PRDX1 monoclonal antibodies are engineered to bind specifically to human PRDX1, with applications spanning Western blot (WB), immunofluorescence (IF), immunoprecipitation (IP), and immunohistochemistry (IHC). Key antibodies include:
Molecular Weight Variability: PRDX1 is detected at 22 kDa in standard WB and 28 kDa in Simple Western systems , likely due to assay-specific conditions.
Species Reactivity: M01845-2 reacts with human, mouse, and rat PRDX1 , while MAB3488 is human-specific .
Subcellular Localization: IF studies show cytoplasmic PRDX1 staining in MCF-7 cells , consistent with its role in oxidative stress defense.
PRDX1 monoclonal antibodies have advanced studies on cancer, oxidative stress, and immune modulation.
Tumor Biomarker: PRDX1 overexpression is linked to esophageal squamous cell carcinoma (ESCC) and other cancers. Monoclonal antibodies detected elevated PRDX1 in ESCC tissues vs. adjacent normal tissues . Autoantibody responses to PRDX1 were observed in 13.2% of ESCC patients vs. 0% in controls .
Mechanistic Insights: PRDX1 knockdown (validated via WB using monoclonal antibodies) enhanced APE1 redox activity, promoting NF-κB-mediated IL-8 expression in cancer cells . PRDX1 silencing increased HCC cell migration and invasion .
NK Cell Protection: PRDX1 upregulation in NK cells (via IL-15 priming) enhances survival under oxidative stress, as demonstrated in breast cancer models .
ERα Protection: PRDX1 overexpression mitigates oxidative damage to estrogen receptor α (ERα), linking PRDX1 expression to better prognosis in ER-positive breast tumors .
Antibody-Based Validation: Monoclonal antibodies confirmed PRDX1’s interaction with APE1 in IP studies, revealing its role in regulating redox-dependent transcription .
Prognostic Biomarker: High PRDX1 expression correlates with low tumor grade and improved recurrence-free survival in breast cancer .
Cross-Reactivity Challenges: While M01845-2 detects PRDX1 across species, other antibodies (e.g., MAB3488) are human-specific, requiring careful selection based on experimental models.
IF Limitations: No commercially available anti-PRDX1 monoclonal antibodies are validated for IF, as noted in studies requiring polyclonal antibodies .
Therapeutic Targeting: PRDX1’s chaperone and antioxidant roles suggest potential as a therapeutic target, but further studies are needed to translate antibody-based findings into clinical applications.
PRDX1 (Peroxiredoxin 1) is a member of the peroxiredoxin family of antioxidant enzymes that reduce hydrogen peroxide and alkyl hydroperoxides to water and alcohols, respectively. It plays a dual role in cellular physiology: protecting cells against oxidative stress by detoxifying peroxides and functioning as a sensor in hydrogen peroxide-mediated signaling pathways . PRDX1 can also act as a molecular chaperone under conditions of oxidative stress, helping to prevent protein damage .
At the molecular level, PRDX1 contains a conserved N-terminal cysteine (Cys51) that becomes oxidized by H₂O₂ to form cysteine-sulfenic acid. This oxidized form subsequently reacts with the Cys172-SH of another PRDX1 protein, forming a protective disulfide dimer structure . This mechanism is essential for PRDX1's antioxidant functions and its role in redox homeostasis maintenance.
PRDX1 plays differential roles across immune cell populations. Notably, natural killer (NK) cells are more susceptible to oxidative stress compared to T and B cells, partly due to lower endogenous PRDX1 expression . In NK cells, priming with interleukin-15 (IL-15) upregulates PRDX1 expression, which subsequently protects these cells from oxidative stress-induced dysfunction .
The relationship between PRDX1 and oxidative stress susceptibility has significant implications for immunotherapy. Research indicates that PRDX1 overexpression in engineered NK cells, particularly PD-L1-targeting CAR NK cells, confers improved survival and enhanced antitumor function under oxidative stress conditions typical of the tumor microenvironment (TME) . This suggests PRDX1 as a potential target for enhancing cellular immunotherapy approaches against solid tumors.
Current commercial PRDX1 monoclonal antibodies are validated for multiple research applications:
Application | Recommended Dilution | Validated in |
---|---|---|
Western Blot (WB) | 1:1000-3000 | Human, Mouse, Rat |
Immunohistochemistry (IHC) | 1:50-300 | Human, Mouse, Rat |
Immunofluorescence (IF) | 1:100-200 | Human, Mouse, Rat |
ELISA | 1:32000 | Human |
These antibodies typically recognize a protein with an observed molecular weight of 21-23 kDa on Western blots, which aligns with the predicted molecular weight of PRDX1 (22.1 kDa) . Validation experiments have confirmed specificity in multiple human cell lines, including HepG2 liver cancer cells .
Quantifying PRDX1 expression in different immune cell populations requires a multi-modal approach. For protein-level assessment, Western blot analysis using validated monoclonal antibodies (0.01-0.03 μg/mL concentration) against PRDX1 is recommended, with β-actin serving as the appropriate loading control .
For gene expression analysis, quantitative PCR (qPCR) has been successfully employed to measure PRDX1 mRNA levels, with normalization to housekeeping genes such as β-actin . When comparing immune cell subsets (T, B, and NK cells), it's critical to isolate pure populations using magnetic or fluorescence-activated cell sorting before analysis.
For single-cell resolution, flow cytometric analysis can be employed using fluorophore-conjugated PRDX1 antibodies, though this requires careful optimization of fixation and permeabilization protocols due to PRDX1's distribution in both cytoplasmic and nuclear compartments. For spatial context in tissues, immunofluorescence microscopy using PRDX1 antibodies (diluted 1:100-200) allows visualization of expression patterns while enabling co-localization studies with cell type-specific markers .
Several strategies leveraging PRDX1's protective functions can enhance NK cell resilience against oxidative stress:
The most robust evidence supports genetic modification approaches, particularly for cell therapy applications targeting solid tumors where oxidative stress in the tumor microenvironment represents a significant barrier to efficacy .
The interaction between PRDX1 and APE1 (Apurinic/apyrimidinic endonuclease 1) represents a previously unrecognized regulatory mechanism in inflammatory signaling. PRDX1 physically associates with APE1 under physiological conditions in both nuclear and cytosolic compartments . This interaction has significant functional consequences:
PRDX1 acts as a negative regulator of APE1's redox activity by blocking its ability to reduce and activate the transcription factor NF-κB .
When PRDX1 is depleted (via shRNA knockdown), APE1's detection in the nucleus is enhanced, and its redox activity increases .
The functional consequence of enhanced APE1 redox activity following PRDX1 knockdown is increased expression of the proinflammatory chemokine interleukin-8 (IL-8) .
This regulatory mechanism positions PRDX1 as an anti-inflammatory protein that prevents excessive proinflammatory gene expression by restraining APE1 redox activity. This interaction was confirmed through multiple methodologies, including tandem affinity purification, co-immunoprecipitation, and gel filtration chromatography, which identified a 60 kDa complex containing both APE1 and PRDX1 .
Detection of PRDX1 in different subcellular compartments requires tailored approaches:
For Western Blot Analysis of Subcellular Fractions:
Perform subcellular fractionation to isolate nuclear and cytoplasmic fractions using commercially available kits or differential centrifugation protocols.
Load 35 μg of protein per lane for reliable detection.
Use RIPA buffer for protein extraction to ensure efficient solubilization of PRDX1 from different compartments .
Employ PRDX1 monoclonal antibodies at 0.01-0.03 μg/mL concentration with 1-hour primary antibody incubation .
Use compartment-specific markers as controls: HDAC1 or Lamin B1 for nuclear fractions and GAPDH or α-tubulin for cytoplasmic fractions.
For Immunofluorescence Microscopy:
Fix cells with 4% paraformaldehyde (10 minutes at room temperature).
Permeabilize with 0.1% Triton X-100 (5 minutes).
Block with 3% BSA in PBS (1 hour).
Incubate with PRDX1 monoclonal antibody at 1:100-200 dilution (overnight at 4°C) .
Counterstain with DAPI to visualize nuclei.
Use confocal microscopy for accurate assessment of nuclear vs. cytoplasmic localization.
When analyzing PRDX1's subcellular distribution, particularly in the context of oxidative stress, it's important to note that rapid changes in localization can occur. Research has demonstrated that PRDX1 detection in the nucleus can be enhanced when its binding partner APE1 is more accessible, as observed in PRDX1 knockdown experiments .
Comprehensive validation of PRDX1 antibody specificity requires multiple approaches:
Positive and Negative Controls:
Multiple Detection Methods:
Cross-Reactivity Assessment:
Evaluate potential cross-reactivity with other peroxiredoxin family members (PRDX2-6) through recombinant protein panels
For antibodies claimed to work across species (human, mouse, rat), confirm specificity in each organism separately
Validation in Knockout/Knockdown Systems:
Epitope Mapping:
When validating monoclonal antibodies against PRDX1, researchers should be aware that some commercial antibodies are produced using specific synthetic peptides (e.g., SDPKRTIAQDYG) corresponding to human PRDX1 , while others use recombinant protein immunogens . This information can be valuable when selecting the most appropriate antibody for a particular application.
Assessing PRDX1 enzymatic activity requires methodologies that can measure its peroxidase function:
Hydrogen Peroxide Consumption Assay:
Principle: Measures the rate of H₂O₂ decomposition by PRDX1
Methodology:
Purify PRDX1 from samples via immunoprecipitation
Add known concentration of H₂O₂
Measure remaining H₂O₂ via colorimetric methods (e.g., FOX assay)
Calculate consumption rate compared to controls
Thioredoxin-Coupled Assay System:
Principle: PRDX1 activity depends on the thioredoxin system for recycling
Methodology:
Combine sample with thioredoxin, thioredoxin reductase, and NADPH
Initiate reaction with H₂O₂
Monitor NADPH oxidation at 340 nm
Activity is proportional to NADPH consumption rate
Redox Western Blot:
Principle: Distinguishes between reduced and oxidized forms of PRDX1
Methodology:
Treat cells with varying concentrations of oxidants
Lyse cells in the presence of alkylating agents to prevent artificial oxidation
Perform non-reducing SDS-PAGE to preserve disulfide bonds
Probe with PRDX1 antibodies to detect monomeric (reduced) and dimeric (oxidized) forms
Cellular Hydrogen Peroxide Sensor Systems:
Principle: Measures impact of PRDX1 on cellular H₂O₂ levels
Methodology:
Express genetically-encoded H₂O₂ sensors (e.g., HyPer) in control and PRDX1-modified cells
Challenge with oxidative stress
Measure fluorescence changes that reflect H₂O₂ levels
Compare peroxide elimination rates
When interpreting PRDX1 activity data, researchers should consider that under extreme oxidative conditions, PRDX1 can form high-molecular-weight oligomers that function as molecular chaperones but lose peroxidase activity . This functional switch represents an important consideration when analyzing PRDX1 behavior under different oxidative stress conditions.
PRDX1 monoclonal antibodies provide valuable tools for investigating immunotherapy resistance mechanisms:
Profiling PRDX1 Expression in Tumor Microenvironments:
Immunohistochemistry using PRDX1 antibodies (1:50-300 dilution) can map expression patterns within tumors and the surrounding microenvironment .
This allows correlation of PRDX1 levels with oxidative stress markers and treatment response.
Multiplex immunofluorescence combining PRDX1 antibodies with immune cell markers can identify which cell populations maintain PRDX1 expression within the tumor.
Monitoring NK Cell Fitness for Immunotherapy:
Western blot analysis with PRDX1 antibodies can assess baseline PRDX1 expression in patient-derived NK cells .
This information may help predict NK cell resilience to oxidative stress in the tumor microenvironment.
Flow cytometry using permeabilized cells and PRDX1 antibodies can evaluate PRDX1 levels at the single-cell level across immune populations.
Evaluating Engineered Therapeutic Cells:
Assessing Oxidative Stress Adaptation Mechanisms:
PRDX1 antibodies can track changes in expression following exposure to oxidative stress or tumor-derived factors.
Co-immunoprecipitation with PRDX1 antibodies followed by mass spectrometry can identify novel interaction partners that emerge under stress conditions.
Research has demonstrated that NK cells are particularly vulnerable to oxidative stress compared to T and B cells, with PRDX1 deficiency identified as a key factor in this susceptibility . This finding has significant implications for NK cell-based cancer immunotherapies, as PRDX1 expression levels may serve as a biomarker for predicting therapeutic efficacy or as a target for enhancing cell therapy products.
Several experimental approaches using PRDX1 monoclonal antibodies can elucidate its role in inflammatory regulation:
PRDX1-APE1 Interaction Analysis:
Co-immunoprecipitation with PRDX1 antibodies followed by APE1 detection can assess whether this interaction is altered in disease states .
Proximity ligation assays can visualize the interaction in situ within tissues.
FPLC gel filtration chromatography can isolate the PRDX1-APE1 complex (60 kDa) from cellular extracts under various conditions .
Functional Impact on NF-κB Signaling:
ChIP assays using PRDX1 antibodies can determine whether PRDX1 is present at NF-κB-regulated promoters.
Reporter gene assays in PRDX1-manipulated cells can quantify NF-κB transcriptional activity.
Immunoblotting for phosphorylated p65 can assess NF-κB activation status in relation to PRDX1 levels.
Inflammatory Cytokine Production:
qPCR and ELISA assays can measure IL-8 production in cells with modulated PRDX1 expression .
Multiplex cytokine assays can provide a broader view of the inflammatory landscape affected by PRDX1.
Single-cell RNA sequencing of PRDX1-manipulated cells can reveal global transcriptional changes in inflammatory pathways.
In Vivo Disease Models:
Immunohistochemistry with PRDX1 antibodies (1:50-300 dilution) can track expression in tissues during disease progression .
Tissue microarrays probed with PRDX1 antibodies can efficiently screen expression across multiple patient samples.
Conditional PRDX1 knockout models combined with antibody validation can establish causality in disease pathology.
Research has established that PRDX1 can act as an anti-inflammatory protein by preventing APE1 from activating the transcription factor NF-κB, thereby checking excessive expression of proinflammatory cytokines like IL-8 . This mechanism represents a novel regulatory pathway in inflammation that could have implications for diseases characterized by chronic inflammatory states, including cancer progression where IL-8 plays roles in invasion and metastasis.
PRDX1 monoclonal antibodies serve as valuable tools for exploring redox-sensitive protein interactions:
Redox-Preserved Immunoprecipitation:
Differential Interaction Mapping Under Oxidative Stress:
Methodology:
Expose cells to graduated levels of H₂O₂ or other oxidants
Perform PRDX1 immunoprecipitation at each condition
Identify condition-specific interaction partners
Validate with reciprocal immunoprecipitation
In Situ Proximity Ligation Assay:
Methodology:
Use PRDX1 antibodies in combination with antibodies against suspected interaction partners
Apply oligonucleotide-linked secondary antibodies that generate fluorescent signals when in close proximity
Quantify interaction signals under different redox conditions
This provides spatial context for interactions within cells
Size Exclusion Chromatography with Antibody Detection:
Methodology:
Fractionate cellular lysates by gel filtration
Analyze fractions by western blot using PRDX1 antibodies
Identify high-molecular-weight complexes containing PRDX1
Re-probe membranes for potential interaction partners
This technique successfully identified a 60 kDa complex containing both PRDX1 and APE1
Bioluminescence Resonance Energy Transfer (BRET):
Methodology:
Generate fusion proteins of PRDX1 and potential partners with appropriate BRET donor/acceptor tags
Measure interaction dynamics in live cells under various redox conditions
Quantify changes in energy transfer as indicator of protein association/dissociation
Research has shown that PRDX1 interactions can be significantly influenced by the cellular redox state. For instance, treatment with (E)-3-[2-(5,6-dimethoxy-3-methyl-1,4-benzoquinonyl)] affects the stability of PRDX1-containing complexes . Additionally, PRDX1's ability to form disulfide-linked dimers and higher-order oligomers under oxidative stress conditions suggests that its interactome likely changes dramatically depending on the redox environment .
Researchers frequently encounter specific challenges when working with PRDX1 antibodies:
Multiple Banding Patterns:
Challenge: Detection of additional bands beyond the expected 21-23 kDa PRDX1 monomer.
Solution:
Variable Nuclear vs. Cytoplasmic Detection:
Challenge: Inconsistent detection of PRDX1 in nuclear fractions.
Solution:
Use gentle cell lysis procedures to avoid nuclear leakage
Employ thiol-blocking agents during fractionation to preserve protein interactions
Consider that PRDX1 nuclear localization may depend on interaction partners like APE1
Use properly validated antibodies known to work in immunofluorescence applications
Cross-Reactivity with Other Peroxiredoxin Family Members:
Challenge: Potential recognition of other PRDX isoforms due to sequence similarity.
Solution:
Fixation-Sensitive Epitopes in Immunohistochemistry/Immunofluorescence:
Challenge: Loss of antibody reactivity with certain fixation protocols.
Solution:
Researchers should note that several commercial PRDX1 antibodies are available with different properties. Some are produced against synthetic peptides corresponding to specific regions of human PRDX1, while others use recombinant proteins as immunogens . For critical applications, it may be worthwhile to compare multiple antibodies to identify those that perform optimally for the specific experimental system and application.
Distinguishing between PRDX1 oxidation states requires specialized methods:
Non-Reducing SDS-PAGE Coupled with Western Blotting:
Methodology:
Prepare lysates in the presence of alkylating agents (e.g., N-ethylmaleimide) to block free thiols
Perform SDS-PAGE without reducing agents
Blot and probe with PRDX1 antibodies
Reduced monomeric PRDX1 appears at ~22 kDa, while disulfide-linked dimers appear at ~44 kDa
Two-Dimensional Redox Western Blotting:
Methodology:
Run non-reducing SDS-PAGE in the first dimension
Cut the lane and expose to reducing conditions
Run the reduced sample in a second dimension SDS-PAGE
Blot and probe with PRDX1 antibodies
This technique separates different oxidation states into a diagonal pattern
Modification-Specific Immunoprecipitation:
Methodology:
Treat cells with dimedone to trap sulfenic acid forms of PRDX1
Perform immunoprecipitation with anti-dimedone antibodies
Probe western blots with PRDX1 antibodies to quantify sulfenic acid-modified PRDX1
Alternatively, immunoprecipitate with PRDX1 antibodies and probe for dimedone adducts
Conformation-Specific Antibodies:
While not mentioned in the search results, developing antibodies specific to particular oxidation states of PRDX1 would be valuable
In the interim, researchers can use general approaches to detect PRDX1 hyperoxidation by probing for sulfinic/sulfonic acid forms using anti-PrxSO₂/₃ antibodies
When analyzing PRDX1 oxidation states, it's important to consider that oxidative stress can cause PRDX1 to form high-molecular-weight oligomers that function as molecular chaperones rather than peroxidases . Additionally, under physiological conditions, PRDX1 forms complexes with other proteins such as APE1, which can be preserved using thiol-blocking agents during sample preparation . These interactions may influence the detection and interpretation of different PRDX1 oxidation states.
Several frontier research areas could leverage PRDX1 monoclonal antibodies:
Cancer Immunotherapy Optimization:
PRDX1 antibodies could help identify patients likely to benefit from NK cell-based therapies by assessing PRDX1 levels in tumor infiltrating lymphocytes
Monitoring PRDX1 expression in therapeutic cell products could serve as a quality control parameter predicting post-infusion efficacy
Target validation studies for approaches combining antioxidants with immunotherapy could use PRDX1 antibodies to track cellular redox responses
Neurodegenerative Disease Research:
PRDX1 binds to and regulates JNK and c-Abl kinases, which are implicated in neurodegeneration
PRDX1 antibodies could help map expression changes in different neurodegenerative conditions
The role of oxidative stress in neurodegeneration could be further elucidated by studying PRDX1-protein interactions in brain tissues
PTEN-Dependent Cancer Biology:
Redox Signalosome Mapping:
Emerging evidence indicates PRDX1 participates in multiple protein-protein interactions influenced by redox state
PRDX1 antibodies combined with proximity labeling approaches could identify novel components of redox-regulated signaling hubs
Sequential immunoprecipitation with PRDX1 antibodies followed by additional purification steps could isolate intact redox signaling complexes
Cellular Senescence Mechanisms:
Oxidative stress is a key driver of cellular senescence
PRDX1 antibodies could track changes in expression and localization during senescence progression
Comparisons between senescent and non-senescent cells could reveal altered PRDX1 interactomes
The expanding understanding of PRDX1's multifaceted roles—beyond simple peroxide detoxification to include chaperone functions, protein-protein interactions, and signaling regulation—opens numerous research avenues where specific antibodies will be invaluable tools for mechanistic studies, biomarker development, and therapeutic target validation .