Prevents aberrant sulfenylation/sulfination of nascent proteins by coupling H₂O₂ elimination with disulfide bond formation
Maintains ER redox balance, critical for preventing misfolded protein accumulation
Modulates receptor tyrosine kinase (RTK) signaling by regulating H₂O₂ levels that influence phosphatase activity
Impacts neurogenesis via GDE2 regulation in motor neuron differentiation
Ovarian aging: PRDX4 deficiency exacerbates oxidative DNA damage (8-OHdG↑) and granulosa cell apoptosis in mice
Metabolic disorders: Protects against hepatic steatosis and insulin resistance via systemic ROS reduction
Piperlongumine: Inactivates PRDX4 in glioblastoma, increasing ROS-mediated apoptosis (IC₅₀ reduction by 60%)
PRDX4 siRNA: Reduces lung tumor invasion (Matrigel penetration ↓35%, p<0.01) and colorectal liver metastasis
Prostate cancer: Serum PRDX4 levels correlate with bone metastasis (AUC=0.82)
Early-stage lung SCC: High PRDX4 predicts shorter DFS (HR=2.1, p=0.008)
PRDX4 (Peroxiredoxin 4) is a member of the peroxiredoxin family, which consists of six antioxidant isozymes (PRDX1-6). These redox family proteins are ubiquitously expressed and widely distributed across human tissues. PRDX4 is unique among the peroxiredoxins as it primarily resides in the endoplasmic reticulum (ER), containing a hydrophobic signal sequence at the N-terminus responsible for its ER localization or secretion into extracellular space .
While the other peroxiredoxins have different subcellular distributions (PRDX1, PRDX2, and PRDX6 in cytosol and nucleus; PRDX3 exclusively in mitochondria; PRDX5 in mitochondria, cytosol and peroxisomes), PRDX4 is the only ER-resident peroxiredoxin . Interestingly, recent research has identified a cytosolic isoform of PRDX4 that is transcribed from an alternative mRNA splicing uniquely in sexually matured testes .
PRDX4 shows variable expression across different human tissues. It is abundantly expressed in the heart, liver, pancreas, and muscle tissues. In contrast, PRDX4 expression is notably lower in the brain, spleen, and peripheral blood leukocytes . This tissue-specific expression pattern suggests specialized functions in different organ systems and potentially tissue-specific regulation mechanisms. Unlike other peroxiredoxins, PRDX4 is not expressed in unicellular organisms such as yeast, indicating its evolutionary development coincided with multicellularity and specialized tissue development .
The primary physiological function of PRDX4 centers on its role in redox homeostasis within the endoplasmic reticulum. PRDX4 catalyzes the reduction of hydrogen peroxide (H₂O₂) and thereby protects the ER lumen from oxidative damage. This activity serves several critical cellular functions:
Protein folding assistance: PRDX4 ensures the correct folding process of nascent proteins, preventing them from undergoing alternative oxidative pathways that could lead to misfolding .
Redox balance maintenance: It couples H₂O₂ catabolism with oxidative protein folding to maintain ER redox balance .
Signal transduction regulation: PRDX4 fine-tunes H₂O₂ concentration, which plays a critical role in cell proliferation signaling pathways .
Protection against misfolded protein accumulation: By maintaining proper redox conditions in the ER, PRDX4 helps prevent the development and accumulation of misfolded proteins that could trigger cellular stress responses .
PRDX4 functions within a complex network of redox shuttles and NADPH metabolism. While PRDX4 itself is involved in H₂O₂ catabolism in the ER, its function is interconnected with broader cellular redox systems. The dependency of certain cancer cells on PRDX4 correlates with cellular NADPH levels, suggesting a functional relationship between PRDX4 activity and NADPH metabolism .
Several redox shuttles contribute to maintaining cellular redox balance. For instance, the pyruvate-malate shuttle enables the production of NADPH through cytosolic malic enzyme (ME1), which can then feed NADPH oxidase 4 (NOX4). Similarly, the pyruvate-isocitrate shuttle allows for the formation of NADPH that can serve as substrate for NOX4 .
PRDX4 appears to play a counterbalancing role against NOX4-generated reactive oxygen species. NOX4, which localizes to the ER membrane, has been identified as a source of ROS that creates cellular dependency on PRDX4, particularly in pancreatic cancer cells. The functional requirement for PRDX4 correlates with cellular NADPH levels across different cancer models and can be rescued by depletion of NOX4 or NADPH . This relationship highlights PRDX4's role in maintaining redox homeostasis in the face of NOX4-generated oxidative stress.
The floodgate model describes a mechanism by which peroxiredoxins, including PRDX4, regulate the diffusion and signaling functions of H₂O₂. According to this model, when peroxiredoxins encounter sustained H₂O₂ flux, they can form high molecular weight (HMW) complexes. These HMW complexes effectively withdraw PRDX molecules from their catalytic cycle, preventing them from metabolizing H₂O₂ in their immediate vicinity .
This temporary inactivation of peroxiredoxins allows H₂O₂ to diffuse to more distant cellular locations, where it can oxidize and thereby activate or inactivate target proteins involved in signaling pathways. The formation of these HMW complexes thus acts as a "floodgate" that permits H₂O₂-mediated redox signaling to occur at specific locations .
The peroxiredoxins containing sulfinyls (oxidized forms) can be returned to their active state through the action of sulfiredoxin (SRX) systems. In mitochondria, this process has been linked to circadian rhythms, with SRX expression alternating with SRX degradation by LON-protease, controlled by clock genes in various tissues including the adrenal gland, brown adipose tissue, and heart .
PRDX4 depletion has significant effects on DNA integrity and repair mechanisms, particularly in cancer cells dependent on PRDX4 for survival. When PRDX4 is depleted in these cells, several consequential events occur:
Increased ROS levels: PRDX4 knockdown results in elevated reactive oxygen species, which can directly damage DNA .
DNA damage induction: Both single-strand breaks (SSBs) and double-strand breaks (DSBs) increase following PRDX4 depletion, as confirmed by Comet assays under alkaline and neutral conditions, respectively .
DNA damage response activation: PRDX4 depletion leads to phosphorylation of histone H2AX at Ser139 (γH2AX), a marker of DNA double-strand breaks. This produces a focal γH2AX signal important for regulation of DNA repair .
DNA-PKcs dependency: The DNA damage response triggered by PRDX4 depletion is primarily governed by DNA-dependent protein kinase catalytic subunit (DNA-PKcs), rather than other phosphoinositide 3-kinase-related kinase (PIKK) family members. This is evidenced by the phosphorylation of RPA32 at the DNA-PK-dependent S4/S8 residue, but not at the ATM/ATR-dependent S33 residue .
Cell cycle effects: PRDX4 knockdown results in a higher proportion of cells in G2/M phase of the cell cycle, but only in cell lines in which PRDX4 is essential, consistent with a DNA damage-induced checkpoint activation .
These findings suggest that PRDX4 plays a crucial role in protecting cellular DNA from oxidative damage, particularly in cancer cells with elevated ROS production.
When investigating PRDX4 dependency in cancer cells, researchers should consider the following methodological approaches:
Genetic manipulation techniques:
Proliferation and survival assays:
ROS measurement:
DNA damage assessment:
In vivo models:
Biomarker identification:
Measuring PRDX4 activity in experimental systems requires multiple complementary approaches:
Protein expression analysis:
Western blotting for baseline PRDX4 protein levels
qRT-PCR for mRNA expression analysis
Immunohistochemistry to assess tissue localization and expression patterns
Redox state assessment:
Non-reducing SDS-PAGE to visualize different redox states of PRDX4 (monomers, dimers, and high molecular weight complexes)
Redox Western blotting with antibodies specific to different PRDX4 oxidation states
Peroxidase activity assays:
Measure H₂O₂ consumption rates in the presence of thioredoxin/thioredoxin reductase systems
Fluorescent or colorimetric peroxide detection assays
Cellular ROS monitoring:
Use of general ROS indicators (e.g., DCFDA) or more specific probes
Comparison of ROS levels between wild-type and PRDX4-depleted cells under basal and stress conditions
Functional readouts:
Analysis of ER stress markers (e.g., UPR-induced transcripts like XBP1s, ERdj4, and CHOP) to assess whether PRDX4 manipulation affects ER homeostasis
Protein folding efficiency assays to measure the impact of PRDX4 on its canonical function
Subcellular localization:
Immunofluorescence or cell fractionation followed by Western blotting to confirm ER localization
Proximity ligation assays to identify interaction partners
Rescue experiments:
When studying PRDX4 in cellular redox systems, the following controls should be included:
Cell type controls:
Genetic manipulation controls:
Non-targeting siRNA/shRNA controls
Multiple independent siRNA/shRNA sequences targeting PRDX4 to rule out off-target effects
Rescue experiments with PRDX4 cDNA resistant to the siRNA/shRNA used
Redox state controls:
Positive controls for oxidative stress (e.g., H₂O₂ treatment)
Antioxidant treatment (e.g., N-acetylcysteine) to confirm ROS involvement
Treatment with specific ROS scavengers to identify critical ROS species
Pathway inhibitor controls:
Other peroxiredoxin family members:
Assessment of other PRDXs (especially PRDX1-3) to determine specificity of PRDX4 effects
Compare cytosolic (PRDX1/2) versus ER (PRDX4) versus mitochondrial (PRDX3) peroxiredoxins to determine compartment-specific effects
Temporal controls:
Time-course experiments to distinguish immediate from adaptive responses
Inducible systems to control the timing of PRDX4 depletion
Quantification controls:
Standard curves for all quantitative assays
Multiple technical and biological replicates
Statistical analysis appropriate to the experimental design
PRDX4 expression shows significant variation across cancer types, with both increased and decreased expression patterns observed depending on the cancer:
Increased PRDX4 expression:
Glioblastoma: PRDX4 is increased in glioblastoma tissue compared to normal brain tissue
Ovarian cancer: PRDX4 expression is significantly higher in borderline than in benign epithelial ovarian tumors
Prostate cancer: Enhanced expression of PRDX4 is observed, which is negatively related to the TMPRSS2-ERG gene fusion status
Decreased PRDX4 expression:
Variable expression in pancreatic cancer:
These varying expression patterns suggest context-dependent roles for PRDX4 in cancer development and progression. The dichotomous expression changes highlight the complexity of redox regulation in cancer biology and suggest that both overexpression and underexpression of PRDX4 can contribute to malignant phenotypes depending on the cancer type and cellular context.
PRDX4 exhibits diverse roles in cancer cell proliferation and tumor growth that vary by cancer type:
Promoting cancer cell proliferation:
In prostate cancer, overexpression of PRDX4 remarkably improves the growth rate of cancer cell lines
In pancreatic cancer, approximately half of cell lines depend on PRDX4 for their proliferation and survival
Both established and primary pancreatic cancer cells show growth inhibition when PRDX4 is depleted, in 2D cultures, 3D spheroid models, and orthotopic xenografts
Supporting tumor survival:
Influencing metastatic potential:
Mediating treatment resistance:
These diverse functions highlight PRDX4's context-dependent role in cancer biology and suggest potential for therapeutic targeting in specific cancer types and contexts.
Based on current research, several approaches for therapeutically targeting PRDX4 in cancer can be considered:
Direct PRDX4 inhibition:
RNA interference approaches (siRNA, shRNA) have demonstrated efficacy in preclinical models
Development of small molecule inhibitors specific to PRDX4 would be valuable, though challenging due to structural similarities with other peroxiredoxins
Targeting the unique N-terminal signal sequence of PRDX4 might provide specificity
Synthetic lethality approaches:
Targeting PRDX4 in combination with radiation therapy, as PRDX4 depletion sensitizes cells to ionizing radiation
Combining PRDX4 inhibition with DNA damaging agents, given that PRDX4 depletion induces DNA damage and activates DNA damage response pathways
Dual targeting of PRDX4 and DNA-PK, as DNA-PK activation appears protective following PRDX4 depletion
Targeting upstream regulators:
Cancer type-specific approaches:
For prostate and breast cancers: Target secreted PRDX4 to prevent bone metastasis and osteolastogenesis
For glioblastoma: Combine PRDX4 inhibition with radiotherapy to overcome radioresistance
For pancreatic cancer: Identify biomarkers (e.g., NOX4 expression, NADPH levels) to select patients likely to respond to PRDX4-targeted therapy
Biomarker-guided therapy:
Targeting the secreted form:
Develop antibodies against extracellular PRDX4 to prevent its role in bone metastasis
Block PRDX4 from functioning as a secreted soluble factor in the tumor microenvironment
These approaches require further validation in preclinical models before advancing to clinical trials, but they represent promising strategies for exploiting PRDX4 dependency in cancer therapy.
Researchers face several technical challenges when studying PRDX4 function:
Distinguishing PRDX4 from other peroxiredoxins:
High sequence homology among peroxiredoxin family members can complicate specific detection
Need for highly specific antibodies that do not cross-react with other PRDX proteins
Challenge of generating PRDX4-specific inhibitors due to structural similarities
Compartment-specific redox measurements:
Accurately measuring ROS specifically within the ER lumen where PRDX4 primarily functions
Distinguishing between cytosolic and ER-localized PRDX4 isoforms
Monitoring real-time changes in PRDX4 oxidation state in living cells
Temporal dynamics of redox signaling:
Capturing rapid redox reactions and signaling events
Distinguishing between immediate PRDX4 functions and adaptive responses
Monitoring the transition between different PRDX4 oxidation states
In vivo models and translation:
Development of appropriate animal models to study PRDX4 function
Potential differences in redox biology between humans and model organisms
Translating findings from cell culture to in vivo settings
Context dependency:
Long-term inhibition challenges:
Contradictory findings regarding PRDX4 in different cancer types can be reconciled through several conceptual frameworks:
Context-dependent role based on redox environment:
Cancer-specific metabolic adaptations:
Different cancers adopt distinct metabolic profiles that may create varying degrees of dependence on redox systems
Cancers with high ER stress or secretory burden may be more dependent on PRDX4 function
The source and quantity of ROS production (e.g., NOX4 activity) likely influences PRDX4 dependency
Integration with complementary redox systems:
Redundancy in redox systems may explain why some cancers are PRDX4-independent
The relative expression of other peroxiredoxins or antioxidant enzymes may determine PRDX4 dependency
The capacity of alternative H₂O₂ scavenging systems may compensate for PRDX4 depletion in some contexts
Dual roles in cancer progression:
Methodological considerations:
Different experimental systems (cell lines, primary cells, animal models, human samples)
Varying methods of PRDX4 manipulation (transient vs. stable, partial vs. complete depletion)
Different readouts and endpoints measured across studies
Integration with genetic background:
Reconciling these contradictions requires comprehensive characterization of PRDX4 function across multiple cancer types, with careful attention to cellular context, genetic background, and metabolic state.
Several promising future directions for PRDX4 research emerge from current findings:
Single-cell resolution studies:
Investigating PRDX4 function and dependency at single-cell level to capture heterogeneity
Correlating PRDX4 activity with specific cellular states and phenotypes
Mapping PRDX4 oxidation dynamics in real-time using advanced redox biosensors
Multi-omics integration:
Combining proteomics, metabolomics, and transcriptomics to build comprehensive models of PRDX4 function
Identifying metabolic signatures that predict PRDX4 dependency
Mapping the PRDX4 interactome under different cellular conditions
Translational applications:
Developing PRDX4 as a biomarker for patient stratification in cancer therapy
Creating PRDX4-targeting therapeutic approaches for PRDX4-dependent cancers
Exploring combination therapies that exploit PRDX4 dependency
Mechanistic studies:
Further characterizing the DNA damage induced by PRDX4 depletion
Investigating the relationship between PRDX4 and circadian rhythms
Understanding how PRDX4 coordinates with unfolded protein response pathways
Systems biology approaches:
Mathematical modeling of redox shuttles and their relationship with PRDX4
Network analysis of PRDX4 interactions within broader redox systems
Prediction of synthetic lethal interactions with PRDX4
Therapeutic development:
Design of specific PRDX4 inhibitors
Development of strategies to target secreted versus ER-resident PRDX4
Exploration of immunotherapeutic approaches targeting PRDX4 in the tumor microenvironment
Role in non-cancer diseases:
Investigating PRDX4 in inflammatory diseases, diabetes, and neurodegenerative disorders
Exploring PRDX4 as a biomarker for various diseases beyond cancer
Understanding the role of PRDX4 in normal physiology and aging
These directions will advance our understanding of PRDX4 biology and potentially lead to new diagnostic and therapeutic approaches for cancer and other diseases.
Prx4 functions as an antioxidant enzyme, reducing hydrogen peroxide to water in a thiol-dependent catalytic cycle . This activity is vital for maintaining cellular redox balance and protecting cells from oxidative damage. Additionally, Prx4 has been linked to the regulation of the pro-inflammatory transcription factor, nuclear factor kappa B (NF-κB) .
Prx4 can switch between peroxidase and chaperone activities through redox-dependent and reversible conversion from disulfide-linked homodimers to higher-order multimers . This versatility allows Prx4 to interact with various binding partners, including stress-responsive kinases, membrane proteins, and immune modulators, thereby fine-tuning hydrogen peroxide signaling .
Prx4 has emerged as a potential biomarker for various diseases due to its differential expression in healthy individuals and patients with acute or chronic conditions . It has been linked to morbidity and mortality in patients with sepsis and non-specific complaints in emergency departments . The measurement of Prx4 levels, redox state, oligomerization, and nitro-oxidative modifications can provide valuable insights into the oxidative state of patients and the progression of diseases .