POLR1E 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
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for specific delivery time information.
Synonyms
A49 antibody; DNA directed RNA polymerase I subunit RPA49 antibody; DNA-directed RNA polymerase I subunit E antibody; DNA-directed RNA polymerase I subunit RPA49 antibody; FLJ 13390 antibody; FLJ13970 antibody; PAF53 antibody; POLR 1 E antibody; POLR1E antibody; Polymerase (RNA) I associated factor 1 antibody; Polymerase (RNA) I polypeptide E antibody; Polymerase (RNA) I polypeptide E, 53kDa antibody; PRAF1 antibody; RNA polymerase I associated factor 1 antibody; RNA polymerase I associated factor 53 antibody; RNA polymerase I polypeptide E antibody; RNA polymerase I subunit A49 antibody; RNA polymerase I-associated factor 1 antibody; RNA polymerase I-associated factor 53 antibody; RP11405L18.3 antibody; RPA49_HUMAN antibody
Target Names
POLR1E
Uniprot No.

Target Background

Function
DNA-dependent RNA polymerase catalyzes the transcription of DNA into RNA, utilizing the four ribonucleoside triphosphates as substrates. It is a component of RNA polymerase I, responsible for synthesizing ribosomal RNA precursors. This enzyme appears to play a role in the formation of the initiation complex at the promoter by mediating the interaction between Pol I and UBTF/UBF.
Gene References Into Functions
  1. Nucleolar retention requires binding of SIRT7 to nascent pre-rRNA, linking the spatial distribution of SIRT7 and deacetylation of PAF53 to ongoing transcription. PMID: 24207024
Database Links

HGNC: 17631

KEGG: hsa:64425

STRING: 9606.ENSP00000367029

UniGene: Hs.591087

Protein Families
Eukaryotic RPA49/POLR1E RNA polymerase subunit family
Subcellular Location
Nucleus, nucleolus.

Q&A

What is POLR1E and why is it significant in cellular biology?

POLR1E (also known as PAF53, PRAF1, or RPA49) is the DNA-directed RNA polymerase I subunit E, a critical component of RNA polymerase I (Pol I) complex. This 53 kDa protein plays an essential role in ribosomal RNA (rRNA) synthesis, which constitutes approximately 60% of cellular RNA production . The full-length protein is 481 amino acids with a molecular weight of 53,962 Da . POLR1E is particularly significant because Pol I activity controls the level of ribosome biogenesis and cell growth, making it relevant to various pathological conditions including cancer and neurodegenerative disorders .

What are the common applications for POLR1E antibodies in research?

POLR1E antibodies are utilized across multiple experimental techniques:

ApplicationTypical Dilution RangeExamples of Validated Use
Western Blot (WB)1:2000-1:12000Detected in HeLa, Jurkat, MCF-7 cells and human brain tissue
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg protein lysateValidated in Jurkat cells
Immunohistochemistry (IHC)1:20-1:200Successfully used in human liver cancer tissue
Immunofluorescence (IF)Varies by antibodyDocumented in multiple published studies
Chromatin Immunoprecipitation (ChIP)Varies by protocolUsed to analyze DNA-protein interactions

These applications enable researchers to investigate POLR1E expression, localization, interactions, and function in various cellular contexts .

What validation methods should be used before implementing a POLR1E antibody in a new experimental system?

Before using a POLR1E antibody in a new experimental system, researchers should conduct comprehensive validation:

  • Positive and negative controls: Use cell lines with known POLR1E expression (e.g., HeLa, Jurkat) as positive controls, and consider knockout/knockdown samples as negative controls .

  • Application-specific validation:

    • For WB: Verify a single band of appropriate molecular weight (47-53 kDa)

    • For IHC/IF: Confirm specific staining pattern (predominantly nucleolar)

    • For IP: Demonstrate specific pull-down by mass spectrometry or western blot

  • Cross-reactivity assessment: Test specificity across species if working with non-human models .

  • Reproducibility testing: Ensure consistent results across multiple experiments and biological replicates .

It's recommended to document all validation steps meticulously, as this will strengthen the reliability of subsequent experimental findings .

How can POLR1E antibodies be used to investigate nucleolar stress in neurodegenerative research?

POLR1E antibodies can be valuable tools for studying nucleolar stress in neurodegenerative disorders through several approaches:

  • Nucleolar morphology assessment:

    • Use immunofluorescence with POLR1E antibodies to visualize nucleolar integrity in neuronal models

    • Compare nucleolar structure between healthy and diseased states

  • Real-time monitoring of RNA Pol I activity:

    • Implement LiveArt (live imaging-based analysis of rDNA transcription) techniques in conjunction with POLR1E antibodies to track rRNA synthesis dynamics

    • This approach can reveal altered Pol I activity characteristic of neurodegeneration

  • Protein-protein interaction studies:

    • Use POLR1E antibodies in proximity ligation assays or co-immunoprecipitation to identify altered interactions with other nucleolar proteins in disease states

  • Therapeutic target assessment:

    • Evaluate the effects of Pol I inhibitors on nucleolar stress using POLR1E antibodies as readouts

    • This is particularly relevant as Pol I inhibition has been proposed as a viable strategy for treating neurodegenerative disorders

Researchers should be aware that abnormalities in Pol I activity may contribute to nuclear and nucleolar stress, DNA damage, and neuronal death, making POLR1E an important marker in understanding the pathophysiology of neurodegenerative conditions .

What are the technical challenges in distinguishing POLR1E from other RNA polymerase subunits?

Distinguishing POLR1E from other RNA polymerase subunits presents several technical challenges:

  • Structural homology: Five Pol I subunits (RPABC1, RPABC2, RPABC3, RPABC4, and RPABC5) are common to all three polymerases (Pol I, II, and III), creating potential cross-reactivity issues .

  • Epitope selection considerations:

    • Antibodies raised against conserved regions may cross-react with similar subunits

    • The immunogen sequence is critical; for example, Sigma's POLR1E antibody uses a specific peptide sequence (LLQFPLGQDPSFLAIPILALPPSDSLVPPYIVWYIVWPSALISFLGCTLTVQFSNGKLQSPGNMRFTLYENKDSTNPRKRNQRILAAETDRLSYVGNNFG) to ensure specificity

  • Validation approaches for ensuring specificity:

    • Immunodepletion experiments with recombinant proteins

    • Comparative immunoprecipitation followed by mass spectrometry

    • CRISPR/Cas9-mediated knockout of POLR1E followed by antibody testing

  • Detection strategies:

    • Use two or more antibodies targeting different epitopes of POLR1E

    • Combine with antibodies against other Pol I-specific subunits to confirm complex identity

    • Implement subcellular fractionation since POLR1E is predominantly nucleolar, while other polymerase subunits may have different distributions

The choice of experimental method also impacts specificity; for example, ChIP applications may require more stringent validation than western blotting due to potential cross-linking artifacts .

How does phosphorylation affect POLR1E detection and function in experimental systems?

Phosphorylation of POLR1E can significantly impact both its detection and functional properties:

  • Detection considerations:

    • Phosphorylation may alter antibody binding affinity, particularly if the epitope contains or is adjacent to phosphorylation sites

    • Some antibodies show preferential binding to phosphorylated forms, as demonstrated in research with other RNA polymerase antibodies

    • Phosphatase treatment of samples prior to analysis can help determine if observed molecular weight shifts are due to phosphorylation

  • Functional implications:

    • Phosphorylation regulates Pol I activity, with evidence suggesting that phosphorylated enzymes may have altered binding properties with antibodies

    • Studies on RNA pol I antibodies from TSK mice showed better binding to phosphorylated enzymes

  • Methodological strategies:

    • Use phospho-specific antibodies when investigating specific phosphorylation states

    • Implement lambda phosphatase treatments as controls

    • Consider combining with phospho-enrichment techniques when studying phosphorylation-dependent processes

  • Data interpretation:

    • Multiple bands in Western blots may represent different phosphorylation states

    • Shifts in apparent molecular weight (47-53 kDa observed range for POLR1E) may indicate post-translational modifications

Researchers should be aware that phosphorylation status can be altered by experimental conditions, including cell lysis methods and buffer compositions, potentially affecting POLR1E antibody performance .

What are the optimal protocols for using POLR1E antibodies in immunohistochemistry?

For optimal immunohistochemistry (IHC) results with POLR1E antibodies, the following methodological approach is recommended:

  • Sample preparation:

    • Fixation: 10% neutral buffered formalin (24-48 hours) is generally effective

    • Embedding: Paraffin embedding with standard protocols

    • Sectioning: 4-5 μm thickness provides optimal staining

  • Antigen retrieval:

    • Primary recommendation: TE buffer pH 9.0 with heat-induced epitope retrieval

    • Alternative method: Citrate buffer pH 6.0 for 20 minutes in pressure cooker/microwave

  • Antibody application protocol:

    • Blocking: 3-5% normal serum (matched to secondary antibody species) or commercial blocking solution

    • Primary antibody: Dilute POLR1E antibody 1:20-1:200 (optimal dilution is antibody-specific)

    • Incubation: Overnight at 4°C or 1-2 hours at room temperature

    • Secondary detection: HRP-polymer system or avidin-biotin complex method

    • Chromogen: DAB (3,3'-diaminobenzidine) for 5-10 minutes

    • Counterstain: Hematoxylin (light)

  • Critical controls:

    • Positive control: Human liver cancer tissue has been validated for POLR1E antibodies

    • Negative control: Primary antibody omission or isotype control

    • Competing peptide control: Pre-incubation with immunogen peptide should eliminate specific staining

  • Expected staining pattern:

    • Predominantly nucleolar staining with potential additional nuclear signal

    • Intensity varies by tissue type and physiological state

Fine-tuning antibody concentration for each specific tissue type is recommended, with initial testing using a dilution series to determine optimal signal-to-noise ratio .

What are the best practices for optimizing Western blot protocols with POLR1E antibodies?

Optimizing Western blot protocols for POLR1E detection requires attention to several critical parameters:

  • Sample preparation:

    • Lysis buffer: RIPA or NP-40 based buffers with protease inhibitors

    • Include phosphatase inhibitors to preserve phosphorylation states

    • Sonication recommended to ensure complete nuclear protein extraction

    • Heat samples at 95°C for 5 minutes in reducing Laemmli buffer

  • Gel electrophoresis and transfer:

    • Protein loading: 20-50 μg total protein per lane

    • Gel percentage: 10-12% SDS-PAGE provides optimal resolution for 47-53 kDa POLR1E

    • Transfer conditions: Semi-dry or wet transfer (90 minutes at 100V or overnight at 30V)

    • Membrane: PVDF (0.45 μm) preferred over nitrocellulose for nuclear proteins

  • Antibody incubation:

    • Blocking: 5% non-fat dry milk in TBST (1 hour at room temperature)

    • Primary antibody: 1:2000-1:12000 dilution in 5% BSA/TBST

    • Incubation: Overnight at 4°C with gentle agitation

    • Washing: 4 × 5 minutes with TBST

    • Secondary antibody: Anti-rabbit HRP at 1:5000-1:10000 (1 hour at room temperature)

  • Detection and troubleshooting:

    • Enhanced chemiluminescence (ECL) recommended

    • Expected molecular weight: 47-53 kDa (variation may indicate different isoforms or post-translational modifications)

    • Positive controls: HeLa, Jurkat, or MCF-7 cell lysates

    • Reduce background: If high background occurs, try increasing blocking time or using alternative blocking agents

  • Quantification considerations:

    • Use housekeeping proteins appropriate for nuclear fraction (e.g., Lamin B1)

    • Perform linear range determination for accurate quantification

    • Consider stripping and reprobing for multiple targets

The protocol should be optimized for each specific cell line or tissue, as POLR1E expression levels and modifications may vary across different biological contexts .

How can researchers implement POLR1E antibodies in chromatin immunoprecipitation (ChIP) studies?

Implementing POLR1E antibodies in ChIP studies requires careful consideration of the following methodological aspects:

  • Experimental design considerations:

    • POLR1E ChIP primarily targets active rDNA transcription sites

    • Consider parallel ChIP with other Pol I subunits (e.g., RPA194) to confirm findings

    • Include appropriate controls: IgG negative control and positive control for active transcription (e.g., H3K4me3)

  • Optimized ChIP protocol:

    • Crosslinking: 1% formaldehyde for 10 minutes at room temperature

    • Sonication: Optimize conditions to achieve 200-500 bp fragments

    • Pre-clearing: Incubate chromatin with protein A/G beads before antibody addition

    • Antibody amount: 2-5 μg per ChIP reaction (adjust based on antibody efficiency)

    • Incubation: Overnight at 4°C with rotation

    • Washing: Stringent washing steps to reduce background

    • Elution and reverse crosslinking: Standard protocols

    • DNA purification: Column-based methods preferred for consistency

  • Analysis approaches:

    • qPCR primers: Design primers targeting rDNA promoter regions, transcribed regions, and terminator elements

    • Sequencing: ChIP-seq allows genome-wide analysis of POLR1E binding

    • Data normalization: Use input normalization and appropriate peak calling algorithms

  • Validation and controls:

    • Perform biological replicates to ensure reproducibility

    • Include treatment controls (e.g., Pol I inhibitors like CX-5461) to validate specificity

    • Consider sequential ChIP (re-ChIP) to analyze co-occupancy with other transcription factors

  • Applications in research contexts:

    • Investigation of rDNA transcription regulation

    • Analysis of nucleolar stress responses

    • Study of cancer-related alterations in Pol I activity

    • Examination of drug effects on rRNA synthesis

Researchers should be aware that POLR1E ChIP efficiency may vary depending on cell type, fixation conditions, and antibody batch, necessitating optimization for each experimental system .

How should researchers interpret multiple bands in Western blots when using POLR1E antibodies?

When encountering multiple bands in Western blots using POLR1E antibodies, researchers should consider these interpretation approaches:

  • Expected pattern interpretation:

    • Primary band: Should appear at 47-53 kDa (the expected molecular weight range for POLR1E)

    • Minor bands: May represent:

      • Alternative splice variants (POLR1E has multiple isoforms)

      • Post-translational modifications (phosphorylation, ubiquitination)

      • Proteolytic fragments (particularly if bands are smaller than expected)

  • Systematic analysis approach:

    • Compare band patterns across different cell lines/tissues

    • Correlate with mRNA expression data (e.g., RT-PCR of different transcripts)

    • Perform immunoprecipitation followed by mass spectrometry to identify proteins in each band

    • Use phosphatase treatment to determine if higher molecular weight bands are due to phosphorylation

  • Validation experiments:

    • siRNA/shRNA knockdown: Should reduce intensity of specific bands

    • Peptide competition: Pre-incubation with immunogenic peptide should block specific signals

    • Compare results from multiple antibodies targeting different POLR1E epitopes

  • Troubleshooting persistent non-specific bands:

    • Optimize blocking conditions (try 5% BSA instead of milk)

    • Increase washing stringency and duration

    • Adjust antibody concentration (try higher dilutions)

    • Consider alternative extraction methods to reduce non-specific protein interactions

The presence of multiple bands is not necessarily indicative of poor antibody quality but requires careful validation to ensure correct interpretation of experimental results .

What controls are essential when using POLR1E antibodies in immunofluorescence studies?

For immunofluorescence studies using POLR1E antibodies, the following control experiments are essential:

  • Primary controls:

    • Negative controls:

      • Primary antibody omission (secondary antibody only)

      • Isotype control (non-specific IgG from same species as primary)

      • Peptide competition (pre-absorb antibody with immunizing peptide)

    • Positive controls:

      • Cell lines with known POLR1E expression (e.g., HeLa cells)

      • Co-staining with established nucleolar markers (e.g., Fibrillarin)

  • Expression modulation controls:

    • siRNA/shRNA knockdown to verify specificity

    • Pol I inhibitor treatment (e.g., CX-5461) to observe expected changes in localization

    • Stress response controls (e.g., actinomycin D treatment disrupts nucleolar structure)

  • Technical controls:

    • Autofluorescence control (no antibodies)

    • Channel bleed-through controls for multi-color imaging

    • Fixed vs. live cell comparisons if applicable

  • Expected staining pattern verification:

    • POLR1E should predominantly localize to nucleoli

    • During mitosis, staining pattern should change consistent with nucleolar disassembly

    • After rRNA synthesis inhibition, reorganization of nucleolar components should be observed

  • Advanced validation approaches:

    • Super-resolution microscopy to confirm precise subcellular localization

    • FRAP (Fluorescence Recovery After Photobleaching) to assess protein dynamics

    • Correlative light-electron microscopy to verify nucleolar substructure localization

Documentation of these controls strengthens result interpretation and should be included in publications to demonstrate antibody specificity and proper experimental design .

How can researchers address discrepancies in POLR1E antibody performance across different applications?

When encountering discrepancies in POLR1E antibody performance across different applications, researchers should implement a systematic troubleshooting approach:

  • Application-specific optimization strategies:

    • Western blot vs. IHC discrepancies:

      • Epitope accessibility may differ in denatured vs. fixed samples

      • Try different fixation methods or antigen retrieval approaches for IHC

      • Consider native vs. denaturing conditions for Western blot

    • IF vs. ChIP discrepancies:

      • Crosslinking may affect epitope recognition differently

      • Optimize fixation time and conditions for each application

      • Consider different antibody clones targeting distinct epitopes

  • Protocol adaptation approaches:

    • Adjust antibody concentration independently for each application

    • Modify incubation times and temperatures based on application requirements

    • Use application-specific blocking reagents to reduce non-specific binding

  • Sample preparation considerations:

    • Pre-treatment options (e.g., heat-induced epitope retrieval, enzymatic digestion)

    • Buffer composition adjustments (detergent types/concentrations, salt concentrations)

    • Fresh vs. frozen vs. fixed material comparisons

  • Antibody selection strategies:

    • Different antibodies may be required for different applications

    • Consider the immunogen type (peptide vs. full-length protein)

    • Polyclonal antibodies may work better for certain applications due to multiple epitope recognition

  • Validation across applications:

    • Implement orthogonal validation techniques

    • Verify results with alternative methods (e.g., RNA expression, mass spectrometry)

    • Document batch-to-batch variations by maintaining reference samples

By systematically addressing each variable, researchers can optimize conditions for specific applications while understanding the limitations of antibody performance across different experimental contexts .

How can POLR1E antibodies contribute to cancer research, particularly in relation to autoimmune conditions?

POLR1E antibodies offer valuable tools for investigating the complex relationship between cancer and autoimmune conditions like scleroderma:

  • Autoantibody profiling and cancer risk assessment:

    • POLR1E antibodies can be used to study how anti-RNA polymerase antibodies in patients correlate with cancer development

    • Research has shown that while anti-RNA pol III antibodies are associated with increased cancer incidence, combinations with other antibodies (e.g., anti-RNA pol III + anti-RPA194) may protect against cancer

    • Implement immunoprecipitation assays to characterize autoantibody profiles in patient cohorts

  • Mechanistic studies of cancer-autoimmunity relationships:

    • Investigate how cancer-associated mutations in RNA polymerase genes may trigger autoimmunity

    • Previous research demonstrated that 15% of scleroderma patients with anti-RNA pol III antibodies have cancer diagnosed concurrent with scleroderma development, often with mutations in polymerase genes

    • Use POLR1E antibodies to track altered Pol I complexes in cancer cells

  • Therapeutic target investigation:

    • RNA polymerase I is a potential therapeutic target in cancer

    • POLR1E antibodies can monitor Pol I inhibitor efficacy in experimental models

    • Implement real-time imaging techniques with POLR1E antibodies for drug screening applications

  • Methodological approaches:

    • Tissue microarray analysis with POLR1E antibodies to examine expression across cancer types

    • ChIP-seq to identify altered binding patterns in cancer vs. normal cells

    • Proximity ligation assays to detect altered protein interactions in disease states

These applications highlight how POLR1E antibodies can bridge basic research on RNA polymerase I biology with clinical investigations into cancer-autoimmunity connections .

What role can POLR1E antibodies play in studying nucleolar stress in disease models?

POLR1E antibodies serve as powerful tools for investigating nucleolar stress in various disease models:

  • Neurodegenerative disease applications:

    • Track nucleolar morphology changes in Alzheimer's, Parkinson's, and related disorders

    • Investigate the relationship between protein aggregation and Pol I activity

    • Pol I inhibition has been proposed as a therapeutic strategy for neurodegenerative conditions

    • Methodological approach: Combine POLR1E immunostaining with markers of protein aggregation in tissue samples

  • Cancer research applications:

    • Monitor nucleolar stress induced by chemotherapeutics

    • Investigate cancer cell adaptation to ribosomal biogenesis stress

    • Study connections between p53 pathways and Pol I activity

    • Approach: Use POLR1E antibodies in conjunction with proliferation markers to assess therapeutic efficacy

  • Aging research:

    • Track age-related changes in nucleolar morphology and function

    • Investigate how restraining Pol I activity might influence aging processes

    • Methodology: Longitudinal studies using POLR1E antibodies to quantify nucleolar changes

  • Live-cell imaging approaches:

    • Implement LiveArt (live imaging-based analysis of rDNA transcription) to visualize real-time Pol I activity

    • This technique reveals dynamic processes including mitotic silencing and reactivation of rDNA transcription

    • Technical approach: Combine with photoactivatable fluorophores for pulse-chase analysis of rRNA synthesis

  • Quantitative assessment strategies:

    • Measure nucleolar size, number, and POLR1E distribution as stress indicators

    • Implement high-content screening approaches for drug discovery

    • Correlate nucleolar stress markers with disease progression

By applying these approaches, researchers can use POLR1E antibodies to gain insights into fundamental disease mechanisms and potential therapeutic interventions targeting nucleolar functions .

How can researchers utilize POLR1E antibodies in multi-omics approaches to study RNA polymerase I biology?

Integrating POLR1E antibodies into multi-omics research frameworks enables comprehensive analysis of RNA polymerase I biology:

  • ChIP-seq integration strategies:

    • Combine POLR1E ChIP-seq with RNA-seq to correlate binding with transcriptional output

    • Integrate with histone modification profiles to understand chromatin environment at active rDNA loci

    • Overlay with DNA methylation data to examine epigenetic regulation of rRNA genes

    • Methodological approach: Sequential ChIP for POLR1E followed by other factors to identify co-occupancy

  • Proteomics applications:

    • Immunoprecipitation with POLR1E antibodies followed by mass spectrometry

    • Identify novel interacting partners under different cellular conditions

    • Quantify post-translational modifications affecting Pol I activity

    • Technical consideration: Cross-linking protocols must be optimized to preserve transient interactions

  • Spatial transcriptomics approaches:

    • Combine POLR1E immunofluorescence with in situ hybridization for nascent rRNA

    • Correlate spatial distribution of Pol I with ribosome biogenesis markers

    • Implementation strategy: Use multiplexed imaging with other nucleolar markers

  • Single-cell multi-omics integration:

    • Single-cell ATAC-seq combined with POLR1E immunofluorescence

    • Link chromatin accessibility with Pol I localization at single-cell resolution

    • Methodology: Index sorting followed by single-cell sequencing technologies

  • Computational integration frameworks:

    • Develop algorithms to integrate multiple data types using POLR1E as a focal point

    • Create predictive models of rRNA synthesis regulation

    • Apply machine learning approaches to identify patterns across diverse datasets

These integrated approaches allow researchers to study RNA polymerase I biology from multiple perspectives simultaneously, providing deeper insights into the complex regulatory mechanisms controlling ribosomal RNA synthesis and nucleolar function .

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.