PRDX2 Mouse

Eukaryotic Translation Initiation Factor 4E Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of PRDX2 Mouse

PRDX2 (peroxiredoxin 2) is a 22 kDa antioxidant enzyme encoded by the Prdx2 gene in mice. It belongs to the peroxiredoxin family and functions as a hydrogen peroxide (H2_2O2_2) scavenger, utilizing thioredoxin to reduce peroxides and regulate redox signaling . PRDX2 is ubiquitously expressed, with high abundance in erythrocytes, where it stabilizes hemoglobin and protects against oxidative damage . It also modulates cellular processes such as apoptosis, inflammation, and differentiation through pathways like TLR4/NF-κB and PI3K/AKT .

Research Models: PRDX2 Knockout Mice

PRDX2-deficient (Prdx2/^{-/-}) mice are viable and fertile but exhibit distinct phenotypic abnormalities:

PhenotypeKey FindingsSource
Hemolytic AnemiaAbnormal erythrocyte morphology, Heinz bodies, and shortened red blood cell lifespan .
AtherosclerosisAccelerated plaque formation in ApoE/^{-/-} mice due to increased vascular inflammation .
Bone HomeostasisHigher bone mass due to impaired osteoclastogenesis and enhanced osteoblast differentiation .
Immune DysregulationExacerbated LPS-induced inflammation and altered T-cell responses .
Metabolic EffectsImproved glucose tolerance under high-fat diet conditions .

Antioxidant Activity

PRDX2 reduces H2_2O2_2 and organic hydroperoxides via its peroxidatic cysteine residue, regenerated by thioredoxin . In erythrocytes, it binds hemoglobin to prevent oxidative denaturation .

Signaling Pathways

  • TLR4/NF-κB: PRDX2 inhibits NF-κB activation, reducing oxidative stress and apoptosis in myocardial infarction models .

  • PI3K/AKT: Overexpression in cervical cancer cells suppresses ER stress and apoptosis induced by bleomycin .

  • Wnt/β-catenin: PRDX2 stabilizes tankyrase, promoting Axin degradation and β-catenin activation in colorectal cancer .

Cardiovascular Disease

PRDX2 deficiency exacerbates myocardial injury post-acute myocardial infarction (AMI) by elevating ROS and caspase activation . In atherosclerosis, Prdx2/^{-/-} mice show increased vascular adhesion molecule expression (VCAM-1/ICAM-1) and immune cell infiltration .

Cancer

  • Cervical Cancer: PRDX2 overexpression reduces bleomycin-induced ROS and ER stress, enhancing cell survival .

  • Colorectal Cancer: PRDX2 promotes tumor progression via Wnt/β-catenin signaling .

Reproductive Health

Reduced PRDX2 levels in placental trophoblasts correlate with recurrent miscarriage due to elevated ROS and impaired cell proliferation .

Therapeutic Implications

  • Recombinant PRDX2: Administering recombinant PRDX2 in AMI models reduces infarct size and improves cardiac function .

  • Antioxidant Therapy: Ebselen (a PRDX mimetic) rescues atherosclerotic phenotypes in Prdx2/^{-/-} mice .

  • Cancer Treatment: Targeting PRDX2 sensitizes cancer cells to ROS-inducing therapies like bleomycin .

Product Specs

Introduction

PRDX2, a member of the peroxiredoxin family, functions as an antioxidant enzyme by reducing hydrogen peroxide and alkyl hydroperoxides. This enzyme plays a crucial role in cellular protection against oxidative stress and contributes to the antiviral response of CD8(+) T-cells. Additionally, PRDX2 has been implicated in cancer development and progression due to its proliferative effects. Insufficient PRDX2 protection against peroxidases can lead to DNA damage, potentially resulting in neurological disorders like Alzheimer's disease or promoting cancer development.

Description

Recombinantly produced in E. coli, PRDX2 Mouse Recombinant is a non-glycosylated polypeptide chain with a single chain. It consists of 222 amino acids (1-198 a.a), with a molecular weight of 24.3 kDa. A 24 amino acid His-tag is fused to the N-terminus of PRDX2. Purification is achieved through proprietary chromatographic techniques.

Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation

The PRDX2 protein solution has a concentration of 1 mg/ml. It is prepared in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, and 1mM DTT.

Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability, consider adding a carrier protein (0.1% HSA or BSA). Multiple freeze-thaw cycles should be avoided.
Purity

SDS-PAGE analysis has determined the purity to be greater than 90.0%.

Biological Activity

The enzyme exhibits a specific activity greater than 700 pmol/min/ug. Specific activity is defined as the amount of hydroperoxide reduced by 1ug of enzyme per minute at a temperature of 25°C.

Synonyms

PRDX2, Peroxiredoxin-2 (EC:1.11.1.15), TSA, Thioredoxin peroxidase 1, Thioredoxin-dependent peroxide reductase 1, Thiol-specific antioxidant protein, Prdx2, Tdpx1, Tpx.  

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSHMASGNA QIGKSAPDFT ATAVVDGAFK EIKLSDYRGK YVVLFFYPLD FTFVCPTEII AFSDHAEDFR KLGCEVLGVS VDSQFTHLAW INTPRKEGGL GPLNIPLLAD VTKSLSQNYG VLKNDEGIAY RGLFIIDAKG VLRQITVNDL PVGRSVDEAL RLVQAFQYTD EHGEVCPAGW KPGSDTIKPN VDDSKEYFSK HN

Q&A

What are the primary phenotypic characteristics of PRDX2 knockout mice?

PRDX2 knockout (Prdx2-/-) mice are viable and fertile but exhibit hemolytic anemia as their most prominent phenotype . The anemia is accompanied by red blood cell abnormalities, including increased intraerythrocytic H₂O₂ levels . Importantly, these mice display multiple other organ-specific phenotypes including defective iron homeostasis, blood vessel defects, platelet abnormalities, and altered inflammatory responses . While the most widely used knockout model was generated by replacing genomic DNA encoding exons 1-5 with a neomycin resistance cassette, various research groups have developed other PRDX2 deletion approaches . When designing experiments, researchers should consider that PRDX2-deficient mice may exhibit a spectrum of hematological and vascular anomalies that could influence experimental outcomes beyond the primary research question.

How does PRDX2 deficiency affect hydrogen peroxide metabolism in erythrocytes?

PRDX2 deficiency significantly alters H₂O₂ metabolism in erythrocytes. Mathematical modeling and experimental data indicate that PRDX2 plays a more substantial role than GPX1 in removing endogenously generated H₂O₂ . When PRDX2 is absent, intracellular H₂O₂ levels increase by approximately 400% (from ~2.5×10⁻¹⁰ M to 10.1×10⁻¹⁰ M), while GPX1 deficiency causes only a 20% increase (to 3.2×10⁻¹⁰ M) . This suggests PRDX2 is the primary enzyme responsible for detoxifying endogenous H₂O₂ in erythrocytes. Interestingly, PRDX2-deficient erythrocytes show no increased sensitivity to exogenous H₂O₂ or organic peroxides compared to wild-type cells . This apparent paradox is explained by the role of catalase, which remains the primary enzyme for detoxifying exogenous H₂O₂, while PRDX2 handles endogenous peroxide. Researchers studying oxidative stress in erythrocytes should differentiate between responses to endogenous versus exogenous peroxide sources when interpreting data from PRDX2-deficient models.

What experimental approaches best measure the impact of PRDX2 deficiency on erythrocyte redox status?

To comprehensively assess erythrocyte redox status in PRDX2-deficient mice, researchers should employ multiple complementary techniques:

  • Catalase inhibition kinetics: Measure the rate of irreversible catalase inhibition by 3-amino-1,2,4-triazole (3-AT), which selectively binds to Compound I of catalase. PRDX2-deficient erythrocytes show approximately twice the rate of catalase inhibition compared to wild-type cells, reflecting elevated H₂O₂ levels .

  • Direct H₂O₂ quantification: While technically challenging in erythrocytes, genetically encoded H₂O₂ sensors (like HyPer or roGFP2-Orp1) can be used in nucleated erythroid precursor cells.

  • Protein oxidation markers: Measure oxidative modifications to hemoglobin and membrane proteins through techniques such as mass spectrometry to detect carbonylation, disulfide formation, and other oxidative modifications.

  • Metabolic pathway analysis: Analyze the pentose phosphate pathway activity and NADPH/NADP⁺ ratios, as these reflect the cell's response to oxidative challenge.

  • Membrane lipid peroxidation: Quantify products like malondialdehyde or 4-hydroxynonenal to assess membrane damage resulting from elevated ROS.

This multi-parameter approach provides a more complete picture of redox disturbances than single measurements and helps differentiate primary from secondary effects of PRDX2 deficiency.

Why do PRDX2-deficient mice develop anemia despite normal responses to exogenous peroxides?

The anemia in PRDX2-deficient mice presents an interesting paradox: mature erythrocytes show normal responses to exogenous peroxides yet the mice develop significant anemia . This suggests the anemia stems from disruptions during erythropoiesis rather than accelerated destruction of mature erythrocytes. PRDX2 appears critical for protecting erythroid precursors from ROS-mediated DNA damage during the highly oxidative process of hemoglobin synthesis . Additionally, PRDX2's absence affects iron homeostasis, which is essential for proper erythropoiesis . Researchers investigating this phenomenon should examine:

  • The erythroid maturation process in bone marrow using flow cytometry with markers for developmental stages

  • DNA damage in erythroid precursors using γH2AX staining

  • Erythrocyte lifespan using in vivo biotinylation assays

  • Compensatory responses in the hematopoietic system, including erythropoietin levels and splenic erythropoiesis

This mechanistic understanding explains why interventions targeting mature erythrocyte peroxide metabolism might not alleviate the anemia phenotype in PRDX2-deficient models.

What control considerations are essential when designing experiments with PRDX2 knockout mice?

When designing experiments with PRDX2 knockout mice, several critical control considerations must be addressed:

  • Genetic background matching: Ensure wild-type controls have the same genetic background as the knockout mice. The search results indicate that PRDX2 knockout mice have been backcrossed to C57BL/6 mice , so matched C57BL/6 controls should be used.

  • Age and sex matching: PRDX2 functions may have age and sex-dependent effects, particularly given its role in ovarian function . Age-matched same-sex comparisons are essential.

  • Littermate controls: When possible, use littermate controls to minimize environmental variables, especially for developmental studies.

  • Compensation by other peroxiredoxins: Consider potential compensatory upregulation of other peroxiredoxins, particularly PRDX1 given its high sequence similarity to PRDX2 . Measurement of other antioxidant enzymes should be included in experimental designs.

  • Secondary phenotype influences: Account for the anemia phenotype as a potential confounder in non-hematological studies, as tissue oxygenation differences could affect experimental outcomes.

  • Housing conditions: Standard housing conditions may represent a relatively low oxidative stress environment. Consider including experimental oxidative stressors (like paraquat challenge or high-fat diet) to reveal phenotypes that might not be apparent under baseline conditions.

These considerations help distinguish direct effects of PRDX2 deficiency from secondary adaptations or strain-specific characteristics.

How can researchers differentiate between the hematological and non-hematological functions of PRDX2?

Differentiating between hematological and non-hematological functions of PRDX2 presents a significant challenge due to the systemic effects of anemia. Several experimental approaches can help address this issue:

  • Bone marrow chimeras: Transplant PRDX2-deficient bone marrow into wild-type recipients and vice versa to isolate hematopoietic from non-hematopoietic PRDX2 functions.

  • Tissue-specific conditional knockouts: If available, use Cre-loxP systems to delete PRDX2 specifically in either erythroid precursors or non-hematological tissues of interest.

  • Ex vivo tissue studies: Compare responses of isolated tissues from wild-type and PRDX2-knockout mice under controlled conditions that minimize the influence of anemia.

  • Anemia correction: Use erythropoietin administration or other approaches to correct anemia in PRDX2-knockout mice, then assess whether non-hematological phenotypes persist.

  • Complementary in vitro studies: Use PRDX2 knockdown or overexpression in relevant cell lines to confirm tissue-specific functions without systemic complications.

This multi-faceted approach allows researchers to dissect the direct tissue-specific roles of PRDX2 from indirect consequences of anemia or systemic redox imbalance.

What are the appropriate molecular markers to verify PRDX2 knockout efficiency?

Verification of PRDX2 knockout efficiency requires comprehensive molecular characterization at multiple levels:

  • Genomic verification: PCR genotyping to confirm the presence of the targeting construct and absence of wild-type alleles.

  • Transcript analysis: RT-qPCR targeting multiple exons to ensure complete ablation of PRDX2 mRNA, particularly checking for potential truncated transcripts or alternative splice variants.

  • Protein verification: Western blotting using antibodies targeting different epitopes of PRDX2 to rule out expression of truncated protein forms. Include positive controls (wild-type tissue) and loading controls.

  • Immunohistochemistry: Tissue staining to confirm absence of PRDX2 protein in the specific cell types relevant to your research question.

  • Functional assays: Measure peroxiredoxin activity using substrate-specific assays to confirm loss of PRDX2 function, distinguishing it from other peroxiredoxin family members.

  • Downstream markers: Assess known PRDX2-dependent changes, such as increased catalase Compound I formation in erythrocytes , as functional confirmation of knockout effectiveness.

This multi-level verification ensures that experimental phenotypes can be confidently attributed to PRDX2 deficiency rather than incomplete knockout or compensatory mechanisms.

How does PRDX2 deficiency interact with other antioxidant systems in mouse models?

PRDX2 deficiency triggers complex interactions with other antioxidant systems, creating a rebalanced redox network:

  • Catalase interaction: In PRDX2-deficient erythrocytes, the fraction of catalase present as Compound I increases from 6% to 14% . This results in accelerated irreversible catalase inhibition when exposed to 3-AT, approximately twice as fast as in wild-type cells .

  • Glutathione system: While PRDX2 and glutathione peroxidase 1 (GPX1) both participate in H₂O₂ removal, kinetic modeling indicates PRDX2 plays a larger role . Interestingly, cells lacking both catalase and GPX1 show greater sensitivity to exogenous H₂O₂ than cells lacking only catalase, while PRDX2 deficiency doesn't affect this exogenous peroxide sensitivity .

  • Peroxiredoxin family compensation: Consider potential upregulation of other peroxiredoxins, particularly PRDX1, which shares high sequence similarity with PRDX2 . This compensation may mask some phenotypes or create tissue-specific differences in antioxidant response.

  • NADPH-generating systems: Examine potential adaptations in the pentose phosphate pathway and other NADPH-generating systems that support glutathione recycling and peroxiredoxin reduction.

When interpreting data from PRDX2-deficient mice, researchers should consider these interactions and measure multiple components of the antioxidant network to capture the system-level reorganization that occurs in response to PRDX2 loss.

What are the most significant contradictions in published data regarding PRDX2 knockout phenotypes?

Several notable contradictions exist in the literature regarding PRDX2 knockout phenotypes that researchers should be aware of:

  • Inflammation regulation: PRDX2 deficiency has been reported to have both pro-inflammatory and anti-inflammatory effects depending on the experimental model and tissue examined. In systemic LPS challenge, PRDX2-deficient macrophages show enhanced pro-inflammatory responses, while in dextran sulfate sodium (DSS)-induced colitis, PRDX2 deficiency appears protective .

  • Tumorigenesis role: Unlike PRDX1 knockouts that consistently show increased tumor susceptibility, PRDX2-deficient mice show anti-tumorigenesis effects in some models (e.g., Apc+/Min intestinal/colon tumors) , suggesting context-dependent tumor suppressor or promoter roles.

  • Vascular phenotypes: Blood vessel responses in PRDX2 knockouts show contradictory reports, with some studies reporting defects in response to balloon injury while others report defects in response to FeCl₃ .

These contradictions likely reflect:

  • Tissue-specific PRDX2 functions

  • Compensatory mechanisms that differ across experimental contexts

  • Interaction of PRDX2 with different signaling pathways depending on the nature of the stimulus

  • Differences in genetic background or environmental factors

Researchers should carefully consider these contradictions when designing experiments and interpret results within the specific experimental context being studied.

How can PRDX2 mouse models inform our understanding of human diseases?

PRDX2 mouse models provide valuable insights into several human disease mechanisms:

  • Hemolytic anemias: PRDX2-deficient mice develop hemolytic anemia with red cell abnormalities , suggesting PRDX2 variants may contribute to unexplained hemolytic anemias in humans. Mouse models can help test therapeutic approaches targeting the redox imbalance in these conditions.

  • Vascular diseases: PRDX2 knockout mice exhibit blood vessel defects and altered responses to vascular injury , indicating potential roles in atherosclerosis, restenosis, and other vascular pathologies. The acceleration of atherosclerosis in PRDX2-deficient mice on atherogenic diets provides a model for studying redox-sensitive vascular disease mechanisms .

  • Inflammatory disorders: The complex pro- and anti-inflammatory phenotypes observed in different tissues of PRDX2-deficient mice parallel the tissue-specific nature of human inflammatory diseases. These models help explain how the same antioxidant protein might have opposing effects in different inflammatory contexts.

  • Metabolic disorders: PRDX2-deficient mice show metabolic defects, particularly on high-fat diets , informing studies of redox regulation in metabolic syndrome and diabetes.

When translating findings from mouse models to human disease, researchers should consider:

  • Human genetic variation in PRDX2 and related pathways

  • Differences in mouse versus human redox biology

  • The potential for therapeutic targeting of PRDX2 or its downstream pathways

What are the key technical challenges in studying PRDX2 function in mouse models?

Studying PRDX2 function in mouse models presents several technical challenges:

  • Redox state preservation: PRDX2's redox state changes rapidly during sample processing. Researchers must use rapid alkylation techniques (like N-ethylmaleimide treatment) to preserve the in vivo redox state before analysis.

  • Peroxide measurement: Accurately measuring H₂O₂ levels in tissues is technically challenging due to the short half-life and reactivity of peroxides. Consider using genetically encoded sensors or specialized probe compounds with appropriate controls.

  • Distinguishing from other peroxiredoxins: PRDX2 shares high homology with other peroxiredoxins (particularly PRDX1) , making specific detection challenging. Validate antibody specificity using knockout tissues and consider using multiple antibodies targeting different epitopes.

  • Compensatory adaptations: PRDX2 knockout mice may develop compensatory mechanisms that mask phenotypes. Consider acute knockdown approaches (e.g., inducible systems) to study immediate effects before compensation occurs.

  • Context dependency: PRDX2 functions appear highly context-dependent, with effects varying by tissue, age, and environmental conditions . Standardize experimental conditions and include appropriate stressors to reveal phenotypes.

  • Secondary effects of anemia: The anemia in PRDX2-deficient mice can confound interpretation of other phenotypes. Consider using tissue-specific knockouts or ex vivo approaches for non-hematological studies.

Addressing these challenges requires careful experimental design and application of complementary methodologies to ensure robust, reproducible results.

What are the optimal experimental conditions for revealing PRDX2-dependent phenotypes?

Revealing PRDX2-dependent phenotypes often requires specific experimental conditions:

  • Oxidative stress challenges: PRDX2-dependent phenotypes may only become apparent under oxidative stress conditions. Consider:

    • Paraquat administration (0.1-0.5 mg/kg) for acute oxidative stress

    • Hyperoxia (80-100% O₂) for respiratory studies

    • Ischemia-reperfusion models for cardiovascular studies

    • High-fat diet for metabolic phenotypes

    • Age-related studies, as older mice may show more pronounced phenotypes

  • Cell-specific analyses: Rather than whole-tissue analysis, examine specific cell populations where PRDX2 function is most critical. For example:

    • Erythroid precursors at different maturation stages

    • Endothelial cells in vascular studies

    • Macrophages in inflammation models

  • Temporal considerations: Some phenotypes may be transient or occur only at specific developmental stages. Design time-course experiments that capture:

    • Early erythroid development

    • Acute phases of inflammatory responses

    • Recovery periods after oxidative challenge

  • Combined genetic models: Cross PRDX2-deficient mice with other relevant genetic models to reveal phenotypes. For example:

    • ApoE-/- for atherosclerosis studies

    • Sod1-/- for enhanced oxidative stress

    • Inflammatory disease models

By systematically applying these conditions, researchers can unmask PRDX2-dependent phenotypes that might remain hidden under standard laboratory conditions.

How can researchers accurately model PRDX2 redox biochemistry in vivo?

Accurately modeling PRDX2 redox biochemistry in vivo requires sophisticated approaches:

  • Mathematical modeling: As demonstrated in the search results, kinetic modeling has successfully predicted H₂O₂ levels in PRDX2-deficient erythrocytes . Researchers should:

    • Incorporate Michaelis-Menten kinetics for relevant enzymes

    • Account for PRDX2's unique kinetic properties (high reactivity, potential for hyperoxidation)

    • Model interactions with other antioxidant systems

    • Validate predictions with experimental measurements

  • Redox proteomics: To capture PRDX2's interactions and substrates:

    • Use redox-sensitive probes coupled with mass spectrometry

    • Apply differential alkylation techniques to quantify the redox state of specific cysteines

    • Perform immunoprecipitation under non-reducing conditions to identify PRDX2 interaction partners

  • In vivo reporters: Deploy genetically encoded sensors to monitor redox changes in real-time:

    • HyPer or roGFP2-Orp1 for H₂O₂ dynamics

    • roGFP2-Grx1 for glutathione redox potential

    • Targeted sensors to specific subcellular compartments

  • Multi-parameter assessment: Combine multiple measurements to build a comprehensive redox profile:

    • Direct peroxide measurements

    • Glutathione redox status (GSH/GSSG ratio)

    • Protein thiol modifications

    • Lipid peroxidation products

    • Metabolic flux through NADPH-generating pathways

This integrated approach allows researchers to move beyond simple oxidative stress measurements to accurately model the complex redox biochemistry influenced by PRDX2 in living systems.

What emerging technologies could advance PRDX2 mouse model research?

Several emerging technologies have the potential to significantly advance PRDX2 mouse model research:

  • CRISPR/Cas9 genome editing: As noted in the search results, CRISPR/Cas9 technology has dramatically reduced the time and effort required to generate precisely modified mouse models . This technology enables:

    • Creation of conditional knockouts with improved tissue specificity

    • Introduction of specific human PRDX2 variants to model disease-associated mutations

    • Generation of reporter knock-ins to monitor PRDX2 expression in real-time

  • Single-cell technologies: Single-cell RNA sequencing and proteomics can reveal cell-type-specific responses to PRDX2 deficiency that may be masked in bulk tissue analyses. This is particularly valuable for heterogeneous tissues like bone marrow, where PRDX2 may differentially affect various cell populations.

  • Live-cell redox imaging: Advanced microscopy techniques combined with genetically encoded redox sensors allow real-time visualization of redox dynamics in PRDX2-deficient cells and tissues, providing spatial and temporal resolution not possible with biochemical assays.

  • Proteomics approaches: Targeted redox proteomics can identify specific proteins affected by altered redox status in PRDX2-deficient mice, helping to elucidate the mechanistic links between PRDX2 deficiency and observed phenotypes.

  • Metabolomics integration: Comprehensive metabolomic profiling can reveal how PRDX2 deficiency affects cellular metabolism beyond direct redox effects, potentially uncovering new therapeutic targets.

These technologies, especially when applied in combination, promise to provide unprecedented insights into PRDX2 function in health and disease.

What are the most critical unanswered questions in PRDX2 mouse model research?

Despite extensive study, several critical questions about PRDX2 biology remain unanswered:

  • Signal transduction vs. antioxidant functions: To what extent does PRDX2 function as a redox-dependent signaling molecule versus a simple antioxidant? Mouse models using redox-inactive PRDX2 mutants could help distinguish these roles.

  • Erythropoietic failure mechanisms: While PRDX2-deficient mice develop anemia , the precise stage at which erythropoiesis is disrupted and the molecular mechanisms involved remain incompletely characterized.

  • Tissue-specific functions: Why does PRDX2 deficiency produce such diverse and sometimes contradictory phenotypes across different tissues ? Understanding tissue-specific PRDX2 interaction partners and substrates is crucial.

  • Compensation mechanisms: How do other antioxidant systems compensate for PRDX2 deficiency, and why is this compensation sufficient in some contexts but not others?

  • Therapeutic potential: Can modulation of PRDX2 activity or expression provide therapeutic benefit in diseases involving oxidative stress? Conditional and inducible mouse models would be valuable for addressing this question.

  • Aging effects: How does PRDX2 deficiency affect the aging process, particularly in tissues with high metabolic activity and ROS production?

Addressing these questions will require integrated approaches combining advanced mouse genetic models with detailed molecular and cellular analyses across multiple tissues and developmental stages.

How should researchers interpret contradictory results between in vitro and in vivo PRDX2 studies?

Contradictions between in vitro and in vivo PRDX2 studies are common and require careful interpretation:

  • Redox environment differences: In vitro systems often have higher oxygen levels and different redox buffering capacities than in vivo environments. When interpreting contradictory results, consider that:

    • Standard cell culture (21% O₂) represents hyperoxic conditions compared to tissues

    • Culture media may lack important redox-active components present in vivo

    • The absence of systemic redox regulation may alter cellular responses

  • Compensatory mechanisms: In vivo systems develop compensatory responses to PRDX2 deficiency that may be absent or different in vitro:

    • Chronic PRDX2 deficiency in vivo triggers adaptive responses

    • Acute PRDX2 knockdown in vitro may reveal primary effects before compensation occurs

    • Consider using inducible knockout systems in vivo to distinguish acute from chronic effects

  • Cell-cell interactions: PRDX2 functions may depend on intercellular communications absent in isolated cell systems:

    • Erythrocyte interactions with splenic macrophages influence redox homeostasis

    • Inflammatory responses involve complex cellular networks

    • Consider co-culture systems to better approximate in vivo cellular environments

  • Technical considerations:

    • Verify knockout/knockdown efficiency using the same methods across systems

    • Use identical stressors and measurement techniques when comparing systems

    • Consider the developmental state of cells (primary cells vs. cell lines vs. in vivo tissues)

When faced with contradictions, researchers should design experiments that bridge in vitro and in vivo approaches, such as ex vivo tissue studies or organoid cultures, to identify the biological basis for the discrepancies rather than simply dismissing one set of results.

How do phenotypes of different peroxiredoxin knockout models compare?

Comparative analysis of peroxiredoxin knockout models reveals distinct and overlapping phenotypes that inform our understanding of their specialized functions:

GenePrimary PhenotypesAffected Organs/TissuesUnique Features
Prdx1Hemolytic anemia, Tumorigenesis, InflammationRBCs, Various tissues, Liver, LungStrong tumor susceptibility not seen in other Prdx knockouts
Prdx2Hemolytic anemia, Blood vessel defects, Inflammation (pro/anti)RBCs, Endothelial cells, Immune cellsMore severe erythrocyte phenotype; context-dependent inflammatory effects
Prdx3Mitochondrial dysfunction, Apoptosis, Muscle defectsMitochondria, Muscle, BrainUnique metabolic phenotypes due to mitochondrial localization
Prdx4ER stress, Defective spermatogenesis, Liver defectsTestis, Liver, ColonOnly peroxiredoxin with secreted form; significant roles in tissue remodeling
Prdx5Metabolic defectsFat, LiverMulti-compartment localization (cytosol, mitochondria, peroxisomes)
Prdx6Tissue vulnerability to oxidative stress, Metabolic defectsLung, Kidney, Liver, SkinOnly 1-Cys peroxiredoxin; unique phospholipase A2 activity

This comparison highlights how subcellular localization, tissue expression patterns, and biochemical properties contribute to the specialized functions of each peroxiredoxin. Researchers should consider these differences when selecting appropriate knockout models for specific research questions and when interpreting phenotypes that may involve multiple peroxiredoxin family members.

When should researchers use PRDX2 knockout mice versus other peroxiredoxin knockouts?

Selection of the appropriate peroxiredoxin knockout model should be guided by the specific research question:

Choose PRDX2 knockout mice for studies of:

  • Erythrocyte redox biology and hemolytic disorders - PRDX2 has the most prominent erythrocyte phenotype

  • Endothelial function and vascular responses - PRDX2 knockouts show significant blood vessel defects

  • Context-dependent inflammatory regulation - PRDX2 has complex pro/anti-inflammatory effects depending on tissue and stimulus

  • Iron homeostasis disorders - PRDX2 knockouts show defective iron handling

Choose alternative peroxiredoxin knockouts when focusing on:

  • Tumorigenesis - PRDX1 knockouts show stronger tumor phenotypes

  • Mitochondrial redox biology - PRDX3 knockouts specifically affect mitochondrial function

  • ER stress responses - PRDX4 is the primary ER-localized peroxiredoxin

  • Specific tissue defects - Consider tissue-specific expression patterns (e.g., PRDX6 for lung studies)

Consider combination knockouts for:

  • Studies of redundancy between PRDX1 and PRDX2, which share high sequence similarity

  • Investigation of compartment-specific redox regulation (e.g., combining cytosolic and mitochondrial peroxiredoxin knockouts)

  • Comprehensive analysis of peroxide metabolism (e.g., combining peroxiredoxin knockouts with catalase or GPX1 deficiency)

The choice of model should be informed by both the biological question and the specific experimental endpoints, with careful consideration of the potential confounding effects of each model's phenotype.

How do compensatory mechanisms differ across peroxiredoxin knockout models?

Compensatory mechanisms in peroxiredoxin knockout models show important differences:

Product Science Overview

Structure and Function

eIF4E is a member of the eukaryotic initiation factor 4 (eIF4) family. It recognizes and binds to the 5’ cap structure of eukaryotic mRNAs, which is a 7-methylguanosine triphosphate (m7GpppN) cap . This binding is essential for the recruitment of the ribosome to the mRNA, facilitating the initiation of translation . eIF4E functions in synergy with other proteins such as the helicase eIF4A and the scaffolding protein eIF4G to form the eIF4F complex . This complex unwinds secondary structures in the mRNA 5’ untranslated regions (UTRs) to allow ribosome binding and translation initiation .

Regulation

The activity of eIF4E is tightly regulated in cells under normal physiological conditions. It can be controlled by post-translational modifications, such as phosphorylation, and by the binding of inhibitory proteins, including eIF4E binding proteins (4E-BPs) and CYFIP1 . These regulatory mechanisms ensure that protein synthesis is appropriately modulated in response to cellular conditions and external stimuli .

Role in Cellular Stress

eIF4E is also involved in the cellular response to stress. It localizes to processing bodies (PBs) and stress granules (SGs) under different stress conditions . For instance, during heat shock, eIF4E can be redirected to stress granules, indicating its role in the cellular stress response . This localization is crucial for the regulation of mRNA stability and translation during stress .

Recombinant Mouse eIF4E

Recombinant mouse eIF4E is a form of the protein that has been produced using recombinant DNA technology. This involves inserting the gene encoding eIF4E into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant proteins are often used in research to study the function and regulation of the protein in a controlled environment .

Applications in Research

Recombinant eIF4E is widely used in biochemical and structural studies to understand its role in translation initiation and its interactions with other proteins. It is also used in studies investigating the regulation of protein synthesis and the cellular response to stress . By using recombinant eIF4E, researchers can gain insights into the molecular mechanisms underlying translation initiation and its regulation in eukaryotic cells .

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.