PARP9 antibodies are primarily used to:
Detect PARP9 expression in cellular and tissue samples (Western blot, immunohistochemistry)
Study PARP9-protein interactions (co-immunoprecipitation)
Validate PARP9 knockdown/overexpression in experimental models
PARP9 antibodies have been critical in elucidating its role as a non-canonical RNA sensor:
Mechanism: PARP9 binds viral dsRNA (1.1–1.4 kb regions) and activates PI3K/AKT3, phosphorylating IRF3/IRF7 to induce type I interferon (IFN) production. This pathway operates independently of mitochondrial antiviral-signaling protein (MAVS) .
In vivo validation: PARP9-knockout mice show impaired IFN responses and heightened susceptibility to RNA viruses (e.g., VSV, reovirus) .
PARP9 antibodies have identified its overexpression in multiple cancers:
Gastric cancer (GC): High PARP9 expression correlates with poor survival (HR = 1.26, p = 0.032) and promotes proliferation, migration, and invasion in AGS/MKN-45 cells .
Breast cancer: PARP9 drives chemoresistance via PI3K/AKT activation, enabling immune evasion .
| Cancer Type | Expression Level vs. Normal | Prognostic Impact (HR) | Functional Role |
|---|---|---|---|
| Gastric cancer | 3.2-fold increase | 1.26 (p = 0.032) | Enhances metastasis |
| Breast cancer | Not quantified | N/A | Mediates immune escape |
PARP9 antibodies are being explored for:
Diagnostic biomarkers: ROC analysis shows PARP9 predicts gastric cancer prognosis with 91.3% accuracy (AUC = 0.913) .
Therapeutic targeting: Knockdown reduces chemoresistance in breast cancer and inhibits GC cell viability by 40–60% .
Current studies lack standardized PARP9 antibody validation across platforms.
Clinical trials targeting PARP9 in humans are pending, despite promising preclinical results.
When selecting a PARP9 antibody, consider multiple factors based on your experimental needs:
Target species reactivity: Different antibodies show reactivity with specific species (human, mouse, rat). For example, product 17535-1-AP shows reactivity with human and rat samples .
Application compatibility: Verify that the antibody has been validated for your intended application (WB, IHC, IF, IP). For instance, anti-PARP9 antibody ab53796 is suitable for IP and WB applications and reacts with human samples .
Clonality: Decide between polyclonal (broader epitope recognition) and monoclonal (higher specificity) based on your research needs.
Validation data: Review existing publications and validation data. Some antibodies like 17535-1-AP have been cited in multiple publications for applications such as KD/KO, WB, and IF .
Immunogen information: Check if the antibody was raised against a region relevant to your research question. For example, HPA066708 was generated against the immunogen sequence "IDNEVLMAAFQRKKKMMEEKLHRQPVSHRLFQQVPYQFCNVVCRVGFQRMYSTPCDPKYGAGIYFTKNLKNLAEKAKKISAA" .
A comprehensive validation strategy includes:
Positive and negative controls: Use cell lines with known PARP9 expression (e.g., Raji cells, THP-1 cells demonstrate positive WB signals) .
Knockdown/knockout verification: Implement PARP9 knockdown/knockout approaches to confirm specificity, as demonstrated in publications using anti-PARP9 antibodies .
Cross-reactivity assessment: Test for cross-reactivity with other PARP family members, especially similar molecules like PARP14, which has functional interactions with PARP9 .
Epitope mapping: Understand the specific region of PARP9 recognized by your antibody to ensure it detects the form relevant to your research.
Multiple technique confirmation: Validate expression using orthogonal methods (e.g., if using for IHC, confirm with WB).
Titration optimization: Determine optimal working dilutions for your specific application (e.g., HPA066708 recommended dilutions: WB 0.04-0.4 μg/mL, IF 0.25-2 μg/mL) .
Optimizing Western blot for PARP9 detection requires specific considerations:
Expected molecular weight: PARP9 has an observed molecular weight of approximately 88-92 kDa , so ensure your gel resolution is appropriate for this range.
Sample preparation:
Antibody dilution: Start with manufacturer's recommended dilution (e.g., 1:2000-1:12000 for 17535-1-AP) and adjust as needed.
Blocking conditions: 5% non-fat milk in TBST for 1 hour at room temperature generally works well, but BSA may be preferred when studying phosphorylation.
Signal detection: For detecting potentially low levels of endogenous PARP9, consider using enhanced chemiluminescence or fluorescent secondary antibodies to improve sensitivity.
Positive controls: Include lysates from Raji cells or THP-1 cells, which have been demonstrated to express detectable levels of PARP9 .
Stripping and reprobing: If examining pathway components, consider gentle stripping methods to probe for interacting partners like DTX3L or signaling molecules in the PI3K/AKT pathway .
For optimal IHC detection of PARP9:
Tissue processing and fixation: Use 10% neutral-buffered formalin fixation for 24-48 hours, as overfixation may mask epitopes.
Antigen retrieval:
Antibody dilution: Start with recommended dilution (e.g., 1:20-1:200 for 17535-1-AP) and optimize for your specific tissue type.
Positive control tissues: Include spleen, skin, kidney, heart, lung, or ovary tissues as positive controls, as these have demonstrated PARP9 expression .
Counterstaining: Hematoxylin counterstaining provides contrast for nuclear visualization, important as PARP9 may show both nuclear and cytoplasmic localization.
Co-staining considerations: For macrophage-specific PARP9 detection in granulomas or tumor tissues, consider dual staining with CD68 as PARP9 has been shown to co-localize with CD68+ macrophages in tuberculosis granulomas .
Image analysis: Quantify PARP9 expression using digital pathology approaches, particularly for comparative studies between different tissue conditions or disease states.
Interpretation of PARP9 expression requires context-specific considerations:
Cancer contexts:
In gliomas, high PARP9 expression correlates with poor prognosis and advanced clinicopathological features . Compare expression levels with tumor grade and clinical outcomes.
In breast cancer, evaluate PARP9 in relation to chemoresistance mechanisms, particularly through the PI3K/AKT/PD-L1 axis .
Analyze subcellular localization differences between tumor and normal tissues.
Immune response contexts:
In infectious diseases like tuberculosis, PARP9 expression in CD68+ macrophages differs between controllers and progressors . Quantify expression in relation to disease progression markers.
For viral infections, evaluate PARP9 in relation to type I interferon responses and PI3K/AKT pathway activation .
Comparative analysis recommendations:
Use standardized scoring systems (H-score, percentage positive cells, intensity scales).
Implement quantitative image analysis with appropriate controls and statistical methods.
Consider single-cell approaches to resolve heterogeneity in expression patterns.
Distinguishing between PARP family members requires attention to:
Molecular weight differentiation: PARP9 has an observed molecular weight of ~88-92 kDa , which differs from other family members (e.g., PARP1: 116 kDa, PARP14: 170-200 kDa).
Expression pattern analysis:
Functional validation approaches:
Use specific PARP9 knockdown/knockout to confirm phenotypes attributed to PARP9 activity.
Implement domain-specific mutants to distinguish between PARP9 macrodomain functions versus catalytic activities.
Bioinformatic analysis strategies:
Perform correlation analysis between PARP9 and inflammatory gene signatures, as PARP9 shows strong correlation with HCK, LCK, interferon, STAT1, MHC I, and MHC II clusters, but negative correlation with IgG clusters .
Use gene set enrichment analysis to identify PARP9-associated pathways, which typically include antigen processing, B-cell receptor signaling, cytokine-receptor interactions, and JAK-STAT signaling .
PARP9's role in DNA damage repair involves several mechanisms that can be investigated using the following approaches:
PARP9-DTX3L complex analysis:
Recruitment dynamics assessment:
Live-cell imaging with fluorescently tagged PARP9 to track recruitment kinetics to laser-induced DNA damage sites.
ChIP-seq to map PARP9 binding at DNA damage sites genome-wide.
Functional readouts:
Mechanistic considerations:
To investigate PARP9's immunoregulatory functions:
Macrophage-specific studies:
Use bone marrow-derived macrophages from wild-type and Parp9-/- mice to compare cytokine responses to pathogen stimulation (demonstrated differences in IFN-β, IL-10, IL-1α, IL-1β production) .
Implement CRISPR-Cas9 knockout of PARP9 in human macrophage cell lines to assess impact on inflammatory responses.
Type I IFN pathway analysis:
RNA sensing mechanisms:
PI3K/AKT pathway activation:
In vivo infection models:
| Experimental Approach | Application to PARP9 Research | Key Readouts | Relevant Controls |
|---|---|---|---|
| CRISPR-Cas9 Knockout | Generate PARP9-deficient cells/animals | Phenotypic changes in immune response, DNA repair | Non-targeting sgRNA, rescue with WT PARP9 |
| Co-immunoprecipitation | Detect PARP9-DTX3L interactions | Protein complex formation | IgG control, DTX3L knockout |
| Phospho-flow Cytometry | Assess STAT1/STAT6 phosphorylation | Phosphorylation state changes | Isotype controls, pathway inhibitors |
| RNA Immunoprecipitation | Identify direct RNA binding | RNA enrichment | Non-specific IgG, PARP9 mutants |
| Infection Models | Evaluate in vivo relevance | Pathogen burden, survival, cytokine production | Wild-type animals, specific pathway blockade |
Researchers frequently encounter these challenges when working with PARP9 antibodies:
Non-specific binding:
Problem: Detection of bands at unexpected molecular weights.
Solution: Use PARP9 knockout/knockdown controls to identify specific bands; optimize antibody concentration; increase washing steps; consider alternative blocking agents (BSA vs. milk).
Low signal intensity:
Problem: Weak detection of endogenous PARP9.
Solution: Implement signal amplification methods; concentrate protein samples; extend primary antibody incubation time (overnight at 4°C); use enhanced detection reagents.
Nuclear protein extraction difficulties:
Problem: Incomplete extraction of nuclear PARP9.
Solution: Include nuclear lysis steps with sonication; use specialized nuclear extraction buffers with higher salt concentrations; ensure complete cell lysis verification.
Inconsistent immunohistochemistry results:
Cross-reactivity with other PARP family members:
Problem: Difficulty distinguishing PARP9 from related proteins.
Solution: Verify specificity using PARP9-knockout samples; perform peptide competition assays; consider using multiple antibodies targeting different epitopes.
When facing contradictory PARP9 functional data:
Context-dependent role reconciliation:
In viral infections, PARP9 acts as a positive regulator of type I IFN responses , while in tuberculosis, it functions as a negative regulator . This apparent contradiction reflects context-specific functions.
Solution: Carefully document experimental conditions (cell types, stimulation protocols, timing) and incorporate pathway-specific readouts.
Model system variations:
PARP9 may show different effects in human versus mouse systems or in different cell types.
Solution: Validate findings across multiple model systems; use primary cells when possible; compare results from cell lines to primary tissue samples.
Temporal dynamics considerations:
PARP9 functions may differ at early versus late timepoints in immune responses.
Solution: Implement time-course experiments; use inducible expression/deletion systems to control timing of PARP9 modulation.
Interaction partner dependencies:
Pathway cross-talk analysis:
Single-cell methodologies offer unique insights into PARP9 biology:
Single-cell RNA sequencing (scRNA-seq):
Single-cell proteomics:
Enables quantification of PARP9 protein levels and post-translational modifications at single-cell resolution.
Method: Mass cytometry (CyTOF) with validated PARP9 antibodies allows simultaneous detection of PARP9 and signaling pathway components.
Spatial transcriptomics:
Multiparameter imaging:
Analytical considerations:
Implement trajectory analysis to identify transitions in PARP9 expression during cellular differentiation or disease progression.
Use computational deconvolution to infer PARP9-associated signatures from bulk tissue data.
For researchers developing PARP9-targeted compounds:
Target specificity assessment:
Challenge: PARP9 shares structural similarities with other PARP family members.
Approach: Implement in vitro activity assays against a panel of recombinant PARP enzymes; use structural biology (crystallography, cryo-EM) to guide selective inhibitor design.
Domain-specific targeting strategies:
Complex-based considerations:
Cellular validation methods:
Verify target engagement using cellular thermal shift assays (CETSA).
Evaluate functional consequences using pathway-specific readouts (DNA damage repair efficiency, IFN signaling outputs).
In vivo evaluation approaches:
Potential therapeutic applications: