PRX Antibody

Shipped with Ice Packs
In Stock

Description

What is PRX Antibody?

The PRX Antibody is a specific reagent designed to detect and study Peroxiredoxins (Prx), a family of antioxidant enzymes critical for cellular protection against oxidative damage. These enzymes neutralize reactive oxygen species (ROS) such as hydrogen peroxide and peroxynitrite, playing roles in redox signaling, inflammation modulation, and immune responses . The antibody is widely used in biochemical assays, including Western blotting (WB), immunoprecipitation (IP), and enzyme-linked immunosorbent assay (ELISA), to analyze Prx protein levels and localization in tissues .

Types of PRX Antibodies

PRX Antibody (B-11)

  • Catalog Number: sc-137222 (Santa Cruz Biotechnology)

  • Format: Mouse monoclonal IgG1 κ antibody

  • Reactivity: Detects human, mouse, and rat Prx proteins

  • Applications: WB, IP, IF, IHC(P), ELISA

  • Conjugates: Available in agarose, HRP, FITC, PE, and Alexa Fluor variants .

Rabbit Polyclonal Anti-PRX Antibody

  • Catalog Number: HPA001868 (Atlas Antibodies)

  • Format: Polyclonal antibody

  • Reactivity: Human Prx proteins

  • Applications: IHC, ICC-IF, WB

  • Validation: Tested for specificity and reproducibility in human tissues .

Applications in Research

a. Antioxidant Mechanisms
The antibody facilitates studies of Prx isoforms (e.g., Prx1, Prx2) in oxidative stress contexts, such as cancer progression and neurodegenerative diseases . For instance, Prx1 overexpression was linked to breast cancer progression .

b. Immunomodulation
Prx proteins interact with Toll-like receptor 4 (TLR4), modulating inflammatory responses. The antibody aids in mapping these interactions in immune cells .

c. Disease Biomarkers
Prx antibodies are used to measure protein levels in Alzheimer’s disease and Down syndrome, where Prx1/2 are upregulated .

d. Drug Development
In Fabry disease, the antibody supports studies of PRX-102 (pegunigalsidase-alfa), a PEGylated enzyme replacement therapy. It helps assess anti-drug antibodies (ADAs) and serum half-life .

Research Findings

  • Anti-PEG Antibodies: Pre-existing anti-PEG antibodies in Fabry patients marginally affect PRX-102 efficacy due to low titers .

  • Prx in Fish Immunity: Fish Prx1/2 enhance natural killer cell activity and protect against viral infections .

  • Cancer Therapy: Prx6 overexpression correlates with poor prognosis in certain tumors, suggesting therapeutic targeting potential .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery time may vary depending on the purchasing method or location. Please contact your local distributor for specific delivery times.
Synonyms
CMT4F antibody; KIAA1620 antibody; Periaxin antibody; PRAX_HUMAN antibody; Prx antibody
Target Names
PRX
Uniprot No.

Target Background

Function
Periaxin is a scaffolding protein that plays a crucial role in the formation and maintenance of myelin sheaths, essential for efficient nerve impulse transmission. It participates in the dystroglycan complex in Schwann cells and interacts with EZR and AHNAK-containing complexes in eye lens fiber cells. Periaxin's functions are multifaceted and critical for the following:

  • Peripheral Myelin Sheath Maintenance: Periaxin is essential for maintaining the structural integrity of the peripheral myelin sheath, which is critical for the rapid and efficient transmission of nerve impulses and proper sensory perception.
  • Myelin Sheath Transport: Periaxin is involved in the proper transport of Myelin Basic Protein (MBP) mRNA from the nucleus to the paranodal regions of the myelin sheath, contributing to efficient myelination.
  • Remyelination After Nerve Injury: Periaxin plays a vital role in the remyelination process after nerve injury, facilitating the repair and restoration of myelin sheaths.
  • Schwann Cell Elongation and Internode Length: Periaxin is critical for the normal elongation of Schwann cells and the proper length of internodes between the nodes of Ranvier, ensuring optimal nerve impulse transmission.
  • Myelin Sheath Apposition Formation: Periaxin mediates the formation of appositions between the abaxonal surface of the myelin sheath and the Schwann cell plasma membrane, effectively compartmentalizing the Schwann cell cytoplasm.
  • Cajal Bands and Schmidt-Lanterman Incisures: Periaxin contributes to the formation of Cajal bands and Schmidt-Lanterman incisures, specialized structures on myelinated nerves.
  • DRP2 Recruitment: Periaxin recruits Dynamin-related protein 2 (DRP2) to the Schwann cell plasma membrane, a process important for maintaining myelin sheath integrity.
  • Eye Lens Fiber Cell Morphology: Periaxin is essential for the proper protein composition and morphology of eye lens fiber cell plasma membranes, contributing to normal eye lens function.
Gene References Into Functions

The critical role of periaxin in maintaining peripheral nerve health is highlighted by studies associating mutations in the PRX gene with various neurological disorders, including:

  1. Congenital Cataract: A variant of periaxin was identified in a Chinese family with congenital cataract, emphasizing its role in eye lens development. (PMID: 27081207)
  2. Periaxin Complex Formation and Stability: Research has shed light on the mechanisms by which periaxin complexes form and maintain their stability, linking the extracellular matrix to the cytoskeleton. (PMID: 26940996)
  3. Dejerine-Sottas Syndrome and Peripheral Neuropathy: A causative mutation (p.R1070*) in the PRX gene has been linked to Dejerine-Sottas syndrome, a severe form of peripheral neuropathy. (PMID: 26059842)
  4. Demyelinating Neuropathy and Scoliosis: Genetic analysis of the periaxin gene is recommended in patients with severe demyelinating neuropathy accompanied by early infantile scoliosis. (PMID: 24011642)
  5. Charcot-Marie-Tooth Disease (CMT4F): Variations in clinical phenotypes for CMT4F have been linked to novel, nonsense PRX mutations, emphasizing the gene's role in this debilitating disorder. (PMID: 22847150)
  6. Unusual Charcot-Marie-Tooth Disease Phenotypes: Novel mutations in both the PRX and MTMR2 genes have been implicated in unusual Charcot-Marie-Tooth disease phenotypes. (PMID: 21741241)
  7. Dejerine-Sottas Neuropathy: Research has established the role of periaxin in maintaining peripheral nerve myelin, with mutations in the PRX gene causing autosomal recessive demyelinating Charcot-Marie-Tooth disease or Dejerine-Sottas neuropathy. (PMID: 21079185)
  8. Vocal Cord Paralysis: A novel mutation in the PRX gene has been identified in cases of vocal cord paralysis, highlighting its potential role in this condition. (PMID: 19950375)
  9. Sensory Neuropathy: Research suggests a prominent role of periaxin gene mutations in causing sensory neuropathy and emphasizes the significance of the carboxyl terminal domain of the periaxin protein. (PMID: 12112076)
  10. Slow-Progressive Charcot-Marie-Tooth Disease: Periaxin mutations have been linked to early-onset, yet slow-progressive Charcot-Marie-Tooth disease, indicating its impact on disease progression. (PMID: 15197604)
  11. Early-Onset Charcot-Marie-Tooth Disease (CMT4F): A specific mutation (S399fsX410) in the PRX gene has been identified in an 8-year-old patient with early-onset CMT4F disease, demonstrating the gene's involvement in this form of CMT. (PMID: 16534116)
  12. PRX Mutation in Japanese Patients: A PRX mutation has been reported in Japanese patients, highlighting the global relevance of the gene's association with neurological disorders. (PMID: 16770524)
  13. Early-Onset Demyelinating Charcot-Marie-Tooth Disease and Sensory Loss: Studies have linked mutations in the PRX gene to early-onset demyelinating autosomal recessive Charcot-Marie-Tooth disease and severe sensory loss. (PMID: 18504680)
Database Links

HGNC: 13797

OMIM: 145900

KEGG: hsa:57716

STRING: 9606.ENSP00000326018

UniGene: Hs.205457

Involvement In Disease
Dejerine-Sottas syndrome (DSS); Charcot-Marie-Tooth disease 4F (CMT4F)
Protein Families
Periaxin family
Subcellular Location
[Isoform 1]: Cell membrane; Peripheral membrane protein; Cytoplasmic side. Nucleus. Cytoplasm.; [Isoform 2]: Cytoplasm.; Cell membrane. Cell junction.
Tissue Specificity
Detected in spinal cord. Isoform 1 and isoform 2 are found in sciatic nerve and Schwann cells.

Q&A

What is the difference between PRX as periaxin and PRX as peroxiredoxin in antibody applications?

PRX antibodies target two distinct proteins that share the same abbreviation but differ significantly in function and structure. Peroxiredoxin (PRX) comprises a family of six antioxidant proteins (PRX I through VI) with molecular weights ranging from 22-25 kDa that protect cells from reactive oxygen species by preventing metal-catalyzed oxidation of enzymes . In contrast, periaxin (PRX) is a significantly larger protein (154.9 kDa) encoded by the PRX gene, also known as CMT4F . When selecting a PRX antibody, researchers must verify which specific protein target the antibody recognizes, as antibodies against these different targets are not interchangeable. Cross-validation with target-specific antibodies and molecular weight confirmation via Western blot is essential to prevent experimental misinterpretation.

How should researchers select the appropriate PRX antibody for their specific experimental applications?

Selection of an appropriate PRX antibody requires consideration of several experimental parameters. First, determine which PRX isoform (for peroxiredoxins) or protein region (for periaxin) is relevant to your research question. For peroxiredoxins, identify whether you need isoform-specific detection (PRX I-VI) or pan-PRX recognition . Second, match the application requirements (Western blot, IHC, IF, ELISA, or IP) with antibodies validated for those specific applications . Third, consider species reactivity—ensure the antibody recognizes your species of interest, as PRX antibodies may vary in cross-reactivity between human, mouse, and rat samples . Fourth, determine whether you require specific redox-state detection (e.g., antibodies that specifically recognize hyperoxidized PRX-SO₂/₃) . Finally, evaluate clone type (monoclonal vs. polyclonal) based on specificity requirements and whether conjugated antibodies (HRP, PE, FITC, Alexa Fluor) would benefit your experimental workflow .

What are the optimal protein extraction methods for preserving native PRX redox states?

Preserving the native redox state of PRX proteins requires specific extraction protocols to prevent artificial oxidation during sample processing. For accurate redox state analysis, use a non-reducing lysis buffer (typically containing 50 mM Tris pH 8.0, 150 mM NaCl, 0.1% SDS, and 0.5% NaDeoxylate) supplemented with protease inhibitor cocktail . To trap PRXs in their native redox state and prevent artifactual over-oxidation during extraction, add 100 mM N-ethyl maleimide (NEM) to the lysis buffer . This alkylating agent irreversibly modifies free thiol groups, preserving the redox state at the time of cell lysis. For comparative studies of different redox forms, prepare parallel samples using both reducing conditions (with T-PER tissue protein extraction reagent containing standard protease inhibitors) and non-reducing conditions to visualize the distribution between monomeric and dimeric forms . Sample processing should be performed quickly at 4°C to minimize spontaneous oxidation, and samples should be analyzed immediately or stored at -80°C with minimal freeze-thaw cycles.

What controls should be included when performing PRX immunodetection experiments?

Robust PRX immunodetection requires multiple controls to ensure specificity and reliability of results. First, include positive controls using tissues or cell lines known to express the target PRX protein (e.g., liver, kidney, testis, and lung for PRX I; mitochondria-rich tissues for PRX III and V) . Second, incorporate negative controls such as tissues from knockout models or cells where the target PRX expression is absent . Third, implement secondary antibody-only controls to identify potential non-specific binding, as demonstrated in immunohistochemistry protocols where "Secondary antibody only control: Secondary antibody is a ready to use LeicaDS9800 (Bond® Polymer Refine Detection)" . Fourth, for oxidation-specific studies, include both oxidized and reduced standards to validate the specificity of oxidation-state specific antibodies . Fifth, when studying multiple PRX isoforms, perform parallel blots with isoform-specific antibodies to distinguish between the similar molecular weight proteins, particularly for PRX 1-3 which are indistinguishable by molecular weight alone (approximately 22 kDa for monomers) . Finally, consider loading controls appropriate for the cellular compartment being studied (cytosolic or mitochondrial) to normalize protein expression levels.

How can researchers accurately distinguish between different PRX isoforms in complex biological samples?

Distinguishing between PRX isoforms presents a significant challenge due to their similar molecular weights and structural homology. For accurate isoform discrimination, implement a multi-faceted approach combining several techniques. First, employ isoform-specific antibodies validated for their specificity against individual PRX family members . Second, utilize subcellular fractionation to exploit the distinct localization patterns—PRX I, II, IV, and VI are predominantly cytoplasmic, while PRX III and V are mitochondrial . Third, perform sequential immunoblotting with stripping and reprobing using different PRX-specific antibodies on the same membrane to identify co-migrating and distinctive bands . Fourth, incorporate knockout or knockdown controls for each specific PRX isoform to unambiguously identify bands. Fifth, combine immunoblotting with mass spectrometry for definitive isoform identification in complex samples. Finally, use 2D electrophoresis (separating by both isoelectric point and molecular weight) to resolve isoforms with similar molecular weights but different post-translational modifications. This comprehensive approach enables reliable discrimination between the PRX isoforms despite their similarities in conventional 1D gel electrophoresis.

What methodological approaches can detect the diverse redox states of PRX proteins in experimental systems?

Detecting diverse redox states of PRX proteins requires specialized methodological approaches that preserve and distinguish between reduced, oxidized, and hyperoxidized forms. First, implement redox state-specific protein extraction by using N-ethyl maleimide (NEM) at 100 mM concentration to alkylate free thiols and trap PRXs in their native redox state . Second, perform non-reducing SDS-PAGE to distinguish between monomeric and dimeric forms, which represent different oxidation states of PRX proteins . Third, utilize redox state-specific antibodies, particularly anti-Prx-SO₂/₃ antibodies that specifically recognize hyperoxidized peroxiredoxins . Fourth, employ a diagonal redox 2D-PAGE approach, where proteins are separated under non-reducing conditions in the first dimension and reducing conditions in the second dimension, creating a pattern where proteins with different redox states appear off the diagonal. Fifth, combine these techniques with high-resolution mass spectrometry to identify specific oxidized residues and quantify the proportion of each redox form. Finally, validate findings using recombinant PRX proteins subjected to controlled oxidation conditions as standards. This multi-technique approach provides comprehensive characterization of PRX redox states in complex biological samples.

How can researchers quantitatively assess PRX hyperoxidation in relation to oxidative stress responses?

Quantitative assessment of PRX hyperoxidation requires a systematic approach combining multiple analytical techniques. Begin with immunoblot analysis using antibodies specifically targeting the hyperoxidized form (PRX-SO₂/₃), alongside antibodies recognizing total PRX protein . Calculate the ratio of hyperoxidized to total PRX to normalize for expression level variations. For temporal dynamics, perform time-course experiments with oxidative stress inducers (H₂O₂, paraquat, or hypoxia/reoxygenation) at physiologically relevant concentrations. Implement mass spectrometry-based approaches, particularly multiple reaction monitoring (MRM), to quantify specific hyperoxidized peptides with precision. For subcellular resolution, combine immunofluorescence using hyperoxidation-specific antibodies with confocal microscopy and quantitative image analysis. In cell populations, flow cytometry with permeabilized cells can quantify the percentage of cells containing hyperoxidized PRX. For in vivo studies, develop transgenic reporter systems where fluorescent proteins are coupled to hyperoxidation-responsive elements. Statistical analysis should include multivariate approaches correlating hyperoxidation levels with other oxidative stress markers (e.g., glutathione depletion, lipid peroxidation) to establish the relationship between PRX hyperoxidation and cellular oxidative damage parameters.

What are the key considerations when designing experiments to study PRX dysfunction in disease models?

Designing experiments to study PRX dysfunction in disease models requires careful consideration of multiple factors for robust, translatable results. First, select appropriate disease models that recapitulate key pathophysiological features—consider both genetic models (knockout/knockin) and induced models (chemical/environmental stressors) . Second, implement tissue-specific analyses, as PRX expression and function vary significantly across tissues (e.g., PRX I in liver, kidney, testis, lung, nervous system; PRX V short form in liver, kidney, heart, and lung) . Third, establish temporal frameworks to distinguish between acute responses and chronic adaptations to PRX dysfunction, particularly in progressive diseases like Alzheimer's disease where PRX I and II levels are elevated . Fourth, incorporate both redox-dependent and redox-independent functions of PRX in experimental designs, as these proteins participate in multiple cellular processes beyond antioxidant defense, including proliferation, differentiation, and gene expression . Fifth, employ multi-omics approaches (transcriptomics, proteomics, metabolomics) to capture system-wide effects of PRX dysfunction. Sixth, validate findings across multiple model systems, including cell lines, primary cultures, animal models, and when possible, human samples. Finally, design rescue experiments using recombinant PRX proteins or gene therapy approaches to establish causality between PRX dysfunction and observed disease phenotypes.

How should researchers address non-specific binding issues when using PRX antibodies?

Non-specific binding with PRX antibodies presents a common technical challenge requiring systematic troubleshooting. First, implement more stringent blocking protocols using 5% BSA or 5% milk in TBS-T with extended blocking times (2 hours at room temperature or overnight at 4°C). Second, optimize antibody concentrations through titration experiments—for immunohistochemistry applications, dilutions ranging from 1/100 (4.8 μg/ml) to 1/2000 (0.24 μg/ml) have been reported as effective . Third, increase washing stringency with higher salt concentrations (up to 500 mM NaCl) in wash buffers and extend washing times. Fourth, pre-absorb antibodies with tissues or lysates from knockout models or unrelated tissues to remove cross-reactive antibodies. Fifth, when possible, validate observations with multiple antibodies targeting different epitopes of the same PRX protein. Sixth, perform parallel experiments with secondary antibody-only controls to identify background signal, as demonstrated in immunohistochemistry protocols . Finally, confirm specificity through peptide competition assays, where pre-incubation of the antibody with excess immunizing peptide should abolish specific signals. For PRX family members with high homology, cross-reactivity between isoforms should be systematically evaluated using recombinant proteins or tissues from isoform-specific knockout models.

What strategies can resolve contradictory results between different detection methods for PRX proteins?

Resolving contradictory results between different PRX detection methods requires systematic investigation of methodological differences. First, compare sample preparation protocols—differences in buffer composition, particularly reducing versus non-reducing conditions, significantly impact PRX detection, as demonstrated in immunoblot analyses . Second, evaluate antibody epitope recognition—antibodies targeting different epitopes may yield discrepant results if post-translational modifications or protein interactions mask specific epitopes. Third, consider detection sensitivity thresholds—techniques vary in sensitivity (mass spectrometry > Western blot > immunohistochemistry), potentially explaining negative results in less sensitive methods. Fourth, assess species-specific differences in antibody recognition, even when sequence homology suggests cross-reactivity. Fifth, implement orthogonal validation approaches—when immunological methods yield contradicting results, employ non-antibody-dependent techniques such as mass spectrometry, RNA-seq, or activity assays. Sixth, evaluate subcellular localization effects—contradictions may arise from differential extraction efficiency of compartmentalized PRX proteins, particularly for mitochondrial PRX III and V versus cytoplasmic PRX I, II, IV, and VI . Finally, establish a systematic validation pipeline where samples are analyzed in parallel using multiple techniques with appropriate controls, allowing direct comparison under identical experimental conditions to identify the source of discrepancies.

How can researchers optimize immunohistochemistry protocols for detecting PRX in different tissue types?

Optimizing immunohistochemistry protocols for PRX detection across diverse tissues requires systematic adjustment of multiple parameters. Begin with antigen retrieval optimization—for PRX detection, heat-mediated antigen retrieval with either Tris-EDTA buffer (pH 9.0) or sodium citrate buffer (10mM citrate pH 6.0 + 0.05% Tween-20) has proven effective . For tissues with high endogenous peroxidase activity (e.g., liver, kidney), incorporate extended quenching steps (3% H₂O₂ for 15-30 minutes). Fixation protocols significantly impact epitope preservation—compare 4% PFA-fixed frozen sections with paraffin-embedded tissues for optimal signal-to-noise ratio . Antibody concentration requires tissue-specific optimization—effective dilutions range from 1/100 (4.8 μg/ml) for frozen sections to 1/2000 (0.24 μg/ml) for paraffin sections . Incubation times should be systematically tested, with 30 minutes at room temperature established as effective for paraffin sections . For tissues with high background (e.g., brain, muscle), implement additional blocking steps with avidin/biotin blocking solutions or mouse-on-mouse blocking reagents for mouse tissues. Signal amplification systems should be selected based on tissue type and expected expression levels—standard polymer detection systems (e.g., LeicaDS9800 Bond® Polymer Refine Detection) have shown efficacy across multiple tissue types . Finally, implement tissue-specific positive and negative controls—for example, capillaries in lung tissue provide positive controls for certain PRX antibodies, while mouse cerebrum may serve as a negative control .

What approaches can accurately distinguish between PRX hyperoxidation and other post-translational modifications?

Distinguishing PRX hyperoxidation from other post-translational modifications (PTMs) requires a multi-faceted analytical approach. First, employ oxidation state-specific antibodies that selectively recognize the sulfinic (SO₂) or sulfonic (SO₃) acid forms of peroxiredoxins, which represent hyperoxidized states . Second, implement differential alkylation protocols—first blocking reduced thiols with NEM followed by reduction of reversibly oxidized thiols and subsequent labeling with a different alkylating agent allows discrimination between reversible and irreversible oxidation states . Third, utilize redox proteomics approaches combining differential labeling with mass spectrometry to identify specific oxidized residues and distinguish between hyperoxidation (sulfinic/sulfonic acid formation) and other oxidative PTMs (S-glutathionylation, S-nitrosylation). Fourth, perform site-directed mutagenesis of catalytic cysteines to confirm specificity of detected signals. Fifth, implement chemical reduction tests—hyperoxidized PRXs (Prx-SO₂/₃) are resistant to reduction by standard reductants like DTT or β-mercaptoethanol, while most other oxidative PTMs are reversible under these conditions. Sixth, employ specific enzymatic assays—sulfiredoxin specifically reduces sulfinic acid in PRXs but not sulfonic acid or other PTMs. Finally, combine these approaches with structural analysis techniques like circular dichroism or limited proteolysis to detect conformational changes specific to hyperoxidation versus other modifications, providing a comprehensive characterization of PRX post-translational status.

How should researchers interpret discrepancies between PRX monomeric and dimeric forms in experimental results?

Interpreting discrepancies between PRX monomeric and dimeric forms requires understanding the relationship between PRX structure and redox state. The distribution between monomeric (~22 kDa for PRX1-3, ~25 kDa for PRX4) and dimeric forms (~44 kDa for PRX1-3, ~50 kDa for PRX4) reflects the functional redox cycle of these proteins . Under non-reducing conditions, reduced (active) PRXs typically appear as monomers, while oxidized PRXs form disulfide-linked dimers . First, confirm whether samples were processed under reducing or non-reducing conditions, as reducing agents will disrupt disulfide bonds and convert dimers to monomers. Second, evaluate whether hyperoxidation has occurred—hyperoxidized PRXs (Prx-SO₂/₃) cannot form disulfide bonds and remain monomeric even under non-reducing conditions . Third, assess whether sample preparation preserved native redox states through the use of alkylating agents like NEM . Fourth, consider PRX isoform-specific behaviors—different isoforms may show distinct monomer-dimer distributions under identical conditions. Fifth, evaluate experimental conditions that might alter PRX redox states (oxidative stress, hypoxia, enzyme inactivation). Sixth, determine whether post-translational modifications beyond oxidation (phosphorylation, acetylation) impact dimerization. Finally, recognize that non-specific bands may confound interpretation—for example, the ~50 kDa band recognized by SO₂/₃ antibodies corresponding to PRX4 dimer has been observed in mutants and overexpressors, suggesting non-specific binding . Careful analysis combining multiple antibodies, redox conditions, and controls is essential for accurate interpretation of monomer-dimer distributions.

How can researchers correlate PRX expression patterns with functional outcomes in tissues and cells?

Correlating PRX expression patterns with functional outcomes requires a comprehensive approach integrating multiple experimental techniques. First, establish baseline expression profiles across tissues and cell types using a combination of immunoblotting, immunohistochemistry, and qPCR to document isoform-specific distribution patterns . Second, implement loss-of-function approaches (siRNA, CRISPR-Cas9, or pharmacological inhibitors) alongside gain-of-function strategies (overexpression systems) to establish causality between PRX levels and functional parameters. Third, measure functional readouts specific to PRX roles—for antioxidant function, quantify ROS levels, oxidative damage markers, and cell survival under oxidative stress; for signaling functions, assess relevant pathway activation (proliferation, differentiation, gene expression) . Fourth, perform subcellular colocalization studies to correlate PRX distribution with intracellular oxidative stress using compartment-specific ROS sensors. Fifth, develop temporal correlations through time-course experiments capturing both acute responses and adaptive changes following PRX modulation. Sixth, implement tissue-specific conditional knockout models to address the varying roles of PRX across tissues—for example, nervous system versus epithelial tissues . Finally, translate findings to disease contexts relevant to specific PRX isoforms—for example, correlating PRX I and II levels with pathological markers in Alzheimer's disease or Down syndrome, or PRX I expression with breast cancer progression . This integrated approach enables robust correlation between expression patterns and functional consequences across physiological and pathological contexts.

What computational approaches can enhance analysis of PRX localization and interaction data?

Computational approaches significantly enhance the analysis of PRX localization and interaction data through multiple advanced techniques. For localization studies, implement machine learning-based image analysis algorithms to quantify immunofluorescence or immunohistochemistry data across tissue sections, providing unbiased quantification of PRX distribution patterns . Develop subcellular prediction algorithms integrating primary sequence information, known targeting motifs, and experimental data to predict novel localization patterns—particularly relevant for distinguishing between mitochondrial PRX III and V versus cytoplasmic PRX I, II, IV, and VI . For interaction analysis, employ molecular docking simulations to predict protein-protein interactions between PRX isoforms and potential partners, generating testable hypotheses for experimental validation. Implement network analysis approaches integrating proteomics data to identify PRX-centered interaction networks and pathway enrichment. Apply statistical colocalization analysis to microscopy data, calculating Pearson's or Manders' coefficients to quantify spatial correlation between PRX and potential interacting proteins or cellular structures. For temporal dynamics, develop computational models simulating PRX redox cycling under various cellular conditions, predicting system behavior under physiological and pathological states. Finally, integrate multi-omics data (genomics, transcriptomics, proteomics, metabolomics) through systems biology approaches to position PRX within broader cellular networks, identifying emergent properties not evident from single-technique studies.

How should differences in PRX tissue expression patterns influence experimental design and data interpretation?

The differential tissue expression patterns of PRX isoforms should fundamentally shape experimental design and data interpretation in several critical ways. First, select appropriate experimental models based on tissue-specific expression patterns—PRX I is widely expressed across liver, kidney, testis, lung, and nervous system; PRX III and V are mitochondria-enriched; PRX V has distinct long and short forms with the long form predominantly in testis and the short form in liver, kidney, heart, and lung . Second, implement tissue-specific normalization strategies—compare PRX expression only within the same tissue type or against appropriate housekeeping proteins specific to that tissue. Third, design isoform-specific detection strategies accounting for tissue-dependent PRX profiles—for example, antibody panels targeting PRX I, II, and III would be appropriate for nervous system studies where these isoforms show elevated expression in pathological conditions like Alzheimer's disease and Down syndrome . Fourth, incorporate tissue-specific positive controls in immunodetection experiments—lung capillaries for certain PRX antibodies, nerve fibers in skeletal muscle for others . Fifth, consider tissue-specific post-translational modification patterns that may affect antibody recognition—palmitoylation of PRX V may affect epitope accessibility in certain tissues . Sixth, account for pathology-induced alterations in PRX expression—upregulation of specific isoforms in cancer or neurodegenerative conditions may create expression patterns divergent from normal tissues . Finally, develop tissue-specific interpretation frameworks recognizing that the functional significance of PRX alterations varies across tissues—changes in mitochondria-rich tissues likely reflect different biological processes than similar changes in epithelial tissues.

How can PRX antibodies be utilized in the study of neurodegenerative diseases?

PRX antibodies offer significant applications in neurodegenerative disease research based on the established involvement of peroxiredoxins in these conditions. First, implement quantitative immunohistochemistry with isoform-specific PRX antibodies to map expression changes across brain regions in Alzheimer's disease and Down syndrome, where elevated levels of PRX I and II have been documented . Second, develop multiplex immunofluorescence protocols combining PRX antibodies with markers of oxidative damage, protein aggregation, and cellular stress responses to establish spatial relationships between PRX expression and pathological features. Third, utilize PRX hyperoxidation-specific antibodies (anti-Prx-SO₂/₃) to quantify redox dysregulation in affected versus spared brain regions, providing insights into differential vulnerability . Fourth, employ PRX antibodies in cerebrospinal fluid biomarker studies to determine whether PRX release correlates with disease progression. Fifth, investigate the relationship between PRX expression and blood-brain barrier function using vascular co-staining, as PRX antibodies show specific labeling of brain capillaries . Sixth, examine periaxin (PRX) antibodies in peripheral nerve studies of neurodegenerative conditions with peripheral involvement, as periaxin plays critical roles in myelin stability . Finally, implement longitudinal studies in animal models to track PRX changes from presymptomatic to advanced disease stages, correlating oxidative stress biomarkers with behavioral and pathological progression. These applications collectively provide a comprehensive framework for understanding redox dysregulation in neurodegenerative pathophysiology.

What methodological approaches can address contradictory findings regarding PRX roles in cancer biology?

Addressing contradictory findings regarding PRX roles in cancer biology requires systematic methodological approaches addressing multiple aspects of experimental design and data interpretation. First, implement comprehensive isoform profiling across diverse cancer types, patient samples, and matched normal tissues using validated isoform-specific antibodies to establish cancer-specific PRX signatures . Second, distinguish between expression levels and functional activity through combined approaches measuring both protein abundance (immunoblotting, IHC) and enzymatic activity (peroxidase assays, redox state analysis). Third, consider intratumoral heterogeneity by employing single-cell approaches and spatial transcriptomics/proteomics to identify cell type-specific PRX functions within the tumor microenvironment. Fourth, develop temporal studies across cancer progression stages, from precancerous lesions to metastatic disease, establishing when and how PRX functions evolve during tumorigenesis. Fifth, implement isogenic cell line models with controlled genetic backgrounds to isolate PRX effects from confounding variables. Sixth, address context-dependent functions through systematic variation of experimental conditions (normoxia vs. hypoxia, presence vs. absence of treatment stressors) to identify condition-specific roles. Seventh, combine PRX modulation with pathway inhibitors to map the signaling networks through which PRXs influence cancer phenotypes beyond their antioxidant functions. Finally, implement meta-analysis approaches integrating published datasets with standardized interpretation frameworks to identify sources of discrepancies across studies. This comprehensive methodological roadmap enables resolution of apparent contradictions by revealing context-specific and multifunctional aspects of PRX biology in cancer.

How should researchers integrate PRX antibody data with other redox biomarkers for comprehensive oxidative stress assessment?

Integrating PRX antibody data with other redox biomarkers requires a systematic multiparametric approach to comprehensively assess oxidative stress. First, develop multiplexed detection systems combining PRX hyperoxidation measurements (using anti-Prx-SO₂/₃ antibodies) with other protein oxidation markers (carbonylation, nitrotyrosine formation) to establish correlation patterns between different oxidative modifications . Second, implement parallel assessment of small molecule antioxidants (glutathione, vitamin E, vitamin C) alongside PRX status to understand compensatory relationships between enzymatic and non-enzymatic antioxidant systems. Third, correlate PRX redox state with activity measurements of related enzymes (thioredoxin, thioredoxin reductase, sulfiredoxin) to establish the functional status of the entire redox network. Fourth, implement multiparametric flow cytometry or imaging cytometry to simultaneously measure multiple redox parameters at the single-cell level, revealing population heterogeneity in oxidative stress responses. Fifth, develop integrated redox panels combining PRX measurements with lipid peroxidation markers (4-HNE, MDA), DNA oxidation products (8-OHdG), and mitochondrial function indicators to capture compartment-specific oxidative damage. Sixth, employ principal component analysis and other dimensionality reduction techniques to identify which redox parameters best discriminate between experimental conditions or disease states. Finally, establish standardized reporting frameworks for integrated redox assessment, ensuring consistent methodology and facilitating meta-analysis across studies. This comprehensive integration approach provides deeper insights into oxidative stress dynamics than any single biomarker alone, revealing system-level redox dysregulation patterns in complex biological processes.

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.