P4HB Antibody Pair

Shipped with Ice Packs
In Stock

Description

Components and Formulations

The P4HB Antibody Pair typically consists of two antibodies: a capture antibody (immobilized on a solid phase) and a detector antibody (conjugated to a reporter enzyme or fluorescent label). Key formulations include:

SupplierComponentsKey FeaturesCitations
Abcam (ab253826)Recombinant capture + detector antibodiesCarrier-free, BSA/azide-free, validated for ELISA/Western blot
VWR (H00005034-PW1)Rabbit polyclonal (IP) + mouse monoclonal (WB)Immunoprecipitation/Western blot compatibility
Bio-Techne (NBP2-79305)Mouse monoclonal capture + HRP-conjugated detectorSandwich ELISA, 47 pg/ml sensitivity

Applications in Research

The antibody pair is utilized across multiple techniques to study P4HB’s role in cellular processes:

ELISA (Enzyme-Linked Immunosorbent Assay)

  • Sensitivity: Detects P4HB levels as low as 47 pg/ml (Bio-Techne kit) .

  • Sample Types: Serum, cell lysates, or tissue homogenates .

  • Example Use: Quantified P4HB in bladder cancer tissues to establish its prognostic value .

Western Blotting

  • Band Size: 57 kDa (validated in HepG2, HeLa, and A431 cell lysates) .

  • Optimal Dilution: 1/1000–1/2000 (Abcam) .

Immunoprecipitation

  • Efficiency: Enriches P4HB from complex mixtures for downstream analysis .

  • Validation: Demonstrated in studies linking P4HB to autophagy in diabetic nephropathy .

Immunohistochemistry (IHC)

  • Tissue Compatibility: Breast carcinoma, brain, and kidney tissues .

  • Dilution: 1/100–1/500 (requires heat-mediated antigen retrieval) .

Role in Cancer

  • Bladder Cancer: High P4HB expression correlates with advanced tumor stages and poor prognosis .

  • Liver Cancer: P4HB knockdown sensitizes chemoresistant cells to adriamycin .

Autophagy and Disease

  • Diabetic Nephropathy: Elevated P4HB levels serve as an autophagy-related biomarker .

  • Hepatocellular Carcinoma (HCC): P4HB promotes tumorigenesis via epithelial-to-mesenchymal transition (EMT) .

ER Stress and Chaperone Activity

  • Chaperone Function: At high concentrations, P4HB prevents protein aggregation; at low concentrations, it facilitates aggregation .

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on your chosen shipping method and location. Please contact your local distributor for specific delivery estimates.
Notes
We recommend using the capture antibody at a concentration of 0.9 µg/mL and the detection antibody at a concentration of 0.25 µg/mL. For optimal results, we suggest determining the ideal dilutions through experimental validation.
Synonyms
ERBA2L, PDI, PDIA1, PO4DB,P4HB,Cellular thyroid hormone-binding protein,Prolyl 4-hydroxylase subunit beta,p55
Target Names

Q&A

What is P4HB and what are its primary cellular functions?

P4HB (Protein disulfide-isomerase) is a multifunctional protein that plays crucial roles in cellular protein homeostasis. It primarily catalyzes the formation, breakage, and rearrangement of disulfide bonds. Within cells, P4HB forms and rearranges disulfide bonds of nascent proteins. At the cell surface, it functions as a reductase that cleaves disulfide bonds of attached proteins, potentially causing structural modifications of exofacial proteins. At high concentrations and following phosphorylation by FAM20C, it acts as a chaperone inhibiting aggregation of misfolded proteins, while at low concentrations, it facilitates protein aggregation (anti-chaperone activity). Additionally, P4HB serves as a structural subunit of various enzymes including prolyl 4-hydroxylase and microsomal triacylglycerol transfer protein (MTTP) .

How does the dual function of P4HB as both chaperone and anti-chaperone impact experimental design?

When designing experiments involving P4HB, researchers must consider its concentration-dependent dual functionality. At high concentrations, particularly after phosphorylation by FAM20C, P4HB functions as a chaperone that prevents misfolded protein aggregation. Conversely, at low concentrations, it demonstrates anti-chaperone activity by facilitating protein aggregation . This dual functionality necessitates careful control of P4HB concentrations in experimental settings. Researchers should include concentration gradients in their protocols and monitor phosphorylation status when studying protein folding dynamics or aggregation pathways. Additionally, experimental designs should incorporate appropriate controls that account for both functions to accurately interpret results related to protein misfolding diseases or protein quality control mechanisms.

How can researchers effectively validate P4HB antibody specificity for their experimental system?

Effective validation of P4HB antibody specificity requires implementing multiple complementary approaches. First, perform Western blot analysis using positive control lysates from cells known to express P4HB (HepG2, HeLa, 293T, or A431 cells) to confirm detection of the expected 57 kDa band . Include negative controls using P4HB knockout cell lines (as demonstrated with P4HB knockout A431 cells) to verify signal absence . For immunocytochemistry applications, conduct parallel staining in wild-type and P4HB-depleted cells, confirming specificity through signal reduction or elimination in knockdown/knockout samples. Additionally, validate antibody performance across multiple experimental methods (Western blot, immunohistochemistry, flow cytometry) to ensure consistent target recognition. When working with new tissue or cell types, perform pre-adsorption tests by pre-incubating the antibody with purified P4HB protein before application to samples. Finally, compare results obtained using different antibodies targeting distinct epitopes of P4HB to confirm consistent localization and expression patterns.

What controls are essential when using P4HB antibody pairs in multiplex immunoassays?

When implementing P4HB antibody pairs in multiplex immunoassays, several essential controls must be incorporated to ensure data validity. First, include isotype controls using irrelevant antibodies of the same isotype as your P4HB antibodies to assess non-specific binding . Second, incorporate positive controls using samples with verified P4HB expression (such as HeLa or HepG2 cell lysates) to confirm assay functionality . Third, employ negative controls using samples lacking P4HB expression, ideally P4HB knockout cell lines, to establish background signal thresholds . For capture-detection antibody pair systems, include controls testing each antibody individually to identify any cross-reactivity issues. Additionally, implement concentration calibration controls using recombinant P4HB protein standards across a physiologically relevant range to establish quantitative relationships between signal intensity and protein concentration. Finally, when multiplexing with other targets, perform single-target controls to identify and mitigate potential cross-reactivity between different antibody sets that could compromise data interpretation.

What are the optimal fixation and permeabilization conditions for detecting P4HB in different subcellular compartments?

Optimization of fixation and permeabilization conditions is critical for accurate detection of P4HB in different subcellular compartments due to its diverse functional locations. For endoplasmic reticulum-localized P4HB detection, 4% paraformaldehyde fixation for 10 minutes provides excellent structural preservation while maintaining antigenicity . This method has been successfully demonstrated in both HeLa and U2OS cells for immunofluorescence applications. For detecting cell surface P4HB, which functions as a reductase for exofacial proteins, gentler fixation with 2% paraformaldehyde without permeabilization is recommended to preserve surface epitopes. When investigating both intracellular and surface P4HB simultaneously, researchers should consider a sequential approach: first staining for surface P4HB before fixation, followed by permeabilization and staining for intracellular pools. For flow cytometry applications, two validated methods are available: 80% methanol fixation for 5 minutes followed by 0.1% PBS-Tween permeabilization for 20 minutes, or alternatively, 100% methanol fixation for 5 minutes with subsequent 0.1% PBS-Triton X-100 permeabilization for 5 minutes . These optimized protocols ensure maximal epitope accessibility while maintaining cellular architecture.

How should researchers design experiments to distinguish between P4HB's chaperone versus anti-chaperone activities?

Designing experiments to differentiate between P4HB's concentration-dependent chaperone and anti-chaperone activities requires careful methodological considerations. Begin by establishing a concentration gradient experiment using purified recombinant P4HB protein (spanning from nanomolar to micromolar ranges) incubated with aggregation-prone substrate proteins. Monitor protein aggregation kinetics using techniques such as thioflavin T fluorescence, light scattering, or sedimentation assays across the concentration range. To specifically examine the chaperone activity at high concentrations, researchers should conduct experiments with and without FAM20C-mediated phosphorylation of P4HB, as this post-translational modification enhances its chaperone function . For cellular studies, develop systems with tunable P4HB expression levels using inducible expression vectors, allowing precise control over intracellular P4HB concentrations. Additionally, create phosphomimetic (e.g., serine to aspartate) and phospho-deficient (serine to alanine) P4HB mutants to distinguish phosphorylation-dependent effects. Time-course analyses should be included to capture the dynamic nature of P4HB's dual functionality. Finally, employ fluorescence resonance energy transfer (FRET) assays to directly visualize P4HB-substrate interactions under different concentration conditions.

What technical considerations are important when using P4HB antibody pairs for quantitative analyses in clinical samples?

When implementing P4HB antibody pairs for quantitative analyses in clinical samples, several technical considerations are essential for generating reliable results. First, thorough validation of the antibody pair's analytical performance is crucial, including assessments of specificity, sensitivity, linear range, and limit of detection using recombinant P4HB standards and reference samples . For clinical applications, researchers must develop standardized protocols with stringent quality control measures, as P4HB has demonstrated potential as a diagnostic and prognostic biomarker in bladder cancer with high discriminatory power (AUC values of 0.888 and 0.881 in different cohorts) . Sample collection, processing, and storage conditions should be rigorously standardized to minimize pre-analytical variables that might affect P4HB levels or antibody binding. When analyzing tissues, consider using adjacent sections for P4HB immunostaining to correlate protein levels with histopathological features. For liquid biopsies, account for potential confounding factors such as hemolysis or platelet activation that might affect circulating P4HB levels. Additionally, implement robust normalization strategies and include appropriate calibration curves with each analytical run. Finally, statistical analysis should incorporate multivariate models that account for potential confounding clinical variables when evaluating P4HB as a biomarker.

How can researchers address inconsistent P4HB antibody staining patterns in immunohistochemistry?

Inconsistent P4HB antibody staining patterns in immunohistochemistry (IHC) can result from multiple technical and biological factors that require systematic troubleshooting. First, optimize antigen retrieval methods, as P4HB detection in formalin-fixed paraffin-embedded (FFPE) tissues requires effective heat-mediated antigen retrieval before IHC staining . Test multiple buffer systems (citrate pH 6.0 vs. EDTA pH 8.0) and retrieval durations to determine optimal conditions for your specific tissue type. Second, evaluate fixation variables, as overfixation can mask P4HB epitopes; standardize fixation times and conditions across specimens. Third, optimize antibody concentration by performing titration experiments to identify the optimal dilution that maximizes specific signal while minimizing background. For the rabbit monoclonal P4HB antibody [EPR9499], a 1/100 dilution has been validated for human breast carcinoma and brain tissues . Fourth, consider tissue-specific factors by implementing positive control tissues with established P4HB expression patterns alongside experimental samples. Fifth, evaluate the detection system by comparing different secondary antibody conjugates and signal amplification methods. Finally, implement rigorous controls, including isotype controls and P4HB-depleted tissues, to distinguish between specific and non-specific staining.

What strategies can resolve non-specific banding patterns in Western blots using P4HB antibodies?

Resolving non-specific banding patterns in Western blots using P4HB antibodies requires implementation of several optimization strategies. First, adjust blocking conditions by testing different blocking agents (BSA, milk, commercial blockers) and concentrations to reduce non-specific binding. For P4HB antibodies, a blocking solution of 1% BSA with 10% normal goat serum and 0.3M glycine in 0.1% PBS-Tween has been validated . Second, optimize antibody dilutions - the rabbit monoclonal P4HB antibody [EPR9499] (ab137110) performs optimally at 1/1000 to 1/2000 dilution for Western blotting applications in various cell lines (HepG2, HeLa, 293T, A431) . Third, implement stringent washing protocols with increased wash duration and detergent concentration to remove weakly bound antibodies. Fourth, validate sample preparation methods, ensuring complete protein denaturation and reducing conditions are maintained to expose the P4HB epitope fully. Fifth, employ gradient gels to achieve better separation of proteins with similar molecular weights to the 57 kDa P4HB target. Sixth, consider peptide competition assays using the immunizing peptide (within Human P4HB aa 50-100 for antibody ab264363) to confirm band specificity. Finally, compare results with alternative P4HB antibodies targeting different epitopes to identify consistently appearing bands that likely represent true P4HB signal.

How should researchers address potential cross-reactivity when using P4HB antibodies in multi-species studies?

Addressing potential cross-reactivity in multi-species studies using P4HB antibodies requires thorough validation and strategic experimental design. Begin by assessing sequence homology of P4HB across target species through bioinformatic analysis, focusing particularly on the antibody's epitope region. For antibody ab264363, the immunogen corresponds to a synthetic peptide within Human P4HB aa 50-100 , and for antibody ab137110, evaluate sequence conservation in this region across species. Validate each antibody empirically in all target species tissues or cells using positive and negative controls specific to each species. For example, while ab137110 has been validated for human, mouse, and rat samples , and ab264363 for human and mouse samples , additional validation may be required for other species. Implement species-specific controls including P4HB-depleted samples from each species when possible. Consider using Western blot analysis to confirm the antibody detects proteins of the expected molecular weight (57 kDa) across all species before proceeding to more complex applications. When cross-reactivity is unavoidable, design experiments to include species-specific antibodies as alternatives or modify experimental approaches to accommodate limitations. Finally, when interpreting results from multi-species studies, acknowledge the potential impact of differential antibody affinity across species on quantitative comparisons.

How can P4HB antibody pairs be utilized in studying endoplasmic reticulum stress responses?

P4HB antibody pairs offer powerful tools for investigating endoplasmic reticulum (ER) stress responses due to P4HB's integral role in protein folding and its regulation during ER stress. Researchers can employ P4HB antibodies in co-localization studies with other ER stress markers to visualize stress-induced changes in P4HB distribution and expression. Immunofluorescence techniques using specific P4HB antibodies like the rabbit monoclonal [EPR9499] (ab137110) at 0.2μg/ml concentration can be combined with antibodies against other ER stress proteins to map the temporal and spatial dynamics of the stress response . For quantitative assessment of ER stress, implement time-course experiments measuring P4HB protein levels via Western blotting, as P4HB expression often changes during the unfolded protein response. P4HB involvement in protein processing in the endoplasmic reticulum, including N-glycan modification and protein metabolic processes responding to ER stress, makes it an informative marker in these pathways . Additionally, researchers can develop pulse-chase experiments with P4HB immunoprecipitation to track client protein interactions during stress conditions. For in vivo models, tissue-specific immunohistochemistry using validated P4HB antibodies can help identify cell populations experiencing ER stress in disease models, particularly in cancer where P4HB shows altered expression patterns .

What approaches can be used to study the interaction between P4HB and LGALS9 in T helper cell migration?

Investigating the interaction between P4HB and galectin-9 (LGALS9) in T helper cell migration requires sophisticated methodological approaches that leverage specific P4HB antibodies. Begin with proximity ligation assays (PLA) using P4HB and LGALS9 antibodies to visualize and quantify direct interactions in situ within Th2 T helper cells. Follow with co-immunoprecipitation experiments using validated P4HB antibodies to pull down protein complexes and confirm interactions with LGALS9, coupled with reciprocal immunoprecipitations. For dynamic studies, implement live cell imaging using fluorescently tagged P4HB antibody fragments to track P4HB-LGALS9 interactions during T cell migration. To assess functional consequences, design transmigration assays comparing wild-type and P4HB-depleted T helper cells, measuring their migratory capacity in response to LGALS9 gradients. Additionally, employ flow cytometry with P4HB antibodies optimized for this application (such as ab137110 at 1/300 dilution) to quantify surface P4HB levels on Th2 cells under different conditions. To directly assess the impact on plasma membrane redox state, use redox-sensitive fluorescent probes in combination with P4HB surface labeling. Finally, validate in vivo relevance through adoptive transfer experiments with P4HB-depleted versus control T helper cells, tracking their migratory patterns and LGALS9 interactions in inflammatory models.

What statistical approaches are most appropriate for analyzing P4HB expression data in cancer biomarker studies?

When analyzing P4HB expression data in cancer biomarker studies, researchers should implement comprehensive statistical approaches tailored to biomarker validation. For diagnostic applications, receiver operating characteristic (ROC) curve analysis is essential to evaluate P4HB's discriminatory power between tumor and normal tissues, as demonstrated in bladder cancer studies where P4HB showed impressive area under the curve (AUC) values of 0.888 (95% CI: 0.801–0.975; P<0.001) and 0.881 (95% CI: 0.825–0.937; P<0.001) in different cohorts . For prognostic evaluation, perform both univariate and multivariate Cox proportional hazards regression analyses to determine if P4HB expression represents an independent risk factor for survival outcomes. Complement this with Kaplan-Meier survival analysis with log-rank tests to visualize and compare survival curves between patient groups with different P4HB expression levels . For robust biomarker development, implement cross-validation techniques and independent validation cohorts to confirm findings across different patient populations. Additionally, consider correlation analyses between P4HB expression and established clinicopathological parameters using appropriate tests (Spearman's or Pearson's correlation). Finally, for mechanistic insights, pathway enrichment analysis and protein-protein interaction network analysis can help position P4HB within relevant biological contexts, as demonstrated by studies linking P4HB to endoplasmic reticulum stress pathways in cancer .

How can researchers reconcile conflicting findings regarding P4HB function across different experimental systems?

Reconciling conflicting findings regarding P4HB function across experimental systems requires systematic consideration of multiple factors that influence its context-dependent activities. First, evaluate concentration-dependent effects, as P4HB exhibits dual functionality – chaperone activity at high concentrations and anti-chaperone activity at low concentrations . These opposing functions may explain apparently contradictory results obtained at different P4HB levels. Second, assess phosphorylation status, as FAM20C-mediated phosphorylation significantly impacts P4HB's chaperone activity . Experiments lacking phosphorylation analysis may miss this regulatory dimension. Third, consider cellular localization context, as P4HB functions differently at the cell surface versus intracellularly . Conflicting findings may result from studies focusing on different subcellular pools without proper compartmentalization analysis. Fourth, examine substrate specificity, as P4HB interacts with diverse client proteins, potentially resulting in substrate-specific functional outcomes. Fifth, analyze tissue-specific factors, as microenvironmental conditions in different tissues may modulate P4HB function, particularly relevant when comparing cancer versus normal contexts . Finally, review methodological differences in experimental systems, including cell types, antibody clones, detection methods, and analysis approaches. When publishing, researchers should explicitly document these parameters to facilitate accurate cross-study comparisons and reconciliation of disparate findings.

What considerations are important when integrating P4HB data into broader systems biology frameworks?

Integrating P4HB data into broader systems biology frameworks requires careful consideration of its multifunctional nature and diverse interaction network. First, implement multi-omics integration strategies that combine P4HB protein expression data with transcriptomics, proteomics, and phenotypic data to position P4HB within functional networks. Pathway enrichment analysis has already revealed P4HB's involvement in protein processing in the endoplasmic reticulum, N-glycan modification, and protein metabolic processes responding to ER stress . Second, construct protein-protein interaction networks centered on P4HB, as has been done using the Search Tool for the Retrieval of Interacting Genes, to identify hub proteins and functional modules . Third, apply dynamic modeling approaches that account for P4HB's dual functionality as both chaperone and anti-chaperone depending on concentration and phosphorylation status . Fourth, incorporate spatial context in your analysis, as P4HB functions differently at the cell surface versus intracellularly. Fifth, consider tissue-specific regulatory networks, particularly in cancer contexts where P4HB shows altered expression patterns with prognostic significance . Sixth, implement network perturbation analysis through simulation of P4HB knockdown/overexpression to predict system-wide effects. Finally, develop visualization tools that effectively communicate P4HB's position within complex biological networks to enhance interpretation of its role in normal physiology and disease states.

What emerging technologies might enhance P4HB research beyond current antibody-based methods?

Several emerging technologies hold promise for advancing P4HB research beyond traditional antibody-based approaches. First, CRISPR-based protein tagging systems could enable endogenous tagging of P4HB, allowing real-time visualization of dynamics without antibody limitations. Second, proximity labeling techniques (BioID, APEX) coupled with mass spectrometry would provide comprehensive mapping of P4HB's context-dependent interactome across different subcellular compartments and concentration conditions. Third, single-cell multi-omics approaches could reveal cell-specific P4HB expression patterns and functions within heterogeneous tissues, particularly relevant in cancer where P4HB shows diagnostic and prognostic value . Fourth, advanced live-cell imaging technologies including lattice light-sheet microscopy could capture P4HB trafficking between cellular compartments with unprecedented temporal and spatial resolution. Fifth, protein structure analysis through cryo-electron microscopy might elucidate structural changes associated with P4HB's transition between chaperone and anti-chaperone functions at different concentrations and phosphorylation states . Sixth, organoid and tissue-on-chip technologies could provide more physiologically relevant models for studying P4HB function in complex 3D environments. Finally, developing P4HB-specific small molecule modulators through high-throughput screening would allow precise temporal control over P4HB activity in experimental systems, enabling more sophisticated functional studies.

How might P4HB antibody research contribute to developing targeted therapies for ER stress-related diseases?

P4HB antibody research could significantly advance targeted therapy development for ER stress-related diseases through multiple translational pathways. First, P4HB antibodies enable precise mapping of altered P4HB expression and localization patterns in disease states, identifying patient populations that might benefit from P4HB-targeted interventions. This approach is particularly promising in cancers where P4HB overexpression correlates with poor prognosis, as demonstrated in bladder cancer studies . Second, therapeutic antibodies targeting extracellular P4HB could modulate its reductase activity at the cell surface, potentially disrupting cancer cell migration that depends on P4HB-mediated plasma membrane redox regulation . Third, antibody-drug conjugates directed against P4HB could deliver cytotoxic payloads specifically to cells overexpressing P4HB, such as cancer cells. Fourth, diagnostic applications using P4HB antibody pairs could facilitate patient stratification for clinical trials and personalized medicine approaches, leveraging P4HB's strong discriminatory power between normal and cancer tissues (AUC values ~0.88) . Fifth, functional P4HB antibodies that modulate its chaperone versus anti-chaperone activities could help rebalance protein homeostasis in diseases characterized by protein misfolding. Finally, antibody-based screening platforms could accelerate identification of small molecule P4HB modulators by providing robust detection methods for high-throughput drug discovery campaigns targeting ER stress pathways.

Table 1: P4HB Antibody Applications and Validated Conditions

ApplicationAntibody TypeOptimal DilutionValidated Cell/Tissue TypesSpecial Considerations
Western BlotRabbit monoclonal [EPR9499]1/1000-1/2000HepG2, HeLa, 293T, A431, mouse spleenExpected band size: 57 kDa
ImmunohistochemistryRabbit monoclonal [EPR9499]1/100Human breast carcinoma, human brainRequires heat-mediated antigen retrieval
Flow CytometryRabbit monoclonal [EPR9499]1/300HeLaFix with 4% paraformaldehyde
ImmunocytochemistryRabbit monoclonal [EPR9499]0.2 μg/mlHeLa, U2OS100% methanol fixation (5 min)
Antibody Pair AssaysBSA and Azide free pairVariable based on assay formatVarious human samplesCarrier-free formulation for quantitative measurement

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.