PARP2 Antibody

Poly (ADP-Ribose) Polymerase 2, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

PARP2 antibodies target the PARP2 enzyme (UniProt ID: Q9UGN5), an ADP-ribosyl transferase that catalyzes poly(ADP-ribosyl)ation (PARylation) of proteins involved in chromatin remodeling, DNA repair, and metabolism . PARP2 is distinct from PARP1 in substrate specificity and biological roles, such as its preference for DNA gaps over nicks and regulation of replication stress .

DNA Repair and Replication Stress

  • PARP2 antibodies have been used to study the enzyme’s role in break-induced replication (BIR), where PARP2 promotes DNA end resection and mitotic DNA synthesis during replication stress .

  • PARP2 deficiency increases telomere fragility and replication stress-induced genomic instability, detectable via reduced PARP2 signal in knockout models .

Cancer and Metastasis

  • In breast cancer models, PARP2 knockdown (validated via antibody-based assays) reduced lung metastasis but unexpectedly increased bone metastasis, highlighting microenvironment-specific roles .

  • PARP2 inhibition in T cells reduced TNFα and IL-17 levels in LPS-induced colitis, suggesting therapeutic potential for inflammatory diseases .

Immunomodulation

  • T cell-specific PARP2 knockout mice (validated using PARP2 antibodies) showed altered ERK/NF-κB signaling and reduced oxidative stress, linking PARP2 to immune regulation .

Technical Considerations

  • Specificity: Antibodies like ABIN6259061 and 20555-1-AP are validated for cross-reactivity with human, mouse, and rat samples .

  • Sample Preparation: Antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) is recommended for IHC .

  • Controls: Include PARP2-deficient cell lysates or tissues to confirm antibody specificity .

Therapeutic Implications

PARP2 antibodies are critical for evaluating PARP inhibitor efficacy in oncology and inflammation. For example:

  • PARP2 suppression in T cells reduced colitis severity in mice, supporting targeted PARP2 inhibition as a strategy for inflammatory bowel disease .

  • PARP2’s role in replication stress makes it a biomarker for cancers with high genomic instability .

Limitations and Future Directions

  • Current antibodies show limited reactivity in non-mammalian models (e.g., zebrafish) .

  • Development of isoform-specific antibodies is needed to dissect PARP2’s roles in overlapping pathways with PARP1 .

Product Specs

Introduction
Poly (ADP-Ribose) Polymerase 2 (PARP2) is an enzyme involved in DNA repair and other cellular processes. Unlike PARP1, it lacks an N-terminal DNA binding domain but possesses a catalytic domain homologous to PARP1. PARP2 catalyzes poly (ADP-ribosyl)ation and may play a role in nuclear and/or nucleolar targeting due to potential DNA-binding properties within its N-terminal region. Two alternatively spliced transcript variants encoding distinct isoforms have been identified.
Physical Appearance
Clear, colorless liquid without any particles.
Formulation
The antibody is supplied in a solution containing 1mg/ml of antibody in PBS at pH 7.4, with 10% Glycerol and 0.02% Sodium Azide.
Storage Procedures
Store at 4°C for up to 1 month. For long-term storage, store at -20°C. Avoid repeated freezing and thawing.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This antibody has been validated for use in ELISA and Western blot applications, demonstrating high specificity and reactivity. Optimal working dilutions should be determined by titration for each specific application. For Western blot analysis, a dilution range of 1:500 to 1:5000 is recommended, with a starting dilution of 1:500.
Synonyms

ADPRT2, ADPRTL2, ADPRTL3, ARTD2, pADPRT-2, PARP-2, Poly [ADP-ribose] polymerase 2, hPARP-2, ADP-ribosyltransferase diphtheria toxin-like 2, NAD (+) ADP-ribosyltransferase 2, Poly [ADP-ribose] synthase 2.

Purification Method
PARP2 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT29G4A.
Immunogen
Anti-human PARP2 mAb, clone PAT29G4A, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human PARP2 protein 233-583 amino acids  purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and Kappa light chain. 

Q&A

What is PARP2 and what is its primary function in cells?

PARP2 is a poly-ADP-ribosyltransferase that mediates the poly-ADP-ribosylation of proteins and plays a key role in DNA repair mechanisms. It catalyzes the transfer of ADP-ribose moieties from NAD+ to target proteins, building polymers with an average chain length of 20-30 units . PARP2 can target glutamate, aspartate, or serine residues on acceptor proteins, with serine ADP-ribosylation being the primary form in response to DNA damage .

Beyond its fundamental role in the Base Excision Repair pathway, PARP2 has recently been identified as a crucial player in the cellular response to replication stress, particularly at telomeres. It promotes replication stress-induced telomere fragility and prevents telomere loss following chronic induction of oxidative DNA lesions and BLM helicase depletion . PARP2 facilitates this process by promoting DNA end resection, strand invasion, and BIR-dependent mitotic DNA synthesis through the orchestration of POLD3 recruitment and activity .

How do PARP2 antibodies differ in their targeting specificity?

PARP2 antibodies vary significantly in their targeting specificity based on the epitope regions they recognize. Available antibodies target different regions of the PARP2 protein, including internal regions and specific amino acid sequences such as AA 14-149, AA 401-450, or the N-terminal region (AA 10-40) . This targeting diversity affects both the specificity of detection and the range of applications for which each antibody is suitable.

The choice of target region directly impacts both the antibody's reactivity across species and its utility in different experimental contexts. Antibodies targeting highly conserved regions typically demonstrate broader cross-species reactivity, with some PARP2 antibodies showing verified reactivity in multiple species including human, mouse, rat, dog, cow, horse, and rabbit . When designing experiments, researchers should consider whether their experimental question requires detection of specific domains or functional regions of PARP2, as this will guide the selection of an appropriate antibody.

What are the key differences between PARP1 and PARP2 functions?

The molecular basis for these functional differences may relate to the distinct roles these enzymes play in DNA repair pathways. While both contribute to base excision repair, PARP2 specifically mediates formation of branched poly-ADP-ribosylation, which is recognized by specialized factors like APLF . Additionally, PARP2 has emerged as a key player in telomere maintenance through the break-induced replication pathway, a function not prominently associated with PARP1 .

What are the optimal applications for different types of PARP2 antibodies?

Different types of PARP2 antibodies are optimized for specific experimental applications based on their characteristics. Polyclonal PARP2 antibodies, which recognize multiple epitopes, are versatile and commonly used in Western blotting (WB), ELISA, immunofluorescence (IF), and immunocytochemistry (ICC) applications . These antibodies are particularly valuable for detecting endogenous levels of total PARP2 protein.

For more specific applications requiring higher specificity, monoclonal antibodies like the Rabbit Recombinant Monoclonal PARP2 antibody [EPR24240-29] are preferred. This particular antibody has been validated for Western blotting applications and shows specific reactivity with human samples . When studying the role of PARP2 in DNA repair mechanisms, researchers should consider antibodies that can effectively detect the protein in chromatin contexts, where PARP2 recognizes and binds DNA breaks.

When designing experiments to monitor poly(ADP-ribosyl)ation activity rather than just PARP2 presence, complementary approaches using anti-PAR (anti-poly(ADP-ribose)) antibodies can be implemented, as demonstrated in studies with Arabidopsis where poly(ADP-ribosyl)ated proteins were monitored following bleomycin treatment .

How can I validate the specificity of a PARP2 antibody for my experimental system?

Validating PARP2 antibody specificity requires a multi-faceted approach to ensure reliable experimental outcomes. One robust validation method is utilizing genetic knockout systems, where antibody reactivity is compared between wild-type samples and PARP2 knockout/knockdown samples. This approach was demonstrated in Arabidopsis studies where anti-PARP2 antibodies were used to detect PARP2 in complementation experiments with T-DNA insertion mutants .

For human systems, validation can include:

  • Western blot analysis with recombinant PARP2 protein as a positive control

  • Peptide competition assays where the antibody is pre-incubated with the immunizing peptide

  • siRNA knockdown of PARP2 followed by immunoblotting to confirm reduced signal

  • Immunoprecipitation followed by mass spectrometry to verify pull-down of PARP2

  • Cross-validation using multiple antibodies targeting different epitopes of PARP2

When validating across species, ensure the chosen antibody has demonstrated reactivity with your species of interest. Published datasets show that some PARP2 antibodies have verified reactivity across multiple species including human, mouse, rat, dog, and others, while some are more species-restricted .

What methods are most effective for studying PARP2-mediated poly(ADP-ribosyl)ation?

Studying PARP2-mediated poly(ADP-ribosyl)ation requires techniques that can detect both the enzyme and its activity. A comprehensive approach includes:

For accurate interpretation of results, researchers should consider the influence of HPF1, which interacts with PARP2 and alters its substrate specificity from glutamate/aspartate to serine residues, while also limiting polymer chain length .

How does the interaction between PARP2 and HPF1 affect experimental design?

The interaction between PARP2 and HPF1 introduces important considerations for experimental design when studying ADP-ribosylation. HPF1 fundamentally alters PARP2 function in two critical ways: it shifts substrate specificity and regulates polymer length . When HPF1 associates with PARP2, the enzyme preferentially catalyzes serine ADP-ribosylation rather than glutamate or aspartate modification. Additionally, HPF1 restricts PARP2's polymerase activity to limit poly-ADP-ribose chain length .

This interaction creates several experimental design implications:

  • Subcellular compartmentalization: Experiments should account for the localization of both PARP2 and HPF1, as their interaction occurs within chromatin contexts.

  • Temporal dynamics: The timing of HPF1 recruitment affects when and where serine ADP-ribosylation occurs, requiring time-course experiments to fully characterize the process.

  • Target protein analysis: Different target proteins may be modified depending on whether HPF1 is present, necessitating comparative proteomic approaches.

  • ADP-ribosylation detection methods: Researchers must employ antibodies or analytical techniques capable of distinguishing between serine ADP-ribosylation (predominant with HPF1) and glutamate/aspartate ADP-ribosylation (occurring without HPF1).

  • Chain length assessment: Methods to determine poly-ADP-ribose chain length are important when comparing conditions with varying HPF1 levels, as HPF1 restricts polymer extension.

When investigating PARP2's role in DNA repair, researchers should consider how HPF1 influences the recruitment of repair factors through its regulation of serine ADP-ribosylation, which promotes chromatin decompaction and facilitates access to DNA lesions .

What experimental approaches are recommended for studying PARP2's role in telomere maintenance?

PARP2's emerging role in telomere maintenance requires specialized experimental approaches. Recent research has demonstrated that PARP2 promotes replication stress-induced telomere fragility and prevents telomere loss under conditions of chronic oxidative DNA damage or BLM helicase depletion . To investigate these functions, researchers should consider the following experimental strategies:

  • Telomere fragility assays: Utilize fluorescence in situ hybridization (FISH) with telomere-specific probes to visualize and quantify telomere fragility in PARP2-proficient versus PARP2-deficient cells under replication stress conditions.

  • Break-induced replication (BIR) assessment: Employ DNA fiber analysis to measure BIR-dependent DNA synthesis at telomeres, comparing control and PARP2-knockdown conditions to evaluate PARP2's contribution to this process.

  • Strand invasion analysis: Develop assays to measure the efficiency of strand invasion during homology-directed repair at telomeres, focusing on how PARP2 facilitates this process.

  • POLD3 recruitment studies: Use chromatin immunoprecipitation or proximity ligation assays to investigate how PARP2 orchestrates POLD3 recruitment to damaged telomeres, which is critical for BIR-dependent mitotic DNA synthesis .

  • Oxidative stress models: Establish experimental systems with controlled induction of oxidative DNA lesions to study how PARP2 prevents telomere loss under these conditions.

  • Combined depletion experiments: Design studies that simultaneously deplete PARP2 and BLM helicase to investigate their functional relationship in telomere maintenance.

These approaches should be implemented with consideration of cell type-specific effects, as telomere biology varies considerably between normal and cancer cells, and between different model organisms .

How can researchers distinguish between PARP1 and PARP2 functions in experimental systems?

Distinguishing between PARP1 and PARP2 functions requires strategic experimental design, particularly given their overlapping roles in DNA repair. Several approaches can help delineate their specific contributions:

  • Genetic knockout models: Generate single and double knockout systems for PARP1 and PARP2 to compare phenotypes. As demonstrated in Arabidopsis studies, knockout of PARP2 severely depleted detectable PARP activity despite PARP1's presence, whereas substantial poly(ADP-ribosyl)ation remained in PARP1 knockout plants . This approach revealed the unexpected predominance of PARP2 in plant systems, contrary to the animal paradigm.

  • Selective inhibition: Employ chemical biology approaches with inhibitors that show selectivity between PARP1 and PARP2, though finding highly selective inhibitors remains challenging.

  • Domain-specific antibodies: Utilize antibodies that recognize unique structural domains of each protein to track their distinct localizations and interactions.

  • Substrate specificity analysis: Investigate the distinct patterns of poly-ADP-ribosylation catalyzed by each enzyme. PARP2 specifically mediates formation of branched poly-ADP-ribosylation recognized by factors like APLF, providing a functional signature .

  • Interaction partner identification: Map the unique protein-protein interaction networks of PARP1 versus PARP2 using techniques like BioID or proximity-dependent biotin labeling.

  • Complementation experiments: Perform cross-complementation studies where PARP1 is expressed in PARP2-deficient systems and vice versa to determine functional redundancy and unique activities.

  • Context-specific activation: Investigate conditions where one enzyme predominates, such as PARP2's emerging specialized role in telomere maintenance and break-induced replication .

These approaches collectively provide a comprehensive framework for dissecting the distinct functions of these related enzymes across different biological contexts and experimental systems.

What are common pitfalls when using PARP2 antibodies in research?

Researchers frequently encounter several challenges when working with PARP2 antibodies. Understanding these pitfalls is essential for generating reliable experimental data:

  • Cross-reactivity issues: Some PARP2 antibodies may cross-react with PARP1 or other PARP family members due to conserved domains. This problem is particularly pronounced in polyclonal antibodies targeting regions with high sequence homology. Always validate antibody specificity using appropriate controls, including PARP2 knockout samples .

  • Species-specific variations: PARP2 antibody reactivity varies significantly across species. While some antibodies show broad reactivity (human, mouse, rat, dog, cow, horse, rabbit), others are more restricted . Always verify that your selected antibody has been validated for your species of interest.

  • Post-translational modification interference: ADP-ribosylation of PARP2 itself or nearby proteins may mask antibody epitopes, leading to inconsistent detection. Consider using PAR-degrading enzymes like PARG in sample preparation to minimize this effect.

  • Fixation and embedding sensitivity: For immunohistochemistry applications, some epitopes may be sensitive to fixation methods or embedding procedures. Optimize fixation protocols specifically for PARP2 detection.

  • Activation state-dependent detection: PARP2 undergoes conformational changes upon activation, which may expose or conceal certain epitopes. Consider using multiple antibodies targeting different regions to ensure detection regardless of activation state.

  • Quantification challenges: When quantifying PARP2 levels by western blotting, inconsistent transfer of higher molecular weight proteins or variable stripping efficiency during reprobing can affect results. Use appropriate loading controls and consistent protocols.

  • Background signal in tissues with high endogenous peroxidase: For IHC applications, thorough blocking of endogenous peroxidase activity is critical to minimize background that can obscure specific PARP2 staining.

How can researchers optimize detection of PARP2 activity in different experimental contexts?

Optimizing PARP2 activity detection requires tailored approaches for different experimental contexts:

  • Cell-free biochemical assays: For in vitro studies, recombinant PARP2 protein can be used with NAD+ and appropriate substrates. Include HPF1 to study serine ADP-ribosylation, or omit it to study glutamate/aspartate modification . Measure activity through NAD+ consumption or direct detection of ADP-ribosylated products.

  • Cellular systems under DNA damage conditions:

    • Induce DNA damage with agents like bleomycin at optimized concentrations (2.5-10 μg/ml has shown effective induction in plant studies)

    • Extract proteins under conditions that preserve ADP-ribosylation (include PARG inhibitors)

    • Detect poly(ADP-ribosyl)ated proteins using anti-PAR antibodies via western blotting

    • Include appropriate controls (PARP1/2 knockout or knockdown cells)

  • Chromatin context studies:

    • Use chromatin fractionation to isolate chromatin-bound PARP2

    • Employ ChIP-seq to map PARP2 binding sites at DNA breaks

    • Combine with proximity ligation assays to detect PARP2 interactions with repair factors

  • Telomere-specific activity:

    • Use telomere-specific ChIP to detect PARP2 recruitment to telomeres

    • Implement DNA fiber analysis to study PARP2's role in break-induced replication at telomeres

    • Assess telomere fragility through FISH following replication stress in PARP2-proficient versus deficient cells

  • Live-cell activity monitoring:

    • Use fluorescently tagged PAR-binding domains to visualize poly(ADP-ribosyl)ation in real-time

    • Implement FRET-based sensors to detect conformational changes associated with PARP2 activation

  • Tissue-specific activity:

    • Optimize protein extraction protocols for different tissue types

    • Consider using tissue-specific knockout models to determine the relative contribution of PARP2 versus PARP1 in different tissues

In all contexts, researchers should include appropriate negative controls (PARP2-deficient samples) and positive controls (samples with known PARP2 activity) to validate their detection methods.

What emerging technologies might enhance PARP2 antibody applications in research?

Several emerging technologies hold promise for enhancing PARP2 antibody applications in research:

  • Nanobodies and single-domain antibodies: These smaller antibody formats offer improved tissue penetration and epitope accessibility, potentially enhancing detection of PARP2 in complex tissues or when investigating protein-protein interactions in constrained nuclear compartments.

  • CRISPR-based tagging: Endogenous tagging of PARP2 using CRISPR/Cas9 technology allows visualization and tracking of the native protein without overexpression artifacts, enabling more physiologically relevant studies of PARP2 dynamics.

  • Proximity-dependent biotinylation: Technologies like BioID or TurboID fused to PARP2 can map its protein interaction landscape in different cellular contexts, providing insights into its functional roles beyond what traditional antibody applications reveal.

  • Super-resolution microscopy: Combining highly specific PARP2 antibodies with techniques like STORM or PALM can reveal the nanoscale organization of PARP2 at DNA damage sites, offering unprecedented spatial resolution of its recruitment and activity.

  • Mass spectrometry-based proteomics: Advanced proteomic approaches can identify specific ADP-ribosylation sites on target proteins and distinguish between modifications catalyzed by PARP1 versus PARP2, particularly when combined with knockout models for each enzyme .

  • Single-cell technologies: Adapting PARP2 antibodies for single-cell proteomics or CyTOF analysis can reveal cell-to-cell variability in PARP2 expression and activity, particularly relevant in heterogeneous tissues or tumors.

  • Intrabodies and intracellular antibody fragments: These engineered antibody formats function within living cells and could be developed to track or even modulate PARP2 activity in real-time without cell fixation.

  • Antibody-based biosensors: Developing conformational sensors using PARP2 antibodies could allow real-time monitoring of PARP2 activation states in response to DNA damage or replication stress.

How might understanding PARP2 function influence future therapeutic approaches?

Recent discoveries about PARP2's unique functions, particularly in replication stress response and telomere maintenance, suggest several promising therapeutic directions:

  • PARP2-specific inhibitors for cancer therapy: The identification of PARP2's role in promoting break-induced replication at telomeres suggests that PARP2-specific inhibitors might selectively target cancer cells with high levels of replication stress . This approach could potentially reduce the side effects associated with pan-PARP inhibitors that also target PARP1.

  • Combination therapies targeting replication stress pathways: PARP2 inhibition could be strategically combined with agents that exacerbate replication stress or target complementary pathways, such as ATR inhibitors or G-quadruplex stabilizers that further challenge telomere replication.

  • Biomarkers for therapeutic response: Understanding the differential expression and activity of PARP2 across cancer types might provide biomarkers to predict which tumors will respond best to PARP-targeted therapies.

  • Targeting PARP2-HPF1 interactions: The critical role of HPF1 in modulating PARP2 substrate specificity suggests that disrupting this interaction could represent a novel therapeutic approach distinct from catalytic inhibition . This strategy could selectively impair serine ADP-ribosylation while leaving other PARP functions intact.

  • Plant pathogen resistance: The predominant role of PARP2 in plant immune responses suggests agricultural applications, where modulating PARP2 activity might enhance crop resistance to pathogens .

  • Neurological applications: Given PARP's roles in DNA repair and cellular stress responses, PARP2-targeted approaches might be relevant for neurodegenerative conditions characterized by DNA damage accumulation.

Future therapeutic development will benefit from deeper understanding of the distinct functions of PARP family members across different biological contexts and disease states, moving beyond the current paradigm of relatively non-selective PARP inhibition toward more precisely targeted approaches.

Product Science Overview

Introduction

Poly (ADP-Ribose) Polymerase 2 (PARP-2) is a member of the PARP family of proteins, which play a crucial role in various cellular processes, including DNA repair, transcription, and chromatin remodeling. PARP-2, along with PARP-1, is involved in the cellular response to DNA damage and is essential for maintaining genomic stability.

Structure and Function

PARP-2 is a nuclear enzyme that catalyzes the transfer of ADP-ribose units from NAD+ to target proteins, forming poly (ADP-ribose) chains. This process, known as poly (ADP-ribosyl)ation, is a post-translational modification that regulates protein function and interactions. PARP-2 has a DNA-binding domain, an automodification domain, and a catalytic domain, which are essential for its activity .

Role in DNA Damage Response

PARP-2 is activated by DNA strand breaks and works in conjunction with PARP-1 to detect and repair DNA damage. Upon activation, PARP-2 synthesizes poly (ADP-ribose) chains that recruit DNA repair proteins to the site of damage. This process is critical for the repair of single-strand breaks and the maintenance of genomic integrity .

Interaction with Other Proteins

PARP-2 interacts with several proteins involved in the DNA damage response and cell cycle regulation. It has been shown to associate with centromeres in a cell-cycle-dependent manner, accumulating at centromeres during prometaphase and metaphase, and disassociating during anaphase . PARP-2 also interacts with PARP-1, Cenpa, Cenpb, and Bub3, but not with Cenpc .

Therapeutic Potential

PARP inhibitors (PARPi) have emerged as promising therapeutic agents for the treatment of cancers with deficiencies in homologous recombination repair, such as BRCA1/2-mutated tumors. PARP-2, along with PARP-1, is a target for these inhibitors, which block poly (ADP-ribosyl)ation and trap PARP proteins on damaged DNA, leading to cell death . However, resistance to PARPi remains a challenge, and ongoing research aims to develop combinatorial treatment strategies to overcome this resistance .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.