while tyrosine phosphorylation inhibits, the protein-binding activity of the RNA Pol II CTD. PMID: 26515650
  19. Co-cultivation of HIV-infected CD4+ cells with antiviral response-expressing CD8+ cells strongly diminishes RNA polymerase II (RNAP II) on the HIV promoter and other viral regions. PMID: 26499373
  20. Ash2L works with P53 promoter occupancy to activate RNA Polymerase II by aiding the formation of a stable transcription pre-initiation complex. PMID: 25023704
  21. RNA polymerase II inhibitors may be useful for targeting dormant leukemia cells. PMID: 23767415
  22. Epstein-Barr virus BcRF1 interacts with a viral pre-initiation complex and cellular RNA polymerase II. PMID: 25165108
  23. E2F-1 forms a complex with RNA polymerase II and protein PURA for transcriptional activation of the secondary promoter. PMID: 24819879
  24. Human CD68 gene expression is associated with changes in Pol II phosphorylation and short-range intrachromosomal gene looping. PMID: 17583472
  25. The NSs protein of Schmallenberg virus (SBV) induces degradation of the RPB1 subunit of RNA polymerase II, inhibiting global cellular protein synthesis and the antiviral response. PMID: 24828331
  26. TCERG1 regulates HIV-1 transcriptional elongation by increasing RNAPII elongation rate and Ser 2 phosphorylation within the CTD. PMID: 24165037
  27. Slow Pol II elongation allows weak splice sites to be recognized, leading to higher inclusion of alternative exons. PMID: 24793692
  28. Sequence-specific double-strand DNA breaks activate positive transcription elongation factor b (P-TEFb), triggering hyperphosphorylation of the Rpb1-CTD and p53-transcriptional axis activation, resulting in cell cycle arrest. PMID: 23906511
  29. Interaction between nuclear CD26 and the POLR2A gene has been observed. PMID: 23638030
  30. RECQL5 interacts with the Rpb1 jaw domain of Pol II, interfering with TFIIS's ability to promote transcriptional read-through. PMID: 23748380
  31. p68/DdX5 immunoprecipitates with RNA polymerase II (RNAP II), facilitating beta-catenin and androgen receptor (AR) transcriptional activity in prostate cancer cells. PMID: 23349811
  32. Inhibition of the transition of paused RNA Pol II to productive elongation, as seen with p21(CIP1), is a mechanism by which transcription factor Sp3 fine-tunes gene expression. PMID: 23401853
  33. RNA polymerase II acts as an RNA-dependent RNA polymerase to extend and destabilize a non-coding RNA. PMID: 23395899
  34. Polyamide treatment activates p53 signaling and depletes the RNA polymerase II (RNAP2) large subunit RPB1. PMID: 23319609
  35. CTCF binding sites regulate mRNA production, RNA polymerase II (RNAPII) programming, and nucleosome organization of the Kaposi's sarcoma-associated herpesvirus latency transcript control region. PMID: 23192870
  36. Site-specific p65 phosphorylation targets NF-kappaB activity to specific gene subsets by influencing p65 and p-RNAP II promoter recruitment. PMID: 23100252
  37. BRD4-driven Pol II phosphorylation at serine 2 regulates lineage-specific gene transcription in human CD4+ T cells. PMID: 23086925
  38. SNAPC1 is a general transcriptional coactivator that functions through RNAPII elongation. PMID: 22966203
  39. Cyclin K1 is essential for CDK12/CrkRS activation to phosphorylate the C-terminal domain of RNA Pol II. PMID: 22988298
  40. The super elongation complex (SEC) is required for rapid transcriptional induction, with or without paused RNA polymerase II (Pol II). PMID: 22895430
  41. The RNA polymerase II CTD and O-GlcNAc regulate transcription initiation. PMID: 22605332
  42. Polo-like kinase 3 phosphorylates Thr4 in mammalian cells. PMID: 22549466
  43. Activator-induced structural shifts within Mediator trigger activation of stalled Pol II. PMID: 21326907
  44. Mediator structural shifts induced by activator binding help orient Pol II before transcription initiation. PMID: 22343046
  45. Phosphorylation of Rpb1 CTD Thr4 residues is required for histone mRNA 3' end processing, facilitating recruitment of 3' processing factors to histone genes. PMID: 22053051
  46. Parcs/Gpn3 is critical for nuclear RNAP II accumulation, impacting cell proliferation. PMID: 21782856
  47. Studies examine the kinetics of RNA polymerase II elongation during co-transcriptional splicing. PMID: 21264352
  48. MicroRNA promoter identification based on RPol II binding patterns provides temporal and spatial measurements of transcription initiation. PMID: 21072189
  49. Deregulation of cellular NIPP1/PP1 holoenzyme affects RNAPII phosphorylation, suggesting NIPP1's role in RNAPII-mediated transcription. PMID: 20941529
  50. Elevated PHD1 and decreased PHD2 are causatively linked to Rpb1 hydroxylation and oncogenesis in human renal clear cell carcinomas with WT VHL gene. PMID: 20978146
Database Links

HGNC: 9187

OMIM: 180660

KEGG: hsa:5430

STRING: 9606.ENSP00000314949

UniGene: Hs.270017

Protein Families
RNA polymerase beta' chain family
Subcellular Location
Nucleus. Cytoplasm. Chromosome.

Q&A

What is Phospho-POLR2A (S2) and why is it important in transcription studies?

Phospho-POLR2A (S2) refers to the phosphorylated form of RNA polymerase II's largest catalytic subunit (POLR2A, also known as RPB1) at the serine 2 position within its C-terminal domain (CTD) heptapeptide repeat sequence (YSPTSPS). This specific phosphorylation plays a crucial role in regulating transcription elongation and mRNA processing mechanisms. The CTD of POLR2A contains multiple repeats of this heptapeptide sequence, and the phosphorylation pattern across these repeats creates a "CTD code" that recruits different factors during transcription progression. Specifically, S2 phosphorylation is associated with productive elongation phases of transcription and facilitates the recruitment of splicing factors and other RNA processing machinery. Using antibodies specific to this phosphorylation state allows researchers to track transcriptionally active polymerase complexes and study elongation dynamics across genes .

What applications have been validated for Phospho-POLR2A (S2) antibodies?

Phospho-POLR2A (S2) antibodies have been rigorously validated across multiple experimental platforms, making them versatile tools for transcription research. According to validation data, these antibodies can be successfully employed in:

ApplicationValidated DilutionNotes
Western Blot (WB)1:500 - 1:2000Detects ~270kDa band
Immunohistochemistry (IHC-P)1:50 - 1:200Paraffin-embedded tissues
Immunofluorescence (IF/ICC)1:50 - 1:200Shows nuclear localization
Chromatin Immunoprecipitation (ChIP)5μg antibody for 10-15μg chromatinFor transcriptional regulation studies
ChIP-sequencing1:50 - 1:100Genome-wide occupancy analysis
CUT&Tag10^5 cells /1 μgHigher resolution alternative to ChIP
ELISAAs recommended by manufacturerFor quantitative analysis

These protocols have been tested in multiple cell lines including MCF7, HeLa, C2C12, and C6, demonstrating cross-reactivity across human, mouse, and rat samples .

How can I validate the specificity of a Phospho-POLR2A (S2) antibody?

Validating antibody specificity is essential for reliable experimental outcomes. For Phospho-POLR2A (S2) antibodies, implement the following validation strategy:

  • Phosphatase treatment control: Treat a portion of your cell lysate with calf intestinal phosphatase (CIP) to remove phosphorylation. Western blot comparison between treated and untreated samples should show signal reduction in the treated lanes, confirming phospho-specificity. Published data shows successful validation using CIP treatment (20μL/400μL) at 37°C for 1 hour in MCF7, HeLa, C2C12, and C6 cell lines .

  • Immunofluorescence co-localization: Perform dual staining with antibodies against total POLR2A and phospho-specific POLR2A (S2). Overlap in nuclear signals with distinct enrichment patterns provides spatial validation of specificity.

  • ChIP-sequencing profile analysis: Phospho-POLR2A (S2) should show characteristic enrichment patterns with higher signal in gene bodies compared to promoters, distinguishing it from Phospho-POLR2A (S5) which shows promoter enrichment. Validated antibodies demonstrate specific enrichment patterns at representative gene loci like GAPDH .

  • Knockdown/knockout validation: Use siRNA against POLR2A or inhibitors of CTD kinases (like CDK9 inhibitors) to reduce the target signal, confirming specificity of detection.

What are essential controls for experiments using Phospho-POLR2A (S2) antibodies?

Robust experimental design requires appropriate controls to ensure valid interpretation of results. When working with Phospho-POLR2A (S2) antibodies, incorporate these essential controls:

How should I optimize immunofluorescence protocols for Phospho-POLR2A (S2) detection?

Immunofluorescence with Phospho-POLR2A (S2) antibodies requires careful optimization to preserve phosphorylation status and nuclear architecture. Follow these methodological recommendations:

  • Fixation protocol: Use freshly prepared 4% paraformaldehyde for 10-15 minutes at room temperature. Overfixation can mask epitopes, while underfixation may cause nuclear material loss.

  • Permeabilization: Gentle permeabilization with 0.2% Triton X-100 for 5-10 minutes preserves nuclear structure while allowing antibody access. Harsher detergents may extract nuclear proteins.

  • Blocking buffer: Use 3-5% BSA in PBS with 0.1% Tween-20 to reduce background. For neuronal cell lines like PC-12, blocking time may need extension to 1-2 hours.

  • Antibody dilution: Optimal dilution for Phospho-POLR2A (S2) antibodies in IF applications is between 1:50 and 1:200. For sensitive detection in PC-12 cells, a 1:100 dilution has been validated with Cy3-conjugated secondary antibodies .

  • Nuclear counterstain: DAPI works effectively as a nuclear counterstain and helps visualize nuclear architecture. Phospho-POLR2A (S2) should show punctate nuclear staining in transcriptionally active cells.

  • Mounting medium: Use anti-fade mounting medium to prevent photobleaching during imaging and analysis.

What factors affect POLR2A S2 phosphorylation levels in experimental systems?

Understanding factors that influence POLR2A S2 phosphorylation is critical for experimental design and data interpretation. Key modulators include:

  • Cell cycle phase: S2 phosphorylation levels fluctuate throughout the cell cycle, with higher levels during S and G2 phases when transcriptional activity increases. Synchronize cells or perform cell cycle analysis in parallel for accurate comparisons.

  • Transcriptional inhibitors: CDK9 inhibitors (flavopiridol, DRB) rapidly reduce S2 phosphorylation by blocking the kinase responsible for this modification. These can serve as experimental tools or controls.

  • Splicing inhibitors: Interestingly, splicing inhibitors like madrasin can indirectly affect POLR2A levels. Research shows that interfering with splicing machinery can lead to intron retention in POLR2A transcripts, reducing its protein levels and consequently affecting S2 phosphorylation patterns .

  • XAB2 levels: XAB2 (XPA-binding protein 2) depletion induces severe splicing defects in POLR2A with significant intron retention, leading to substantial loss of POLR2A at both RNA and protein levels. This subsequently impairs global transcription and can confound phosphorylation studies .

  • Cellular stress: Various stressors including DNA damage, oxidative stress, and heat shock can alter the CTD phosphorylation pattern by activating stress-responsive kinases and phosphatases.

How does ChIP-seq with Phospho-POLR2A (S2) antibodies inform our understanding of transcription dynamics?

ChIP-seq using Phospho-POLR2A (S2) antibodies provides genome-wide insights into active transcription and has revolutionized our understanding of transcriptional regulation. This methodology reveals:

  • Elongation dynamics: S2 phosphorylation increases as RNA polymerase II proceeds from promoter into gene body, creating a characteristic profile that rises through the gene and often extends beyond the polyadenylation site. This pattern distinguishes actively transcribed genes from paused or inactive genes.

  • Correlation with mRNA processing: Peaks of S2 phosphorylation often correlate with exon-intron boundaries and RNA processing sites, reflecting the role of this modification in coupling transcription with mRNA processing.

  • Enhancer transcription: Low-level S2 phosphorylation at enhancers indicates enhancer RNA (eRNA) transcription, helping identify active enhancer elements genome-wide.

  • Protocol optimization: For optimal results, use 5μg antibody for 10-15μg of cross-linked chromatin. Following ChIP, construct sequencing libraries using standard protocols with appropriate size selection (150-300bp fragments) .

  • Bioinformatic analysis: Analyze ChIP-seq data using peak-calling algorithms optimized for broad peaks rather than sharp transcription factor binding sites. Normalize to input and compare with total POLR2A profiles to distinguish changes in phosphorylation from changes in occupancy.

ChIP-seq experiments with Phospho-POLR2A (S2) antibodies can identify enrichment patterns at specific genomic regions, as demonstrated in studies showing characteristic profiles at representative gene loci like GAPDH .

What are the advantages of CUT&Tag over ChIP-seq for studying POLR2A S2 phosphorylation?

CUT&Tag (Cleavage Under Targets and Tagmentation) represents a significant methodological advancement for epigenomic profiling, offering several advantages over traditional ChIP-seq for studying POLR2A S2 phosphorylation:

  • Higher sensitivity: CUT&Tag requires significantly fewer cells (as low as 10^5 cells with 1μg of Phospho-POLR2A (S2) antibody) compared to ChIP-seq, making it suitable for rare cell populations or limited samples .

  • Improved signal-to-noise ratio: CUT&Tag typically yields cleaner data with lower background, as the tagmentation reaction occurs only at antibody-bound sites within the native chromatin environment.

  • Preserved nuclear architecture: The method uses intact cells/nuclei rather than sonicated chromatin, potentially preserving higher-order chromatin structures relevant to transcription regulation.

  • Faster protocol: CUT&Tag can be completed in 1-2 days versus 3-4 days for ChIP-seq, with fewer washing steps and handling losses.

  • Recommended protocol: Successful CUT&Tag has been performed using the CUT&Tag Assay Kit (pAG-Tn5) from 10^5 HeLa cells with 1μg Phospho-POLR2A CTD-S2 Rabbit mAb, along with a secondary Goat Anti-Rabbit IgG(H+L). This approach revealed characteristic enrichment patterns at representative gene loci like GAPDH .

When deciding between methods, consider sample availability, required resolution, and compatibility with downstream analyses. For limited samples or higher resolution needs, CUT&Tag offers advantages, while ChIP-seq remains valuable for cross-comparison with existing datasets.

How does XAB2 depletion specifically affect POLR2A and what mechanisms underlie this relationship?

Research has uncovered a critical relationship between XAB2 (XPA-binding protein 2) and POLR2A expression that impacts global transcription. The mechanistic details include:

  • Splicing regulation: XAB2 depletion leads to severe splicing defects in POLR2A transcripts, characterized by significant intron retention. This splicing disruption results in substantial loss of POLR2A at both RNA and protein levels, subsequently impairing global transcription .

  • mRNA surveillance pathway involvement: Proteomics analysis after XAB2 depletion revealed upregulation of proteins involved in mRNA surveillance, including Dom34 (PELO). Knockdown of Dom34 partially rescues POLR2A expression by stabilizing its mRNA, suggesting that aberrantly spliced POLR2A transcripts are targeted for degradation by mRNA surveillance mechanisms .

  • Spliceosome component interactions: Immunoprecipitation experiments confirmed that XAB2 associates with spliceosome components critical for POLR2A expression. Depletion of specific factors including SNRNP200, EFTUD2, SNW1, PRPF8, PLRG1, and AQR reduces POLR2A expression, indicating a specialized splicing machinery requirement .

  • Domain mapping insights: The TPR motifs 2-4 and 11 of XAB2 are particularly important for POLR2A expression, functioning through interaction with SNW1. This structural information provides mechanistic insight into how XAB2 regulates POLR2A processing .

  • Cellular senescence connection: XAB2 or POLR2A depletion induces cellular senescence by upregulating p53 and p21. Re-expression of POLR2A after XAB2 depletion alleviates cellular senescence, positioning POLR2A as a major mediator of senescence induced by XAB2 deficiency .

This complex regulatory relationship highlights how splicing defects in a single critical gene (POLR2A) can profoundly impact global transcription and cellular aging pathways.

What is the relationship between POLR2A S2 phosphorylation and cellular senescence?

POLR2A S2 phosphorylation has emerged as a critical regulator in cellular senescence pathways through several interconnected mechanisms:

  • Global transcription maintenance: POLR2A S2 phosphorylation is essential for productive transcription elongation. Research demonstrates that disruption of POLR2A expression or function leads to impaired global transcription, triggering stress responses that culminate in cellular senescence .

  • p53-p21 pathway activation: Depletion of POLR2A induces significant cellular senescence markers, with approximately 49.7% of cells showing positive SA-β-gal staining compared to control levels. This senescence phenotype correlates with upregulation of p53 and p21, key mediators of cell cycle arrest and senescence .

  • Cell cycle effects: POLR2A deficiency results in cell cycle arrest predominantly at the G2/M phase and inhibits cell proliferation, characteristic features of senescent cells .

  • Rescue experiments: Importantly, re-expression of POLR2A after depletion of upstream regulators like XAB2 alleviates cellular senescence, confirming POLR2A's central role in this process rather than being a secondary effect .

  • Transcription-coupled stress signaling: Disruption of POLR2A function may trigger transcription-coupled stress signaling pathways that activate p53, potentially through R-loop formation or other transcriptional stress mechanisms.

These findings position POLR2A and its phosphorylation state as central regulators in the cellular senescence program, linking transcriptional regulation directly to cellular aging pathways. This connection has significant implications for understanding age-related pathologies and potential therapeutic interventions.

How do alterations in POLR2A S2 phosphorylation patterns contribute to disease states?

Dysregulation of POLR2A S2 phosphorylation has been implicated in various pathological conditions through disruption of transcriptional regulation:

  • Cancer biology: Alterations in POLR2A phosphorylation patterns can drive aberrant gene expression programs in cancer cells. Cancer-specific transcriptional dependencies may emerge from changes in CTD phosphorylation, creating potential therapeutic vulnerabilities. Research confirms that dysregulation of POLR2A phosphorylation has been implicated in various diseases, including cancer .

  • Neurological disorders: Proper POLR2A function and phosphorylation is critical for neuronal gene expression programs. Disruption of POLR2A regulation has been associated with neurological conditions, highlighting its importance in neural development and function .

  • Senescence-related pathologies: Given POLR2A's role in cellular senescence pathways, dysregulation may contribute to age-related diseases. Research shows that XAB2 depletion causes POLR2A reduction and induces cellular senescence through p53/p21 activation, potentially connecting to age-related tissue dysfunction .

  • Developmental disorders: As a master regulator of transcription, POLR2A phosphorylation abnormalities may underlie certain developmental disorders through global perturbation of gene expression programs.

  • Stress response pathways: POLR2A phosphorylation serves as an integration point for cellular stress signals. Chronic dysregulation may contribute to stress-related pathologies through persistent activation or inhibition of stress-responsive genes.

Understanding these connections offers potential therapeutic opportunities, including the development of compounds that modulate CTD phosphorylation or target specific transcriptional dependencies in disease states.

What are the most common technical challenges when working with Phospho-POLR2A (S2) antibodies?

Researchers face several technical challenges when working with Phospho-POLR2A (S2) antibodies that can impact experimental outcomes:

How can I optimize Western blot protocols for detecting Phospho-POLR2A (S2)?

Western blot detection of Phospho-POLR2A (S2) requires careful optimization due to the high molecular weight and phosphorylation-specific nature of the target. Follow these methodological recommendations:

  • Sample preparation: Lyse cells directly in SDS sample buffer containing phosphatase inhibitors (10mM sodium fluoride, 1mM sodium orthovanadate, 10mM β-glycerophosphate) to immediately preserve phosphorylation status.

  • Gel selection: Use 6-8% polyacrylamide gels to properly resolve the ~270kDa POLR2A protein. Commercial gradient gels (4-15%) can also work well for this high molecular weight protein .

  • Transfer conditions: Perform wet transfer at low voltage (30V) overnight at 4°C to ensure complete transfer of high molecular weight proteins. Use 0.45μm PVDF membrane rather than 0.2μm for better retention of large proteins.

  • Blocking conditions: Block membranes with 3% BSA in TBST rather than milk, as milk contains phosphatases that can remove the phosphorylation being detected .

  • Antibody dilution: Optimal results have been achieved with 1:1000 dilution of primary antibody and 1:10000 dilution of HRP-conjugated secondary antibody (goat anti-rabbit IgG). Incubate primary antibody overnight at 4°C for best results .

  • Detection system: Use ECL detection systems with appropriate sensitivity. For weak signals, enhanced chemiluminescence substrates or longer exposure times may be necessary. Published protocols have successfully used ECL Basic Kit with 10-second exposure time .

  • Controls: Always include a phosphatase-treated control sample (using CIP treatment: 20μL/400μL at 37°C for 1 hour) to confirm phospho-specificity of detection .

What methodological approaches can resolve contradictory data in POLR2A phosphorylation studies?

When faced with contradictory results in POLR2A phosphorylation studies, employ these methodological approaches to resolve discrepancies:

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 2024 Thebiotek. All Rights Reserved.