POLD1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The POLD1 antibody is a rabbit monoclonal antibody (clone EPR15118) specifically designed to target the catalytic subunit of DNA polymerase delta (Polδ), encoded by the POLD1 gene. This enzyme plays a critical role in DNA replication and repair, including lagging strand synthesis, mismatch repair, and translesion synthesis . The antibody is widely used in research to study POLD1’s involvement in genomic stability, cancer progression, and immune evasion mechanisms.

Role in Cancer Progression

POLD1 overexpression has been linked to aggressive tumor phenotypes, including enhanced proliferation, metastasis, and resistance to immunotherapy . Studies employing the POLD1 antibody revealed:

Cancer TypePOLD1 ExpressionClinical Correlation
ccRCCHighPoor OS, advanced stage
Breast/LiverElevatedHigher mutation burden

Mechanistic Insights

  • DNA Damage Repair: POLD1’s exonuclease domain ensures high-fidelity replication. Mutations impairing this domain increase mutation load and cancer risk .

  • Therapeutic Target: POLD1 knockdown inhibits tumor growth and sensitizes cells to ferroptosis, suggesting potential therapeutic applications .

Clinical Relevance and Diagnostic Potential

The POLD1 antibody has been utilized in immunohistochemical analyses of paraffin-embedded tissues to assess tumor aggressiveness. For example:

  • ccRCC Diagnosis: High POLD1 expression in tumor samples predicts resistance to immune-checkpoint inhibitors (ICIs) and correlates with somatic mutations .

  • Biomarker for Immunotherapy: POLD1 proofreading domain mutations may predict clinical responses to ICIs, highlighting its utility in personalized medicine .

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributor for specific delivery times.
Synonyms
Polymerase (DNA directed) delta 1 catalytic subunit antibody; CDC2 antibody; CDC2 homolog antibody; CRCS10 antibody; DNA directed DNA polymerase delta 1 antibody; DNA directed polymerase delta 1 antibody; DNA pol delta 1 antibody; DNA polymerase delta catalytic subunit antibody; DNA polymerase subunit delta p125 antibody; DPOD1_HUMAN antibody; MDPL antibody; POLD antibody; POLD 1 antibody; POLD1 antibody; Polymerase (DNA directed) delta 1 catalytic subunit 125kDa antibody; Polymerase (DNA) delta 1 catalytic subunit antibody; Polymerase DNA directed delta 1 catalytic subunit 125kD antibody; polymerase, DNA, delta antibody
Target Names
Uniprot No.

Target Background

Function
As the catalytic component of the trimeric (Pol-delta3 complex) and tetrameric DNA polymerase delta complexes (Pol-delta4 complex), POLD1 plays a crucial role in high-fidelity genome replication. This includes its involvement in lagging strand synthesis and DNA repair. POLD1 exhibits both DNA polymerase and 3'- to 5'-exonuclease activities. For full activity, it requires the presence of accessory proteins POLD2, POLD3, and POLD4. The presence or absence of POLD4 (Pol-delta3 vs. Pol-delta4) influences catalytic activity. Notably, Pol-delta3 displays higher proofreading activity than Pol-delta4. While both Pol-delta3 and Pol-delta4 process Okazaki fragments in vitro, Pol-delta3 may be better suited for this task, exhibiting near-absence of strand displacement activity compared to Pol-delta4. It also stalls upon encountering 5'-blocking oligonucleotides. The idling process of Pol-delta3 may prevent gap formation while maintaining a nick that can be readily ligated. Along with DNA polymerase kappa, DNA polymerase delta carries out approximately half of nucleotide excision repair (NER) synthesis following UV irradiation. In the presence of POLD3 and POLD4, under conditions of DNA replication stress, POLD1 may catalyze the repair of broken replication forks through break-induced replication (BIR). It is also involved in the translesion synthesis (TLS) of templates carrying O6-methylguanine, 8oxoG, or abasic sites.
Gene References Into Functions
  1. Research suggests that SIRT1 acts as an oncogenic factor in breast cancer cells and may contribute to the progression of breast cancer by inhibiting p53 and activating POLD1. PMID: 29807012
  2. Findings demonstrate a key role of POLD1 and POLD3 in genome stability and S-phase progression. The study revealed RNA-DNA hybrids-dependent effects for POLD3, which might be partly attributed to its interaction with Pol zeta. PMID: 27974823
  3. Mutations in the DNA Polymerase III gene have been linked to MMR deficiency in cancer. PMID: 28512192
  4. POLD1 plays significant roles in regulating cell cycle- and DNA replication-related pathways. E2F can upregulate POLD1 expression levels by deregulating promoter methylation, potentially promoting relapse in Acute Lymphoblastic Leukemia. PMID: 29768346
  5. The catalytic subunit of DNA polymerase delta controls noncentrosomal gammaTuRC activity and regulates the organization of Golgi-derived microtubules. PMID: 28916777
  6. Both POLH and POLK can exchange with PolD1 stalled at repetitive CFS (common fragile sites) sequences. POLD1 synthesis is inhibited by replication stress caused by aphidicolin, preventing replication past CFS. Notably, POLH and POLK remain proficient in rescuing stalled POLD1 synthesis. POLD1 stalling at CFSs allows for free exchange with specialized polymerases, independent of PCNA. PMID: 28605669
  7. To our knowledge, the four Valencian families included in this study represent the only families where the POLD1 Leu474Pro mutation has been identified. PMID: 28306219
  8. An association has been observed between six previously reported single nucleotide polymorphisms (rs15869 [BRCA2], rs1805389 [LIG4], rs8079544 [TP53], rs25489 [XRCC1], rs1673041 [POLD1], and rs11615 [ERCC1]) and subsequent CNS tumors in childhood cancer survivors treated with radiation therapy. PMID: 28976792
  9. The proofreading activity of DNA polymerase delta contributes to shunting DNA mismatch repair towards an EXO1-dependent excision pathway rather than directly participating in gap formation through its 3'-5' exonuclease activity. PMID: 28934474
  10. Frameshift mutations in the POLD1 gene are associated with mismatch repair-deficiency and Lynch syndrome. PMID: 28218421
  11. Research highlights the pathogenic role of the POLD1-R689W mutation in the development of human tumors, emphasizing the need to experimentally determine the significance of Poldelta variants found in sporadic tumors. PMID: 28368425
  12. This work emphasizes that mutations in different POLD1 domains can lead to varying phenotypes, ranging from dominantly inherited cancer predisposition syndromes to milder MDPL phenotypes. PMID: 28521875
  13. Germline or somatic variants in POLE/POLD1 have been identified in unresolved suspected Lynch syndrome cancers with mismatch repair defects. PMID: 26648449
  14. WRN or the Bloom syndrome helicase (BLM) stimulate DNA polymerase delta progression across telomeric G-rich repeats. However, only WRN promotes sequential strand displacement synthesis and FEN1 cleavage. PMID: 27849570
  15. This study investigated complete exonuclease domains of POLE and POLD1 in 529 families characterized by familial or early-onset mismatch repair proficient colorectal cancer, and/or APC-negative and MUTYH-negative polyposis. The results broaden the phenotypic spectrum of the POLE/POLD1-associated syndrome and identify novel pathogenic variants. PMID: 26133394
  16. POLD1 is a central mediator of DNA replication and repair, implicated in cancer and other pathologies. (Review) PMID: 27320729
  17. Inactivating POLD1 mutations are associated with colorectal cancer. PMID: 26755646
  18. Mutations in POLE and POLD1 in Southeast Asian women with grade 3 endometrioid endometrial carcinomas are associated with improved recurrence-free survival. PMID: 26748215
  19. Germline mutations in POLD1 can result in a variably expressed and potentially underdiagnosed segmental progeroid syndrome. PMID: 26172944
  20. On the sequence of Escherichia coli oriC plasmid DNA, it was found that hPold replicates DNA across CCG repeats, while hPole stalls at CCG repeats, even when the secondary structure is eliminated by a single-stranded binding protein. PMID: 26271349
  21. None of the 1188 patients carried the POLD1 p.(Ser478Asn) variant. Carriers of POLE germline variants are also associated with microsatellite instable colorectal cancer. PMID: 25370038
  22. POLD1 plays a crucial role in the regulation of cell cycle progression and DNA damage repair. PMID: 26087769
  23. This study analyzed the phenotype of variants of the essential replicative polymerase-delta carrying missense mutations in its active site. PMID: 25241845
  24. Mutations in POLE and POLD1 explain a portion of familial CRC and polyposis. Sequencing the proofreading domains of POLE and POLD1 should be considered in routine genetic diagnostics. PMID: 24501277
  25. Germline mutations in the proofreading domains of two DNA polymerases (POLE and POLD1) have been linked to a dominantly inherited, highly penetrant syndrome characterized by oligoadenomatous polyposis and early-age-of-diagnosis colorectal and endometrial cancer. PMID: 24509466
  26. POLE and POLD1 mutations are associated with endometrial cancer. PMID: 23528559
  27. A single-codon deletion in POLD1 affecting the polymerase active site causes a distinctly different phenotype that includes lipodystrophy. PMID: 23770608
  28. Germline and somatic polymerase epsilon and delta mutations define a new class of hypermutated colorectal and endometrial cancers. PMID: 23447401
  29. The downregulation of DNA pol delta1 is age-related and has minimal impact on diseases and nutrition. DNA pol delta1 holds potential as a new biomarker for aging. PMID: 22915169
  30. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose individuals to colorectal adenomas and carcinomas. PMID: 23263490
  31. The human lagging strand DNA polymerase delta holoenzyme is distributive. PMID: 22942285
  32. Reducing expression of individual PRMT7 target DNA repair genes showed that only the catalytic subunit of DNA polymerase, POLD1, could resensitize PRMT7 knock-down cells to DNA-damaging agents. PMID: 22761421
  33. Pol switches at replication-blocking lesions occur through the exchange of the Pol delta and Pol zeta catalytic subunits on a preassembled complex of accessory proteins retained on DNA during translesion DNA synthesis. PMID: 22465957
  34. Coronavirus nsp13 and DNA polymerase delta induced DNA replication stress in IBV-infected cells. PMID: 21918226
  35. POLD1 gene mutations are not responsible for the elevated HPRT mutation rates in a colon cancer cell line. PMID: 14767555
  36. Ubiquitin and ubiquitin-like proteins modify the p66 and p12 subunits of DNA polymerase delta. PMID: 16934752
  37. Results suggest that the CDE/CHR-like sequence is an active functional element in the POLD1 promoter, which plays a role in cell cycle regulation of the POLD1 gene. PMID: 19557333

Show More

Hide All

Database Links

HGNC: 9175

OMIM: 174761

KEGG: hsa:5424

STRING: 9606.ENSP00000406046

UniGene: Hs.279413

Involvement In Disease
Colorectal cancer 10 (CRCS10); Mandibular hypoplasia, deafness, progeroid features, and lipodystrophy syndrome (MDPL)
Protein Families
DNA polymerase type-B family
Subcellular Location
Nucleus.
Tissue Specificity
Widely expressed, with high levels of expression in heart and lung.

Q&A

What is POLD1 and why is it significant in cancer research?

POLD1 (polymerase DNA directed, delta 1, catalytic subunit) is a 1107 amino acid protein with a molecular weight of 124 kDa that belongs to the DNA polymerase type-B family. It serves as the catalytic component of the DNA polymerase delta complexes, playing a crucial role in high-fidelity genome replication, particularly in lagging strand synthesis and DNA repair . POLD1 exhibits both DNA polymerase and 3'-5'-exonuclease activities and requires accessory proteins POLD2, POLD3, and POLD4 for full activity .

POLD1 has emerged as significant in cancer research due to:

  • Its frequent overexpression in various tumors including clear cell renal cell carcinoma (ccRCC), colorectal cancer, breast cancer, and bladder cancer

  • Its correlation with pathologic tumor stage and histologic grade in multiple cancers

  • Its potential as a prognostic biomarker, with high expression often associated with poor clinical outcomes

  • The role of POLD1 mutations in hypermutation phenotypes that influence immunotherapy response

What are the common experimental applications for POLD1 antibodies?

POLD1 antibodies have been validated and utilized in multiple experimental techniques:

ApplicationValidated DilutionsCommon Cell/Tissue Types
Western Blot (WB)1:500-1:3000Jurkat, K-562, HeLa, MOLT4, C6, Raw264.7, PC12 cells
Immunohistochemistry (IHC)1:50-1:500Human breast cancer tissue, colon cancer tissue, cervix carcinoma, ovarian carcinoma
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg proteinHeLa cells
Immunocytochemistry/Immunofluorescence (ICC/IF)1:250-1:500HeLa, K562 cells
Flow Cytometry1:250HeLa cells
ELISA1:40000Various human samples

For optimal results, antibody concentration should be titrated for each specific experimental system .

How should POLD1 antibodies be stored and handled for maximum stability?

Based on manufacturer recommendations across multiple antibody products:

Note that some preparations (particularly 20μl sizes) may contain 0.1% BSA, which should be considered when designing experiments .

How can I validate the specificity of a POLD1 antibody for my particular experimental system?

Comprehensive validation of POLD1 antibody specificity requires a multi-faceted approach:

  • Positive and negative control selection:

    • Validated positive controls for Western blot include Jurkat, K-562, HeLa, and MOLT4 cells which consistently show a band at 124 kDa .

    • For immunohistochemistry, human breast cancer, colon cancer, and cervix carcinoma tissues serve as reliable positive controls .

    • Consider using POLD1 knockdown or knockout cell lines as negative controls to confirm antibody specificity.

  • Cross-reactivity assessment:

    • While many POLD1 antibodies are validated for human samples, some also react with mouse and rat samples . Confirm cross-reactivity through literature or manufacturer data before using in non-human systems.

    • Sequence alignment analysis between human, mouse, and rat POLD1 can predict potential cross-reactivity based on epitope conservation.

  • Blocking peptide verification:

    • When available, use the immunogen peptide as a competitive inhibitor in parallel experiments to confirm binding specificity .

    • A true signal should be significantly reduced or eliminated when pre-incubated with the blocking peptide.

  • Multiple detection methods concordance:

    • Verify POLD1 detection across multiple techniques (e.g., WB, IHC, IF) to ensure consistent localization and expression patterns.

    • Nuclear localization is expected in positive samples based on POLD1's function .

  • Molecular weight verification:

    • POLD1 should appear at 124 kDa in denaturing conditions, consistent with its calculated molecular weight .

    • Any additional bands should be carefully investigated for potential isoforms or degradation products.

What are the optimal antigen retrieval conditions for POLD1 detection in FFPE tissues?

Optimal antigen retrieval for POLD1 in formalin-fixed paraffin-embedded (FFPE) tissues depends on the specific antibody and tissue type. Based on published protocols:

Primary recommendation:

  • Heat-mediated antigen retrieval with EDTA buffer at pH 9.0 .

  • This method has been validated for human breast cancer, colon cancer, and cervix carcinoma tissues.

Alternative method:

  • Citrate buffer at pH 6.0 can also be effective for some tissue types .

  • This approach may be considered if EDTA buffer retrieval yields suboptimal results.

Protocol optimization considerations:

  • Retrieval duration: Typically 10-20 minutes at 95-100°C, with exact timing requiring optimization for each tissue type.

  • Cooling period: Allow slides to cool to room temperature gradually (15-20 minutes) before proceeding with blocking steps.

  • Section thickness: 4-5 μm sections generally provide optimal results for POLD1 detection.

  • Background reduction: After antigen retrieval, thorough washing and effective blocking are crucial. Consider 3-5% BSA or 5-10% normal serum from the same species as the secondary antibody.

For reliable results, each new tissue type should undergo a systematic optimization of these parameters.

How can I accurately quantify POLD1 expression in tumor samples for correlation with clinical outcomes?

Accurate quantification of POLD1 expression for clinical correlations requires standardized approaches:

For IHC-based quantification:

  • Scoring system development:

    • Implement a combined intensity and percentage scoring system:

      • Intensity scale: 0 (negative), 1 (weak), 2 (moderate), 3 (strong)

      • Percentage scale: 0-100% of positive cells

    • Calculate H-score (0-300) by multiplying intensity (0-3) by percentage (0-100%)

    • Alternatively, use Allred scoring (intensity + proportion, 0-8)

  • Cut-off determination:

    • Define high vs. low expression categories using:

      • Median H-score from your cohort (as used in ccRCC studies)

      • ROC curve analysis for outcome prediction

      • Statistical methods like minimum P-value approach

  • Controls and normalization:

    • Include positive and negative controls on each slide

    • Use automated image analysis software when possible for objective quantification

    • Consider tissue microarrays (TMAs) for high-throughput analysis

For Western blot quantification:

  • Use housekeeping proteins (β-actin, GAPDH) for normalization

  • Include standard curves with recombinant POLD1 protein

  • Employ densitometry software for band intensity measurement

  • Run at least three technical replicates

For RT-qPCR quantification:

  • Select appropriate reference genes verified for stability in your tissue type

  • Use the 2^-ΔΔCt method for relative quantification

  • Include no-template and no-RT controls

  • Validate primers for specificity and efficiency (90-110%)

How do POLD1 mutations affect response to immunotherapy across different cancer types?

POLD1 mutations have emerged as promising predictive biomarkers for immunotherapy response across multiple cancer types:

Mechanism of immunotherapy sensitization:
POLD1 mutations, particularly in the exonuclease domain, impair proofreading function during DNA replication, leading to:

  • Accumulation of somatic mutations and ultra-high mutation load

  • Increased neoantigen production that can enhance tumor immunogenicity

  • Altered tumor microenvironment with increased immune cell infiltration

Clinical evidence across cancer types:
A cohort study analyzing 47,721 patients with various cancers found that:

  • POLD1 mutations were frequently observed in endometrial, colorectal, skin, esophagogastric, bladder, and lung cancers

  • POLD1 mutations served as a negative prognostic marker in untreated patients but predicted survival benefit from immune checkpoint inhibitor (ICI) therapy

  • Mutations in all exons, not just the exonuclease domain, were associated with improved outcomes on ICI therapy

Relationship with established biomarkers:

  • POLD1 mutations often correlate with high tumor mutation burden (TMB) but represent a distinct predictive biomarker

  • The predictive value appears independent of microsatellite instability (MSI) status, suggesting utility even in non-MSI-high tumors

Ongoing clinical investigation:

  • A phase 2 clinical trial has been initiated to test the treatment outcomes of toripalimab (PD-1 antibody) in patients with solid cancers harboring POLD1 mutations who are non-MSI-high

These findings suggest that POLD1 mutation testing could help identify additional patients likely to benefit from immunotherapy beyond established biomarkers like PD-L1 expression and MSI status.

What experimental approaches can differentiate functional from non-functional POLD1 mutations?

Distinguishing functional from non-functional POLD1 mutations requires multiple experimental approaches:

Computational prediction methods:
Different pathogenicity prediction tools yield variable results for POLD1 variants:

POLD1 VariantPON-P2PolyPhen-2PROVEANMutationAssessor
G10VNeutralBenignNeutralLow impact
R506HPathogenicBenignDeleteriousMedium impact
R689WPathogenicProbably damagingDeleteriousHigh impact
S746INeutralBenignNeutralLow impact

These computational predictions should be verified through functional assays .

CRISPR/Cas9 variant modeling:

  • Generate isogenic cell lines expressing specific POLD1 variants using CRISPR/Cas9

  • This approach has been used to study variants like R689W in colorectal cancer cell lines

Functional assays to assess variant impact:

  • DNA replication fidelity assessment:

    • Measure mutation rates using reporter assays

    • Analyze microsatellite stability in variant-expressing cells

  • DNA damage response analysis:

    • Monitor CHK1 phosphorylation levels

    • Assess γH2AX foci formation as indicator of DNA damage

  • Cell cycle and apoptosis evaluation:

    • Flow cytometry for cell cycle distribution

    • Apoptosis assays (Annexin V/PI staining)

  • Drug sensitivity testing:

    • Measure sensitivity to ATR inhibitors

    • Evaluate response to other DNA-damaging agents

  • Mutational signature analysis:

    • Functional POLD1 mutations generate specific mutational signatures

    • Logistic regression models based on COSMIC SBS signatures can predict functional mutations

Research has shown that the R689W variant specifically increases sensitivity to ATR inhibitors in colorectal cancer cells, demonstrating how functional analysis can identify therapeutic vulnerabilities associated with specific POLD1 variants .

How does POLD1 expression influence the tumor microenvironment and immune cell infiltration?

POLD1 expression has significant effects on the tumor immune microenvironment, with high expression generally associated with immunosuppressive features:

Immune cell infiltration patterns:
High POLD1 expression correlates with specific immune cell infiltration profiles:

  • Increased infiltration of:

    • CD56dim natural killer cells

    • Regulatory T cells (Tregs)

    • Myeloid-derived suppressor cells (MDSCs)

    • Activated CD8 T cells

  • Decreased infiltration of:

    • Immature dendritic cells

    • Natural killer cells

    • Neutrophils

Association with T cell exhaustion markers:
POLD1 expression positively correlates with T cell exhaustion markers, suggesting a role in immune escape:

  • Significant correlation with CTLA4, LAG3, LGALS9, TGFB1, and PDCD1 (PD-1)

  • Strong association with markers of Tregs and T cell exhaustion

Immunomodulator correlations:
POLD1 levels show significant associations with both immunoinhibitors and immunostimulators:

  • Immunoinhibitors: CTLA4, LAG3, LGALS9, TGFB1, and PD-1

  • Immunostimulators: TNFRSF18, TNFRSF25, TNFRSF8, TNFRSF14, and LTA

  • Chemokines: CCL5, CXCL13, XCL1, and XCL2

  • Chemokine receptors: CXCR3, CXCR5, and CXCR6

Experimental validation approaches:
Researchers have utilized multiple databases and experimental methods to establish these correlations:

  • TIMER and TISIDB databases for immune cell infiltration analysis

  • RT-qPCR, Western blot, and immunohistochemistry for validation

  • Functional and animal experiments for in vitro and in vivo verification

These findings suggest that POLD1 may influence tumor progression partly by creating an immunosuppressive microenvironment, which could have important implications for immunotherapy approaches.

What are the optimal experimental designs for studying POLD1's role in cancer cell proliferation and metastasis?

Based on published studies, a comprehensive experimental design to investigate POLD1's role in cancer should include:

In vitro experimental approaches:

  • POLD1 expression modulation:

    • Knockdown: siRNA or shRNA targeting POLD1 (at least two siRNA sequences for validation)

    • Overexpression: POLD1-expressing plasmids

    • Validation: RT-qPCR and Western blot to confirm efficiency

  • Proliferation assays:

    • MTT/CCK-8 assay at multiple time points (24h, 48h, 72h, 96h)

    • Colony formation assay (14-21 days)

    • Cell cycle analysis by flow cytometry with PI staining

    • EdU incorporation assay for DNA synthesis

  • Migration and invasion assays:

    • Transwell migration assay (24-48 hours)

    • Wound healing assay with time-lapse imaging

    • 3D invasion assays with Matrigel coating

  • Mechanistic investigations:

    • RNA-seq after POLD1 knockdown to identify altered pathways

    • GSEA and GO analysis for functional annotation

    • Immunoblotting for cell cycle proteins (Cyclin E1, Cyclin D1) and EMT markers (E-cadherin, N-cadherin, Vimentin, Snail)

    • Immunofluorescence staining for proliferation markers (Ki67) and EMT proteins

In vivo experimental approaches:

  • Tumor growth models:

    • Subcutaneous injection of POLD1-knockdown or overexpressing cells in immunodeficient mice

    • Tumor size and weight measurements over time

    • IHC staining of tumor tissues for POLD1 and Ki67

  • Metastasis models:

    • Tail vein injection to assess lung metastasis potential

    • Quantification of metastatic foci number and size

    • Optional: orthotopic models for tissue-specific assessment

  • Rescue experiments:

    • Co-expression of downstream targets (e.g., MYC) with POLD1 knockdown to verify mechanism

    • In vivo and in vitro validation of rescue effect

  • Drug sensitivity studies:

    • Treatment with targeted therapies (e.g., ATR inhibitors) in POLD1-modulated cells

    • Combination therapy approaches based on pathway analysis

Studies implementing these approaches have revealed that POLD1 promotes cancer cell proliferation by facilitating G1-S phase transition and enhances metastasis through EMT activation, with potential mechanistic involvement of MYC stabilization .

What controls and validation steps are crucial when using POLD1 antibodies in different experimental techniques?

Proper controls and validation are essential for generating reliable data with POLD1 antibodies:

For Western Blotting:

  • Loading controls:

    • Use appropriate housekeeping proteins (β-actin, GAPDH, α-tubulin)

    • Consider nuclear loading controls (Lamin B1, Histone H3) as POLD1 is predominantly nuclear

  • Specificity controls:

    • Positive controls: Known POLD1-expressing cell lines (Jurkat, K-562, HeLa, MOLT4)

    • Negative controls: POLD1 knockdown/knockout cells

    • Peptide competition assay with immunizing peptide

  • Molecular weight verification:

    • POLD1 should appear at 124 kDa

    • Pre-stained protein ladder to confirm size

  • Additional validation:

    • Multiple antibodies targeting different epitopes

    • Gradient dilution series to determine optimal concentration (1:500-1:3000)

For Immunohistochemistry:

  • Tissue controls:

    • Positive tissue controls: Human breast cancer, colon cancer tissue

    • Negative tissue controls: Tissues known to lack POLD1 expression

    • Isotype control: Normal IgG from same species as primary antibody

  • Antibody controls:

    • Omission of primary antibody

    • Serial antibody dilutions (1:50-1:500) to optimize signal-to-noise ratio

    • Peptide competition control

  • Antigen retrieval optimization:

    • Compare EDTA buffer (pH 9.0) vs. citrate buffer (pH 6.0)

    • Test multiple retrieval durations

  • Staining pattern verification:

    • Nuclear localization expected for POLD1

    • Comparison with RNA-seq or other expression data

For Immunofluorescence:

  • Fixation optimization:

    • Compare 4% paraformaldehyde vs. acetone fixation

    • Test different permeabilization methods (0.1% Triton X-100)

  • Fluorescence controls:

    • Secondary antibody only control

    • Autofluorescence control (unstained sample)

    • Nuclear counterstain (DAPI) for co-localization

  • Dilution optimization:

    • Test range of dilutions (1:250-1:500)

    • Signal intensity quantification

For Immunoprecipitation:

  • IP controls:

    • IgG control from same species as POLD1 antibody

    • Input sample (pre-IP lysate)

    • Optimized antibody amount (0.5-4.0 μg for 1.0-3.0 mg protein)

  • Interaction validation:

    • Reverse IP with interacting protein antibodies

    • IP under different conditions (± DNA damage)

Implementing these controls ensures reliable and reproducible results across different experimental techniques and research questions involving POLD1.

How should researchers integrate POLD1 expression data with other tumor characteristics for comprehensive cancer studies?

Comprehensive integration of POLD1 expression data with other tumor characteristics requires a multidimensional approach:

Data integration framework:

  • Multi-omics data correlation:

    • Genomics: Correlate POLD1 expression with mutation status, copy number variations

    • Transcriptomics: Identify co-expressed genes and pathways through RNA-seq

    • Proteomics: Analyze protein interaction networks involving POLD1

    • Epigenomics: Investigate methylation patterns of POLD1 promoter

  • Clinical data integration:

    • Patient demographics (age, sex, ethnicity)

    • Tumor characteristics (stage, grade, histological subtype)

    • Treatment history and response

    • Survival outcomes (OS, PFS, DSS)

  • Immune landscape correlation:

    • Immune cell infiltration profiles (quantified by deconvolution algorithms)

    • Expression of immune checkpoint molecules

    • Cytokine/chemokine expression patterns

    • Tumor mutational burden and neoantigen load

Analytical approaches:

  • Stratification strategies:

    • Divide patients into high vs. low POLD1 expression groups based on median expression

    • Create patient clusters based on POLD1 and related pathway genes

    • Use machine learning for patient classification

  • Statistical methods:

    • Cox regression for survival analysis (univariate and multivariate)

    • ANOVA or t-tests for group comparisons

    • Correlation coefficients (Pearson, Spearman) for continuous variables

    • Multiple testing correction (FDR, Bonferroni)

  • Pathway and network analysis:

    • Gene Set Enrichment Analysis (GSEA) for biological pathways

    • Protein-protein interaction networks

    • Regulatory network inference

  • Visualization techniques:

    • Heatmaps for expression patterns

    • Kaplan-Meier curves for survival analysis

    • Forest plots for multivariate analysis

    • t-SNE or UMAP for dimension reduction

Validation strategies:

  • Cross-validation in independent cohorts:

    • Use multiple patient datasets (e.g., TCGA, GEO)

    • Split discovery and validation cohorts

  • Experimental validation:

    • In vitro confirmation of key findings

    • Patient-derived xenograft models

    • Prospective clinical validation

Studies implementing these approaches have revealed that POLD1 expression is associated with pathologic tumor stage, histologic grade, immune cell infiltration patterns, and patient survival across multiple cancer types . For example, ccRCC patients with high POLD1 expression show poorer OS, PFS, and DSS, along with specific immune infiltration profiles characterized by increased Treg cells and MDSCs .

How can researchers troubleshoot inconsistent POLD1 antibody staining patterns in immunohistochemistry?

Inconsistent POLD1 antibody staining in IHC can be systematically resolved through the following troubleshooting approach:

Problem: Weak or absent staining

Potential CauseSolution
Insufficient antigen retrievalTry EDTA buffer pH 9.0 as primary method; increase retrieval time or temperature
Antibody concentration too lowTitrate antibody concentration (start with 1:50 for weak signals; recommended range 1:50-1:500)
Tissue fixation issuesOptimize fixation time; consider using tissue fixed for 24-48 hours in 10% neutral buffered formalin
Antibody degradationUse fresh antibody aliquot; check storage conditions (-20°C, with glycerol)
Detection system sensitivitySwitch to more sensitive detection system (polymer-HRP or tyramide signal amplification)

Problem: High background staining

Potential CauseSolution
Antibody concentration too highDilute antibody further (try 1:500 for high background)
Insufficient blockingExtend blocking time; try different blocking reagents (5-10% normal serum, 3-5% BSA)
Cross-reactivityUse more specific antibody; confirm with peptide competition assay
Endogenous peroxidase activityEnhance peroxidase blocking (3% H₂O₂ for 10-15 minutes)
Non-specific bindingInclude 0.1-0.3% Triton X-100 or Tween-20 in wash buffers

Problem: Inconsistent staining across samples

Potential CauseSolution
Tissue processing variationsStandardize fixation and processing protocols; use tissue microarrays for batch consistency
Antigen degradationMinimize time between sectioning and staining; store unstained slides at 4°C
Antibody batch variationUse the same lot number for entire study; include standard control slide in each batch
Protocol inconsistenciesUse automated staining platforms; detailed protocol documentation
Regional tissue variationsTake multiple cores per sample; analyze larger tissue areas

Problem: Unexpected staining pattern

Potential CauseSolution
Non-specific antibodyValidate with alternative antibodies targeting different epitopes
Post-translational modificationsConsider phospho-specific antibodies if relevant
Isoform expressionVerify epitope region against known isoforms
Technical artifactsInclude negative controls (isotype IgG, no primary antibody)
Aberrant POLD1 localizationConfirm with immunofluorescence co-localization studies

Recommended validation workflow:

  • Start with recommended protocol (antigen retrieval with EDTA buffer pH 9.0, 1:100 antibody dilution)

  • Systematically optimize each variable independently

  • Include positive control tissues (human breast cancer, colon cancer)

  • Compare multiple POLD1 antibodies when possible

  • Validate findings with orthogonal methods (WB, IF)

Following these troubleshooting steps will help ensure consistent and reliable POLD1 IHC staining across experimental samples.

What factors should be considered when comparing results from different POLD1 antibodies across studies?

When comparing results from different POLD1 antibodies across studies, researchers should consider several critical factors:

Antibody characteristics comparison:

FactorPotential ImpactAssessment Method
Epitope locationDifferent domains detect different isoforms or truncated proteinsCompare immunogen sequences in product datasheets
ClonalityMonoclonals (higher specificity); Polyclonals (better signal, multiple epitopes)Check antibody type: monoclonal vs. polyclonal
Host speciesAffects secondary antibody selection and background in certain tissuesCompare host species (rabbit most common for POLD1)
Validation extentMore extensively validated antibodies provide higher confidenceReview validation data and publication history
Cross-reactivityDifferent species reactivity profilesCheck tested reactivity (human, mouse, rat)

Methodological considerations:

  • Protocol differences:

    • Antigen retrieval methods (EDTA pH 9.0 vs. citrate pH 6.0)

    • Antibody incubation conditions (time, temperature, concentration)

    • Detection systems (chromogenic vs. fluorescent; amplification methods)

    • Blocking reagents and washing protocols

  • Sample preparation variations:

    • Fixation protocols and duration

    • Processing and embedding techniques

    • Storage conditions and section thickness

    • Fresh vs. archival tissue samples

  • Quantification methods:

    • Scoring systems (H-score, Allred, percentage positive)

    • Manual vs. automated analysis

    • Different thresholds for positive/negative classification

    • Image acquisition parameters

Reconciliation strategies:

  • Direct comparison experiments:

    • Test multiple antibodies on the same sample set

    • Create concordance tables between antibodies

    • Determine conversion factors if possible

  • Validation with orthogonal methods:

    • Correlate IHC with mRNA expression data

    • Confirm with Western blot analysis

    • Verify with functional assays

  • Meta-analysis approaches:

    • Standardize effect sizes rather than absolute measurements

    • Subgroup analysis by antibody type

    • Sensitivity analysis excluding outlier studies

  • Reporting standards implementation:

    • Detailed MIQE guidelines for PCR studies

    • Complete antibody reporting (catalog #, lot #, dilution, protocol)

    • Raw data sharing when possible

When examining literature, researchers should carefully evaluate reported antibody details – for instance, multiple studies have used rabbit polyclonal antibodies (15646-1-AP) , while others employed rabbit monoclonal antibodies (EPR15118) or alternative polyclonal antibodies (ab168827) , each with potentially different detection characteristics.

How can researchers accurately distinguish between wild-type POLD1 and mutant variants in experimental systems?

Accurately distinguishing between wild-type POLD1 and mutant variants requires a targeted experimental approach combining molecular, biochemical, and functional techniques:

Genomic and transcript-level detection:

  • Allele-specific PCR:

    • Design primers that selectively amplify wild-type or mutant POLD1 sequences

    • Include positive controls with known genotypes

    • Optimize annealing temperatures for maximum specificity

  • Sanger sequencing:

    • Direct sequencing of POLD1 exons, particularly exonuclease domains

    • Analysis of chromatograms for heterozygous mutations

    • Cloning and sequencing of individual alleles when necessary

  • Next-generation sequencing:

    • Targeted deep sequencing for low-frequency variants

    • RNA-seq to determine allele-specific expression

    • Confirmation of variant allele frequencies

  • Droplet digital PCR:

    • Absolute quantification of wild-type and mutant alleles

    • Detection of low-frequency variants (<1%)

    • Determination of copy number variations

Protein-level detection:

  • Variant-specific antibodies:

    • Develop antibodies recognizing specific mutations (e.g., R689W)

    • Validate specificity with recombinant proteins

    • Use in Western blot or IHC applications

  • Mass spectrometry:

    • Targeted proteomics to detect variant-specific peptides

    • Label-free quantification of wild-type vs. mutant proteins

    • Phosphoproteomics to detect differential post-translational modifications

  • Immunoprecipitation followed by sequencing:

    • Pull down POLD1 protein and sequence associated DNA

    • Analyze error rates and mutation patterns

Cellular model systems:

  • CRISPR/Cas9 engineered cell lines:

    • Generate isogenic cell lines with specific POLD1 variants

    • Create heterozygous and homozygous mutant models

    • Example: DLD-1 colorectal cancer cells with R689W variant

  • Allele-specific knockout:

    • Selectively target wild-type or mutant alleles

    • Validate by allele-specific expression analysis

    • Example: Heterozygous cell clones expressing only R689W variant

Functional discrimination methods:

  • Polymerase activity assays:

    • In vitro DNA synthesis with purified proteins

    • Measurement of processivity and fidelity

    • Analysis of error rates and types

  • DNA damage response analysis:

    • CHK1 phosphorylation status by Western blot

    • γH2AX foci formation by immunofluorescence

    • Cell cycle checkpoint activation

  • Drug sensitivity profiling:

    • Differential sensitivity to ATR inhibitors

    • Synthetic lethality screening

    • POLD1-R689W variant shows increased sensitivity to ATR inhibitors

  • Mutational signature analysis:

    • Whole-genome sequencing to detect characteristic mutation patterns

    • Application of computational models to predict functional mutations

    • Correlation with COSMIC mutational signatures

Studies have successfully implemented these approaches to distinguish between wild-type POLD1 and variants such as G10V, R506H, R689W, and S746I in colorectal cancer models, revealing functional differences particularly in DNA damage response and drug sensitivity .

Comprehensive POLD1 Antibody Research FAQs for Advanced Scientific Investigations

Based on comprehensive analysis of research data, this document provides authoritative answers to frequently asked questions about POLD1 antibody in scientific research. The information is structured to support both foundational understanding and advanced experimental applications.

What is POLD1 and why is it significant in cancer research?

POLD1 (polymerase DNA directed, delta 1, catalytic subunit) is a 1107 amino acid protein with a molecular weight of 124 kDa that belongs to the DNA polymerase type-B family. It serves as the catalytic component of the DNA polymerase delta complexes, playing a crucial role in high-fidelity genome replication, particularly in lagging strand synthesis and DNA repair . POLD1 exhibits both DNA polymerase and 3'-5'-exonuclease activities and requires accessory proteins POLD2, POLD3, and POLD4 for full activity .

POLD1 has emerged as significant in cancer research due to:

  • Its frequent overexpression in various tumors including clear cell renal cell carcinoma (ccRCC), colorectal cancer, breast cancer, and bladder cancer

  • Its correlation with pathologic tumor stage and histologic grade in multiple cancers

  • Its potential as a prognostic biomarker, with high expression often associated with poor clinical outcomes

  • The role of POLD1 mutations in hypermutation phenotypes that influence immunotherapy response

What are the common experimental applications for POLD1 antibodies?

POLD1 antibodies have been validated and utilized in multiple experimental techniques:

ApplicationValidated DilutionsCommon Cell/Tissue Types
Western Blot (WB)1:500-1:3000Jurkat, K-562, HeLa, MOLT4, C6, Raw264.7, PC12 cells
Immunohistochemistry (IHC)1:50-1:500Human breast cancer tissue, colon cancer tissue, cervix carcinoma, ovarian carcinoma
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg proteinHeLa cells
Immunocytochemistry/Immunofluorescence (ICC/IF)1:250-1:500HeLa, K562 cells
Flow Cytometry1:250HeLa cells
ELISA1:40000Various human samples

For optimal results, antibody concentration should be titrated for each specific experimental system .

How should POLD1 antibodies be stored and handled for maximum stability?

Based on manufacturer recommendations across multiple antibody products:

Note that some preparations (particularly 20μl sizes) may contain 0.1% BSA, which should be considered when designing experiments .

How can I validate the specificity of a POLD1 antibody for my particular experimental system?

Comprehensive validation of POLD1 antibody specificity requires a multi-faceted approach:

  • Positive and negative control selection:

    • Validated positive controls for Western blot include Jurkat, K-562, HeLa, and MOLT4 cells which consistently show a band at 124 kDa .

    • For immunohistochemistry, human breast cancer, colon cancer, and cervix carcinoma tissues serve as reliable positive controls .

    • Consider using POLD1 knockdown or knockout cell lines as negative controls to confirm antibody specificity.

  • Cross-reactivity assessment:

    • While many POLD1 antibodies are validated for human samples, some also react with mouse and rat samples . Confirm cross-reactivity through literature or manufacturer data before using in non-human systems.

    • Sequence alignment analysis between human, mouse, and rat POLD1 can predict potential cross-reactivity based on epitope conservation.

  • Blocking peptide verification:

    • When available, use the immunogen peptide as a competitive inhibitor in parallel experiments to confirm binding specificity .

    • A true signal should be significantly reduced or eliminated when pre-incubated with the blocking peptide.

  • Multiple detection methods concordance:

    • Verify POLD1 detection across multiple techniques (e.g., WB, IHC, IF) to ensure consistent localization and expression patterns.

    • Nuclear localization is expected in positive samples based on POLD1's function .

  • Molecular weight verification:

    • POLD1 should appear at 124 kDa in denaturing conditions, consistent with its calculated molecular weight .

    • Any additional bands should be carefully investigated for potential isoforms or degradation products.

What are the optimal antigen retrieval conditions for POLD1 detection in FFPE tissues?

Optimal antigen retrieval for POLD1 in formalin-fixed paraffin-embedded (FFPE) tissues depends on the specific antibody and tissue type. Based on published protocols:

Primary recommendation:

  • Heat-mediated antigen retrieval with EDTA buffer at pH 9.0 .

  • This method has been validated for human breast cancer, colon cancer, and cervix carcinoma tissues.

Alternative method:

  • Citrate buffer at pH 6.0 can also be effective for some tissue types .

  • This approach may be considered if EDTA buffer retrieval yields suboptimal results.

Protocol optimization considerations:

  • Retrieval duration: Typically 10-20 minutes at 95-100°C, with exact timing requiring optimization for each tissue type.

  • Cooling period: Allow slides to cool to room temperature gradually (15-20 minutes) before proceeding with blocking steps.

  • Section thickness: 4-5 μm sections generally provide optimal results for POLD1 detection.

  • Background reduction: After antigen retrieval, thorough washing and effective blocking are crucial. Consider 3-5% BSA or 5-10% normal serum from the same species as the secondary antibody.

For reliable results, each new tissue type should undergo a systematic optimization of these parameters.

How can I accurately quantify POLD1 expression in tumor samples for correlation with clinical outcomes?

Accurate quantification of POLD1 expression for clinical correlations requires standardized approaches:

For IHC-based quantification:

  • Scoring system development:

    • Implement a combined intensity and percentage scoring system:

      • Intensity scale: 0 (negative), 1 (weak), 2 (moderate), 3 (strong)

      • Percentage scale: 0-100% of positive cells

    • Calculate H-score (0-300) by multiplying intensity (0-3) by percentage (0-100%)

    • Alternatively, use Allred scoring (intensity + proportion, 0-8)

  • Cut-off determination:

    • Define high vs. low expression categories using:

      • Median H-score from your cohort (as used in ccRCC studies)

      • ROC curve analysis for outcome prediction

      • Statistical methods like minimum P-value approach

  • Controls and normalization:

    • Include positive and negative controls on each slide

    • Use automated image analysis software when possible for objective quantification

    • Consider tissue microarrays (TMAs) for high-throughput analysis

For Western blot quantification:

  • Use housekeeping proteins (β-actin, GAPDH) for normalization

  • Include standard curves with recombinant POLD1 protein

  • Employ densitometry software for band intensity measurement

  • Run at least three technical replicates

For RT-qPCR quantification:

  • Select appropriate reference genes verified for stability in your tissue type

  • Use the 2^-ΔΔCt method for relative quantification

  • Include no-template and no-RT controls

  • Validate primers for specificity and efficiency (90-110%)

How do POLD1 mutations affect response to immunotherapy across different cancer types?

POLD1 mutations have emerged as promising predictive biomarkers for immunotherapy response across multiple cancer types:

Mechanism of immunotherapy sensitization:
POLD1 mutations, particularly in the exonuclease domain, impair proofreading function during DNA replication, leading to:

  • Accumulation of somatic mutations and ultra-high mutation load

  • Increased neoantigen production that can enhance tumor immunogenicity

  • Altered tumor microenvironment with increased immune cell infiltration

Clinical evidence across cancer types:
A cohort study analyzing 47,721 patients with various cancers found that:

  • POLD1 mutations were frequently observed in endometrial, colorectal, skin, esophagogastric, bladder, and lung cancers

  • POLD1 mutations served as a negative prognostic marker in untreated patients but predicted survival benefit from immune checkpoint inhibitor (ICI) therapy

  • Mutations in all exons, not just the exonuclease domain, were associated with improved outcomes on ICI therapy

Relationship with established biomarkers:

  • POLD1 mutations often correlate with high tumor mutation burden (TMB) but represent a distinct predictive biomarker

  • The predictive value appears independent of microsatellite instability (MSI) status, suggesting utility even in non-MSI-high tumors

Ongoing clinical investigation:

  • A phase 2 clinical trial has been initiated to test the treatment outcomes of toripalimab (PD-1 antibody) in patients with solid cancers harboring POLD1 mutations who are non-MSI-high

These findings suggest that POLD1 mutation testing could help identify additional patients likely to benefit from immunotherapy beyond established biomarkers like PD-L1 expression and MSI status.

What experimental approaches can differentiate functional from non-functional POLD1 mutations?

Distinguishing functional from non-functional POLD1 mutations requires multiple experimental approaches:

Computational prediction methods:
Different pathogenicity prediction tools yield variable results for POLD1 variants:

POLD1 VariantPON-P2PolyPhen-2PROVEANMutationAssessor
G10VNeutralBenignNeutralLow impact
R506HPathogenicBenignDeleteriousMedium impact
R689WPathogenicProbably damagingDeleteriousHigh impact
S746INeutralBenignNeutralLow impact

These computational predictions should be verified through functional assays .

CRISPR/Cas9 variant modeling:

  • Generate isogenic cell lines expressing specific POLD1 variants using CRISPR/Cas9

  • This approach has been used to study variants like R689W in colorectal cancer cell lines

Functional assays to assess variant impact:

  • DNA replication fidelity assessment:

    • Measure mutation rates using reporter assays

    • Analyze microsatellite stability in variant-expressing cells

  • DNA damage response analysis:

    • Monitor CHK1 phosphorylation levels

    • Assess γH2AX foci formation as indicator of DNA damage

  • Cell cycle and apoptosis evaluation:

    • Flow cytometry for cell cycle distribution

    • Apoptosis assays (Annexin V/PI staining)

  • Drug sensitivity testing:

    • Measure sensitivity to ATR inhibitors

    • Evaluate response to other DNA-damaging agents

  • Mutational signature analysis:

    • Functional POLD1 mutations generate specific mutational signatures

    • Logistic regression models based on COSMIC SBS signatures can predict functional mutations

Research has shown that the R689W variant specifically increases sensitivity to ATR inhibitors in colorectal cancer cells, demonstrating how functional analysis can identify therapeutic vulnerabilities associated with specific POLD1 variants .

How does POLD1 expression influence the tumor microenvironment and immune cell infiltration?

POLD1 expression has significant effects on the tumor immune microenvironment, with high expression generally associated with immunosuppressive features:

Immune cell infiltration patterns:
High POLD1 expression correlates with specific immune cell infiltration profiles:

  • Increased infiltration of:

    • CD56dim natural killer cells

    • Regulatory T cells (Tregs)

    • Myeloid-derived suppressor cells (MDSCs)

    • Activated CD8 T cells

  • Decreased infiltration of:

    • Immature dendritic cells

    • Natural killer cells

    • Neutrophils

Association with T cell exhaustion markers:
POLD1 expression positively correlates with T cell exhaustion markers, suggesting a role in immune escape:

  • Significant correlation with CTLA4, LAG3, LGALS9, TGFB1, and PDCD1 (PD-1)

  • Strong association with markers of Tregs and T cell exhaustion

Immunomodulator correlations:
POLD1 levels show significant associations with both immunoinhibitors and immunostimulators:

  • Immunoinhibitors: CTLA4, LAG3, LGALS9, TGFB1, and PD-1

  • Immunostimulators: TNFRSF18, TNFRSF25, TNFRSF8, TNFRSF14, and LTA

  • Chemokines: CCL5, CXCL13, XCL1, and XCL2

  • Chemokine receptors: CXCR3, CXCR5, and CXCR6

Experimental validation approaches:
Researchers have utilized multiple databases and experimental methods to establish these correlations:

  • TIMER and TISIDB databases for immune cell infiltration analysis

  • RT-qPCR, Western blot, and immunohistochemistry for validation

  • Functional and animal experiments for in vitro and in vivo verification

These findings suggest that POLD1 may influence tumor progression partly by creating an immunosuppressive microenvironment, which could have important implications for immunotherapy approaches.

What are the optimal experimental designs for studying POLD1's role in cancer cell proliferation and metastasis?

Based on published studies, a comprehensive experimental design to investigate POLD1's role in cancer should include:

In vitro experimental approaches:

  • POLD1 expression modulation:

    • Knockdown: siRNA or shRNA targeting POLD1 (at least two siRNA sequences for validation)

    • Overexpression: POLD1-expressing plasmids

    • Validation: RT-qPCR and Western blot to confirm efficiency

  • Proliferation assays:

    • MTT/CCK-8 assay at multiple time points (24h, 48h, 72h, 96h)

    • Colony formation assay (14-21 days)

    • Cell cycle analysis by flow cytometry with PI staining

    • EdU incorporation assay for DNA synthesis

  • Migration and invasion assays:

    • Transwell migration assay (24-48 hours)

    • Wound healing assay with time-lapse imaging

    • 3D invasion assays with Matrigel coating

  • Mechanistic investigations:

    • RNA-seq after POLD1 knockdown to identify altered pathways

    • GSEA and GO analysis for functional annotation

    • Immunoblotting for cell cycle proteins (Cyclin E1, Cyclin D1) and EMT markers (E-cadherin, N-cadherin, Vimentin, Snail)

    • Immunofluorescence staining for proliferation markers (Ki67) and EMT proteins

In vivo experimental approaches:

  • Tumor growth models:

    • Subcutaneous injection of POLD1-knockdown or overexpressing cells in immunodeficient mice

    • Tumor size and weight measurements over time

    • IHC staining of tumor tissues for POLD1 and Ki67

  • Metastasis models:

    • Tail vein injection to assess lung metastasis potential

    • Quantification of metastatic foci number and size

    • Optional: orthotopic models for tissue-specific assessment

  • Rescue experiments:

    • Co-expression of downstream targets (e.g., MYC) with POLD1 knockdown to verify mechanism

    • In vivo and in vitro validation of rescue effect

  • Drug sensitivity studies:

    • Treatment with targeted therapies (e.g., ATR inhibitors) in POLD1-modulated cells

    • Combination therapy approaches based on pathway analysis

Studies implementing these approaches have revealed that POLD1 promotes cancer cell proliferation by facilitating G1-S phase transition and enhances metastasis through EMT activation, with potential mechanistic involvement of MYC stabilization .

What controls and validation steps are crucial when using POLD1 antibodies in different experimental techniques?

Proper controls and validation are essential for generating reliable data with POLD1 antibodies:

For Western Blotting:

  • Loading controls:

    • Use appropriate housekeeping proteins (β-actin, GAPDH, α-tubulin)

    • Consider nuclear loading controls (Lamin B1, Histone H3) as POLD1 is predominantly nuclear

  • Specificity controls:

    • Positive controls: Known POLD1-expressing cell lines (Jurkat, K-562, HeLa, MOLT4)

    • Negative controls: POLD1 knockdown/knockout cells

    • Peptide competition assay with immunizing peptide

  • Molecular weight verification:

    • POLD1 should appear at 124 kDa

    • Pre-stained protein ladder to confirm size

  • Additional validation:

    • Multiple antibodies targeting different epitopes

    • Gradient dilution series to determine optimal concentration (1:500-1:3000)

For Immunohistochemistry:

  • Tissue controls:

    • Positive tissue controls: Human breast cancer, colon cancer tissue

    • Negative tissue controls: Tissues known to lack POLD1 expression

    • Isotype control: Normal IgG from same species as primary antibody

  • Antibody controls:

    • Omission of primary antibody

    • Serial antibody dilutions (1:50-1:500) to optimize signal-to-noise ratio

    • Peptide competition control

  • Antigen retrieval optimization:

    • Compare EDTA buffer (pH 9.0) vs. citrate buffer (pH 6.0)

    • Test multiple retrieval durations

  • Staining pattern verification:

    • Nuclear localization expected for POLD1

    • Comparison with RNA-seq or other expression data

For Immunofluorescence:

  • Fixation optimization:

    • Compare 4% paraformaldehyde vs. acetone fixation

    • Test different permeabilization methods (0.1% Triton X-100)

  • Fluorescence controls:

    • Secondary antibody only control

    • Autofluorescence control (unstained sample)

    • Nuclear counterstain (DAPI) for co-localization

  • Dilution optimization:

    • Test range of dilutions (1:250-1:500)

    • Signal intensity quantification

For Immunoprecipitation:

  • IP controls:

    • IgG control from same species as POLD1 antibody

    • Input sample (pre-IP lysate)

    • Optimized antibody amount (0.5-4.0 μg for 1.0-3.0 mg protein)

  • Interaction validation:

    • Reverse IP with interacting protein antibodies

    • IP under different conditions (± DNA damage)

Implementing these controls ensures reliable and reproducible results across different experimental techniques and research questions involving POLD1.

How should researchers integrate POLD1 expression data with other tumor characteristics for comprehensive cancer studies?

Comprehensive integration of POLD1 expression data with other tumor characteristics requires a multidimensional approach:

Data integration framework:

  • Multi-omics data correlation:

    • Genomics: Correlate POLD1 expression with mutation status, copy number variations

    • Transcriptomics: Identify co-expressed genes and pathways through RNA-seq

    • Proteomics: Analyze protein interaction networks involving POLD1

    • Epigenomics: Investigate methylation patterns of POLD1 promoter

  • Clinical data integration:

    • Patient demographics (age, sex, ethnicity)

    • Tumor characteristics (stage, grade, histological subtype)

    • Treatment history and response

    • Survival outcomes (OS, PFS, DSS)

  • Immune landscape correlation:

    • Immune cell infiltration profiles (quantified by deconvolution algorithms)

    • Expression of immune checkpoint molecules

    • Cytokine/chemokine expression patterns

    • Tumor mutational burden and neoantigen load

Analytical approaches:

  • Stratification strategies:

    • Divide patients into high vs. low POLD1 expression groups based on median expression

    • Create patient clusters based on POLD1 and related pathway genes

    • Use machine learning for patient classification

  • Statistical methods:

    • Cox regression for survival analysis (univariate and multivariate)

    • ANOVA or t-tests for group comparisons

    • Correlation coefficients (Pearson, Spearman) for continuous variables

    • Multiple testing correction (FDR, Bonferroni)

  • Pathway and network analysis:

    • Gene Set Enrichment Analysis (GSEA) for biological pathways

    • Protein-protein interaction networks

    • Regulatory network inference

  • Visualization techniques:

    • Heatmaps for expression patterns

    • Kaplan-Meier curves for survival analysis

    • Forest plots for multivariate analysis

    • t-SNE or UMAP for dimension reduction

Validation strategies:

  • Cross-validation in independent cohorts:

    • Use multiple patient datasets (e.g., TCGA, GEO)

    • Split discovery and validation cohorts

  • Experimental validation:

    • In vitro confirmation of key findings

    • Patient-derived xenograft models

    • Prospective clinical validation

Studies implementing these approaches have revealed that POLD1 expression is associated with pathologic tumor stage, histologic grade, immune cell infiltration patterns, and patient survival across multiple cancer types . For example, ccRCC patients with high POLD1 expression show poorer OS, PFS, and DSS, along with specific immune infiltration profiles characterized by increased Treg cells and MDSCs .

How can researchers troubleshoot inconsistent POLD1 antibody staining patterns in immunohistochemistry?

Inconsistent POLD1 antibody staining in IHC can be systematically resolved through the following troubleshooting approach:

Problem: Weak or absent staining

Potential CauseSolution
Insufficient antigen retrievalTry EDTA buffer pH 9.0 as primary method; increase retrieval time or temperature
Antibody concentration too lowTitrate antibody concentration (start with 1:50 for weak signals; recommended range 1:50-1:500)
Tissue fixation issuesOptimize fixation time; consider using tissue fixed for 24-48 hours in 10% neutral buffered formalin
Antibody degradationUse fresh antibody aliquot; check storage conditions (-20°C, with glycerol)
Detection system sensitivitySwitch to more sensitive detection system (polymer-HRP or tyramide signal amplification)

Problem: High background staining

Potential CauseSolution
Antibody concentration too highDilute antibody further (try 1:500 for high background)
Insufficient blockingExtend blocking time; try different blocking reagents (5-10% normal serum, 3-5% BSA)
Cross-reactivityUse more specific antibody; confirm with peptide competition assay
Endogenous peroxidase activityEnhance peroxidase blocking (3% H₂O₂ for 10-15 minutes)
Non-specific bindingInclude 0.1-0.3% Triton X-100 or Tween-20 in wash buffers

Problem: Inconsistent staining across samples

Potential CauseSolution
Tissue processing variationsStandardize fixation and processing protocols; use tissue microarrays for batch consistency
Antigen degradationMinimize time between sectioning and staining; store unstained slides at 4°C
Antibody batch variationUse the same lot number for entire study; include standard control slide in each batch
Protocol inconsistenciesUse automated staining platforms; detailed protocol documentation
Regional tissue variationsTake multiple cores per sample; analyze larger tissue areas

Problem: Unexpected staining pattern

Potential CauseSolution
Non-specific antibodyValidate with alternative antibodies targeting different epitopes
Post-translational modificationsConsider phospho-specific antibodies if relevant
Isoform expressionVerify epitope region against known isoforms
Technical artifactsInclude negative controls (isotype IgG, no primary antibody)
Aberrant POLD1 localizationConfirm with immunofluorescence co-localization studies

Recommended validation workflow:

  • Start with recommended protocol (antigen retrieval with EDTA buffer pH 9.0, 1:100 antibody dilution)

  • Systematically optimize each variable independently

  • Include positive control tissues (human breast cancer, colon cancer)

  • Compare multiple POLD1 antibodies when possible

  • Validate findings with orthogonal methods (WB, IF)

Following these troubleshooting steps will help ensure consistent and reliable POLD1 IHC staining across experimental samples.

What factors should be considered when comparing results from different POLD1 antibodies across studies?

When comparing results from different POLD1 antibodies across studies, researchers should consider several critical factors:

Antibody characteristics comparison:

FactorPotential ImpactAssessment Method
Epitope locationDifferent domains detect different isoforms or truncated proteinsCompare immunogen sequences in product datasheets
ClonalityMonoclonals (higher specificity); Polyclonals (better signal, multiple epitopes)Check antibody type: monoclonal vs. polyclonal
Host speciesAffects secondary antibody selection and background in certain tissuesCompare host species (rabbit most common for POLD1)
Validation extentMore extensively validated antibodies provide higher confidenceReview validation data and publication history
Cross-reactivityDifferent species reactivity profilesCheck tested reactivity (human, mouse, rat)

Methodological considerations:

  • Protocol differences:

    • Antigen retrieval methods (EDTA pH 9.0 vs. citrate pH 6.0)

    • Antibody incubation conditions (time, temperature, concentration)

    • Detection systems (chromogenic vs. fluorescent; amplification methods)

    • Blocking reagents and washing protocols

  • Sample preparation variations:

    • Fixation protocols and duration

    • Processing and embedding techniques

    • Storage conditions and section thickness

    • Fresh vs. archival tissue samples

  • Quantification methods:

    • Scoring systems (H-score, Allred, percentage positive)

    • Manual vs. automated analysis

    • Different thresholds for positive/negative classification

    • Image acquisition parameters

Reconciliation strategies:

  • Direct comparison experiments:

    • Test multiple antibodies on the same sample set

    • Create concordance tables between antibodies

    • Determine conversion factors if possible

  • Validation with orthogonal methods:

    • Correlate IHC with mRNA expression data

    • Confirm with Western blot analysis

    • Verify with functional assays

  • Meta-analysis approaches:

    • Standardize effect sizes rather than absolute measurements

    • Subgroup analysis by antibody type

    • Sensitivity analysis excluding outlier studies

  • Reporting standards implementation:

    • Detailed MIQE guidelines for PCR studies

    • Complete antibody reporting (catalog #, lot #, dilution, protocol)

    • Raw data sharing when possible

When examining literature, researchers should carefully evaluate reported antibody details – for instance, multiple studies have used rabbit polyclonal antibodies (15646-1-AP) , while others employed rabbit monoclonal antibodies (EPR15118) or alternative polyclonal antibodies (ab168827) , each with potentially different detection characteristics.

How can researchers accurately distinguish between wild-type POLD1 and mutant variants in experimental systems?

Accurately distinguishing between wild-type POLD1 and mutant variants requires a targeted experimental approach combining molecular, biochemical, and functional techniques:

Genomic and transcript-level detection:

  • Allele-specific PCR:

    • Design primers that selectively amplify wild-type or mutant POLD1 sequences

    • Include positive controls with known genotypes

    • Optimize annealing temperatures for maximum specificity

  • Sanger sequencing:

    • Direct sequencing of POLD1 exons, particularly exonuclease domains

    • Analysis of chromatograms for heterozygous mutations

    • Cloning and sequencing of individual alleles when necessary

  • Next-generation sequencing:

    • Targeted deep sequencing for low-frequency variants

    • RNA-seq to determine allele-specific expression

    • Confirmation of variant allele frequencies

  • Droplet digital PCR:

    • Absolute quantification of wild-type and mutant alleles

    • Detection of low-frequency variants (<1%)

    • Determination of copy number variations

Protein-level detection:

  • Variant-specific antibodies:

    • Develop antibodies recognizing specific mutations (e.g., R689W)

    • Validate specificity with recombinant proteins

    • Use in Western blot or IHC applications

  • Mass spectrometry:

    • Targeted proteomics to detect variant-specific peptides

    • Label-free quantification of wild-type vs. mutant proteins

    • Phosphoproteomics to detect differential post-translational modifications

  • Immunoprecipitation followed by sequencing:

    • Pull down POLD1 protein and sequence associated DNA

    • Analyze error rates and mutation patterns

Cellular model systems:

  • CRISPR/Cas9 engineered cell lines:

    • Generate isogenic cell lines with specific POLD1 variants

    • Create heterozygous and homozygous mutant models

    • Example: DLD-1 colorectal cancer cells with R689W variant

  • Allele-specific knockout:

    • Selectively target wild-type or mutant alleles

    • Validate by allele-specific expression analysis

    • Example: Heterozygous cell clones expressing only R689W variant

Functional discrimination methods:

  • Polymerase activity assays:

    • In vitro DNA synthesis with purified proteins

    • Measurement of processivity and fidelity

    • Analysis of error rates and types

  • DNA damage response analysis:

    • CHK1 phosphorylation status by Western blot

    • γH2AX foci formation by immunofluorescence

    • Cell cycle checkpoint activation

  • Drug sensitivity profiling:

    • Differential sensitivity to ATR inhibitors

    • Synthetic lethality screening

    • POLD1-R689W variant shows increased sensitivity to ATR inhibitors

  • Mutational signature analysis:

    • Whole-genome sequencing to detect characteristic mutation patterns

    • Application of computational models to predict functional mutations

    • Correlation with COSMIC mutational signatures

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.