XPA antibodies are immunodetection reagents designed to target the XPA protein (xeroderma pigmentosum group A-complementing protein), which is encoded by the XPA gene (chromosome 9q22.3). XPA is a 273-amino-acid nuclear protein (31–40 kDa) essential for DNA damage recognition and repair via the NER pathway . These antibodies are used to quantify XPA expression, assess DNA repair capacity, and investigate its role in diseases like cancer and xeroderma pigmentosum (XP) .
XPA contains a zinc-finger domain critical for DNA binding . It acts as a scaffold in NER, stabilizing repair complexes and verifying DNA damage (e.g., UV-induced lesions or cisplatin adducts) . Key interactions include:
PCNA: Recruitment to replication foci .
Deficiencies in XPA cause XP, characterized by extreme UV sensitivity, neurodegeneration, and skin cancer .
Commercial XPA antibodies vary in specificity, host species, and applications:
Antibody Name | Clone | Applications | Source |
---|---|---|---|
ab65963 | 5F12 (Mouse) | WB, ICC/IF, ELISA | Abcam |
#14607 | D9U5U (Rabbit) | WB, IHC | Cell Signaling |
SAB4503090 | Polyclonal | WB, IHC, ELISA | Sigma-Aldrich |
XPA expression correlates with chemotherapy resistance and patient outcomes:
XPA mutations (e.g., c.553C>T) cause truncated proteins, leading to severe neurodegeneration or mild phenotypes based on mutation location .
A 2024 NIH study classified 18 XP-A patients by XPA variant location:
Mutation Location | Phenotype | DNA Repair Capacity |
---|---|---|
Exons 3/5 (nonsense) | Severe | <10% of normal |
Intron 4 (splice) | Intermediate | 10–30% |
Exon 1 (frameshift) | Mild | 30–50% |
Specificity: Anti-XPA antibodies must distinguish full-length XPA from truncated isoforms (e.g., XP1UMN fibroblasts show no detectable XPA) .
Storage: Most antibodies require storage at −20°C in buffered solutions .
Cross-Reactivity: Confirmed in human, mouse, and rat samples .
This antibody solution is provided at a concentration of 1mg/ml and is formulated in a buffer consisting of PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
This antibody has undergone rigorous testing in various applications, including ELISA, Western blot analysis, and immunocytochemistry/immunofluorescence (ICC/IF), to ensure its specificity and reactivity. As optimal working dilutions may vary depending on the specific application and experimental conditions, it is recommended to perform a titration to determine the most effective concentration for your experiment.
PAT71H3AT.
Anti-human XPA mAb, clone PAT71H3, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human XPA protein 1-273 amino acids purified from E. coli.
Mouse IgG1 heavy chain and k light chain.
XPA is a DNA repair protein that plays a critical role in the nucleotide excision repair (NER) pathway. It is essential for repairing DNA damage caused by ultraviolet (UV) light and other sources that create bulky DNA adducts . XPA's importance stems from its role in coordinating the assembly of the preincision complex necessary for recognizing and processing damaged DNA. Deficiencies in NER can lead to xeroderma pigmentosum, a genetic disorder that significantly increases the risk of skin cancer due to the accumulation of unrepaired DNA lesions . When designing experiments to study DNA repair mechanisms, XPA antibodies serve as valuable tools for investigating normal repair processes and their dysfunction in disease states.
XPA antibodies are versatile tools that can be employed across multiple experimental platforms. They are validated for western blotting (WB), immunoprecipitation (IP), immunofluorescence (IF), immunohistochemistry with paraffin-embedded sections (IHCP), and enzyme-linked immunosorbent assay (ELISA) . For western blotting, researchers typically use dilutions ranging from 1:2000 to 1:10000, while immunohistochemistry applications generally require dilutions between 1:200 and 1:1000 . When conducting immunoprecipitation experiments, 2-10 μg of antibody per mg of lysate is recommended . For optimal results in immunohistochemistry with paraffin-embedded tissues, epitope retrieval with citrate buffer pH 6.0 is recommended .
To maintain antibody efficacy, XPA antibodies should be stored at 4°C for short-term use (up to one week). For long-term storage, they should be aliquoted and stored at temperatures between -20°C and -80°C to avoid repeated freeze-thaw cycles that may compromise antibody integrity . Most XPA antibodies are formulated in buffers containing preservatives such as sodium azide (typically 0.09%) and may include stabilizers like BSA (0.1%) . When working with these antibodies, it's important to avoid contamination and minimize exposure to light, particularly for fluorescently conjugated variants. Before use, allow the antibody to equilibrate to room temperature and gently mix rather than vortex to prevent protein denaturation.
XPA antibodies are available in multiple formats with different host species and clonal characteristics. The most common types include mouse monoclonal antibodies such as clone 12F5 (IgG2a kappa light chain) and clone AT71H3 (IgG1 kappa) , as well as polyclonal rabbit antibodies . These antibodies can be obtained in non-conjugated forms or with various conjugations including agarose, horseradish peroxidase (HRP), phycoerythrin (PE), fluorescein isothiocyanate (FITC), and multiple Alexa Fluor® conjugates for different detection methods . The selection of antibody type should be based on the specific experimental application, with monoclonals offering higher specificity while polyclonals may provide stronger signals through recognition of multiple epitopes.
When designing controls for XPA antibody experiments, positive controls should include cell lines known to express XPA protein. HeLa, K-562, HEK293T, and Jurkat cells are well-documented to express detectable levels of XPA and are suitable as positive controls for western blot and immunostaining applications . For negative controls, consider using either isotype controls (antibodies of the same isotype but irrelevant specificity) or cells where XPA expression has been knocked down using siRNA or CRISPR-Cas9 methods. Additionally, primary antibody omission controls are essential to confirm the specificity of secondary antibody binding. When performing immunoprecipitation experiments, include a control using non-immune IgG from the same species as the XPA antibody to assess non-specific binding .
For optimal western blotting results with XPA antibodies, cell lysates should be prepared using effective lysis buffers such as NETN (as used in reference studies) . A protein load of 40-50 μg per lane is typically sufficient for detection of endogenous XPA . Recommended antibody dilutions range from 1:2000 to 1:10000, though this may vary by specific antibody and should be optimized . For detection, secondary antibodies conjugated to HRP followed by ECL detection systems provide good sensitivity . When analyzing results, be aware that human XPA protein appears at approximately 40 kDa on western blots. To enhance specificity, blocking with 3-5% BSA or milk in TBS-T is recommended. If signal strength is an issue, consider longer incubation times with primary antibody (overnight at 4°C) rather than increasing antibody concentration.
For immunofluorescence applications, fixation with 4% paraformaldehyde for 15-20 minutes at room temperature preserves XPA protein structure while maintaining cellular architecture. Permeabilization with 0.1-0.5% Triton X-100 for 5-10 minutes allows antibody access to nuclear XPA protein. For paraffin-embedded tissue sections, antigen retrieval is critical; citrate buffer at pH 6.0 is specifically recommended for XPA detection . Typical working dilutions for immunohistochemistry range from 1:200 to 1:1000 , while immunofluorescence may require more concentrated antibody solutions. When analyzing results, remember that XPA is predominantly localized to the nucleus, so co-staining with DAPI helps confirm proper subcellular localization . Non-specific background can be minimized by thorough blocking (2-5% normal serum from the same species as the secondary antibody) and including 0.1-0.3% Triton X-100 in antibody diluents.
For successful immunoprecipitation of XPA protein, begin with fresh cell lysates prepared in non-denaturing buffer systems such as NETN . Use 2-10 μg of XPA antibody per mg of total protein lysate , and incubate the antibody-lysate mixture overnight at 4°C with gentle rotation to maximize antigen-antibody binding. Protein A/G beads are suitable for capturing most XPA antibodies, though protein G is preferable for mouse IgG1 isotypes like clone AT71H3 . After immunoprecipitation, thorough washing (at least 4-5 washes) with lysis buffer containing reduced detergent concentrations is critical to minimize non-specific binding. For elution, either denaturing conditions (SDS sample buffer at 95°C for 5 minutes) or native elution (using excess immunogenic peptide) can be employed depending on downstream applications. When analyzing immunoprecipitated XPA by western blotting, use concentrations of 0.1 μg/ml of detection antibody for optimal results .
XPA antibodies can be powerfully applied to investigate DNA damage response pathways through several advanced approaches. One method involves conducting chromatin immunoprecipitation (ChIP) assays to identify XPA binding sites at DNA damage locations following UV irradiation or treatment with DNA-damaging agents. This approach can reveal the kinetics of XPA recruitment to damage sites and its interaction with other NER factors. Another application is proximity ligation assays (PLA) to visualize and quantify in situ protein-protein interactions between XPA and other NER components like RPA, TFIIH, or ERCC1-XPF. Researchers can also employ XPA antibodies in combination with DNA damage markers (such as CPD or 6-4PP antibodies) in dual immunofluorescence to correlate damage recognition with repair complex assembly. For studying pathway dynamics, XPA antibodies can be used in live-cell imaging experiments when conjugated to appropriate fluorophores or through immunoprecipitation followed by mass spectrometry to identify novel interaction partners in specific damage contexts .
When investigating XPA-RPA interactions in nucleotide excision repair, several methodological considerations are critical. First, be aware that RPA alone can form nonspecific aggregates with DNA at high concentrations, which can be prevented or reversed by the addition of XPA . Therefore, protein concentration ratios are crucial when designing in vitro binding experiments. Second, the order of addition (whether pre-incubating RPA with XPA before adding DNA, or allowing RPA to interact with DNA first before adding XPA) appears to yield similar results, suggesting the DNA-RPA aggregation effect is both preventable and reversible by addition of XPA . For detecting these interactions, ELISA-based protein-protein interaction assays can be performed by coating plates with one purified protein and detecting bound XPA with anti-XPA antibodies at dilutions of approximately 1:1000 . When designing experiments to study these interactions in cells, consider that both proteins are predominantly nuclear, so nuclear extraction protocols that preserve protein-protein interactions are preferable to whole-cell lysates.
Detection of post-translational modifications (PTMs) of XPA requires specialized approaches. While standard XPA antibodies recognize the protein regardless of modification status, phospho-specific or other PTM-specific antibodies may be necessary to detect particular modifications. For studying XPA phosphorylation (particularly at Ser196, which is ATR-dependent and UV-induced), use phospho-specific antibodies in combination with phosphatase inhibitors during sample preparation. To confirm specificity, include controls treated with lambda phosphatase. When investigating ubiquitination or SUMOylation of XPA, perform immunoprecipitation with XPA antibodies followed by western blotting with anti-ubiquitin or anti-SUMO antibodies. Alternatively, perform the reverse experiment by immunoprecipitating with anti-ubiquitin or anti-SUMO antibodies and then detecting XPA in the precipitates. Mass spectrometry following XPA immunoprecipitation can provide comprehensive identification of PTMs. To study the dynamics of modifications, combine these approaches with treatments that induce DNA damage or inhibit specific enzymes involved in adding or removing the modifications of interest.
To investigate XPA's potential roles beyond canonical NER, several methodological approaches using XPA antibodies are valuable. First, employ co-immunoprecipitation with XPA antibodies followed by mass spectrometry to identify novel interaction partners that may connect XPA to other repair pathways. Second, use XPA antibodies in ChIP-seq experiments before and after various types of DNA damage (not limited to UV) to identify whether XPA is recruited to non-canonical damage sites. Third, perform systematic immunofluorescence co-localization studies with markers of different repair pathways (such as γH2AX for double-strand breaks or XRCC1 for base excision repair) following various genotoxic stresses. Fourth, utilize proximity ligation assays to visualize potential in situ interactions between XPA and components of other repair pathways. When designing these experiments, it's crucial to include appropriate positive controls (known NER substrates) and negative controls (damages not expected to involve XPA). Additionally, validation through orthogonal methods is essential, as unexpected findings may result from antibody cross-reactivity or indirect effects rather than novel XPA functions.
When faced with weak or absent signals in XPA antibody applications, a systematic troubleshooting approach is necessary. First, verify protein expression in your samples using positive control cell lines known to express XPA, such as HeLa, K-562, HEK293T, or Jurkat cells . For western blotting, increase protein loading to 50-70 μg per lane and consider extended exposure times. Optimize antibody concentration by testing a range of dilutions, starting with more concentrated solutions than recommended (e.g., 1:500 instead of 1:2000 for western blot). Ensure complete transfer of protein to membranes by using pre-stained markers and post-transfer staining with Ponceau S. For immunostaining applications, thorough antigen retrieval is crucial; try extending citrate buffer treatment time or consider alternative retrieval methods like EDTA buffer (pH 8.0) or enzymatic retrieval. If problems persist, the antibody's epitope may be masked by protein interactions or post-translational modifications; try denaturing conditions or treating samples with appropriate enzymes (phosphatases, deglycosylases) before immunodetection.
High background is a common challenge when working with antibodies, including those targeting XPA. Primary causes include insufficient blocking, excessive antibody concentration, or non-specific binding. For western blotting, increase blocking time to 2 hours or overnight using 5% milk or BSA in TBS-T, and ensure thorough washing between steps (at least 3 x 10 minutes). Dilute antibodies in fresh blocking buffer and filter solutions if necessary. For immunostaining, include 0.1-0.3% Triton X-100 in blocking and antibody diluents to reduce non-specific membrane associations. Consider using alternative blocking agents such as normal serum (5-10%) from the species of the secondary antibody. If using fluorescent detection methods, ensure samples are protected from light, and include an autofluorescence reduction step (such as brief treatment with 0.1% sodium borohydride). For all applications, titrate antibody concentrations carefully; optimal signal-to-noise ratio often occurs at higher dilutions than expected. As a last resort, pre-adsorption of the antibody with cell/tissue lysates lacking XPA can reduce non-specific binding.
When encountering unexpected bands in western blots using XPA antibodies, careful analysis and additional controls are required for accurate interpretation. Human XPA protein typically appears at approximately 40 kDa . Bands at higher molecular weights may represent post-translationally modified forms (phosphorylated, ubiquitinated, or SUMOylated XPA), protein complexes that weren't fully denatured, or non-specific cross-reactivity. Lower molecular weight bands could indicate degradation products, alternative splice variants, or non-specific binding. To distinguish between these possibilities, perform validation experiments: (1) Include positive control lysates from cells with confirmed XPA expression; (2) Compare results with a second XPA antibody recognizing a different epitope; (3) Perform peptide competition assays where pre-incubation of the antibody with its immunizing peptide should eliminate specific bands; (4) For suspected post-translational modifications, treat samples with appropriate enzymes (phosphatases, deubiquitinases) before western blotting; (5) For potential degradation products, add protease inhibitors during sample preparation and minimize freeze-thaw cycles.
Accurate quantification of XPA protein requires careful experimental design and appropriate controls. For western blot quantification, always include a concentration curve of recombinant XPA or a well-characterized reference sample on the same blot to establish linearity of signal. Normalize XPA signals to appropriate loading controls (such as β-actin, GAPDH, or total protein staining with methods like Ponceau S), being mindful that traditional housekeeping genes may vary under certain experimental conditions. For relative quantification between samples, densitometric analysis should be performed within the linear range of detection. When using ELISA for XPA quantification, generate standard curves using purified recombinant XPA protein and ensure samples fall within this range, diluting as necessary. For immunofluorescence quantification, conduct z-stack imaging to capture the total cellular signal and use software that can measure nuclear intensity specifically (as XPA is predominantly nuclear). Include untreated and positive control samples in each experiment, and perform technical replicates (at least triplicate) to account for methodological variation. Statistical analysis should consider both biological and technical variability when determining significance of observed differences.
XPA antibodies offer valuable tools for cancer research applications, particularly given the connection between DNA repair deficiencies and cancer development and treatment. In diagnostic contexts, XPA antibodies can be used in immunohistochemistry to assess XPA expression levels in tumor samples, potentially correlating with prognosis or treatment response. Research applications include studying XPA as a biomarker for chemotherapy resistance, as elevated XPA expression may decrease sensitivity to platinum-based agents and other DNA-damaging therapeutics. For investigating synthetic lethality approaches, XPA antibodies can monitor protein levels following genetic or pharmacological interventions targeting complementary repair pathways. In therapeutic development, XPA antibody-based proximity assays can be employed for high-throughput screening to identify compounds that disrupt XPA interactions with other repair factors, potentially sensitizing cancer cells to DNA-damaging treatments. Additionally, XPA antibodies conjugated to nanoparticles or liposomes might serve as delivery vehicles for targeting cancer cells with elevated nuclear XPA expression, though such applications require extensive validation .
When incorporating XPA antibodies into multiplex imaging systems, several technical considerations are critical for successful implementation. First, antibody compatibility with fixation and antigen retrieval protocols must be verified, as multiplex systems often require compromises in sample preparation that may affect epitope accessibility. Second, carefully select fluorophore conjugates or detection systems that minimize spectral overlap with other targets in your panel. XPA antibodies are available with various conjugations including phycoerythrin (PE), fluorescein isothiocyanate (FITC), and multiple Alexa Fluor® conjugates , allowing flexibility in experimental design. Third, conduct single-staining controls to establish baseline signals and identify any unexpected cross-reactivity or background. Fourth, when designing sequential staining protocols, consider the order of antibody application; applying XPA antibodies early in the sequence may be advantageous if the epitope is sensitive to harsh elution conditions used between rounds. Finally, for quantitative analysis of XPA in multiplex systems, include internal reference standards and ensure image acquisition parameters remain consistent across all samples to allow for meaningful comparisons of expression levels.
XPA antibodies provide valuable tools for investigating the connections between DNA repair capacity and aging processes. One approach involves immunohistochemistry or immunofluorescence analysis of XPA expression in tissue samples from individuals of different ages or in aged animal models, correlating expression patterns with markers of cellular senescence or tissue degeneration. For more mechanistic studies, co-immunoprecipitation using XPA antibodies can identify age-associated changes in XPA interaction partners that might impair repair efficiency. In cell culture models of senescence, XPA antibodies can monitor changes in protein levels, subcellular localization, and recruitment kinetics to DNA damage sites through live-cell imaging or fixed-cell immunofluorescence. For studying post-translational regulation during aging, XPA immunoprecipitation followed by mass spectrometry can identify age-specific modifications that might alter protein function. Additionally, chromatin immunoprecipitation (ChIP) with XPA antibodies can assess whether aging affects the recruitment of XPA to endogenous DNA damage sites genome-wide. When designing these experiments, include appropriate age-matched controls and consider tissue-specific differences in DNA repair capacity and aging rates.
XPA antibodies offer significant potential for understanding neurodegenerative conditions linked to DNA repair defects, particularly since progressive neurodegeneration is a hallmark of xeroderma pigmentosum when it involves neurological symptoms. Research applications include immunohistochemical analysis of post-mortem brain tissue to compare XPA expression and localization patterns between patients with neurodegenerative diseases and healthy controls. In cellular models of neurodegeneration, XPA antibodies can monitor the protein's nuclear localization, which may be disrupted in disease states. Co-immunoprecipitation experiments using XPA antibodies can identify altered protein interactions in affected neurons, potentially revealing disease mechanisms. For studying the relationship between oxidative stress (common in neurodegenerative conditions) and XPA function, immunofluorescence co-localization with markers of oxidative DNA damage can determine whether XPA is recruited to these lesions in neuronal cells. In animal models, immunohistochemistry with XPA antibodies can track age-dependent changes in different brain regions, correlating with behavioral deficits. When designing these studies, it's important to account for regional and cell type-specific differences in XPA expression within the nervous system, using appropriate neuronal markers for co-localization studies.
Xeroderma Pigmentosum (XP) is a rare autosomal recessive disorder characterized by extreme sensitivity to ultraviolet (UV) rays from sunlight, with a high predisposition to skin cancers and, in some cases, neurological abnormalities . The disease is caused by defects in the nucleotide excision repair (NER) pathway, which is responsible for repairing UV-induced DNA damage .
XPA is one of the seven complementation groups (A-G) of XP, each corresponding to a different gene involved in the NER pathway . The XPA gene encodes a protein that plays a crucial role in the NER process by verifying DNA damage and stabilizing the repair complex . Mutations in the XPA gene lead to a defective NER pathway, resulting in the accumulation of unrepaired DNA damage and increased mutagenesis .
The mouse anti-human XPA antibody is a monoclonal antibody developed to target the human XPA protein. This antibody is used in various research applications, including immunofluorescence, Western blotting, and immunoprecipitation, to study the expression and function of the XPA protein in human cells .
Research involving the XPA protein and its corresponding antibody has provided significant insights into the mechanisms of DNA repair and the pathogenesis of XP. For instance, studies have shown that the XPA protein is essential for repairing clustered oxidative DNA adducts, which are complex forms of DNA damage caused by ionizing radiation . Additionally, the mouse anti-human XPA antibody has been used to investigate the localization and function of the XPA protein in various cell types .
Current therapeutic approaches for XP primarily focus on preventing UV exposure and managing skin cancers. However, recent research has explored novel strategies to enhance DNA repair and reduce the disease burden. For example, specific sulfonylureas have been shown to promote the clearance of DNA damage and increase resistance to UV radiation in cellular models of XP . Moreover, nicotinamide and caloric restriction have demonstrated potential in attenuating the effects of UV radiation and decreasing DNA damage burden in animal models .