PDIA4 Human, Active

Protein Disulfide Isomerase A4 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Biochemical Activity

PDIA4 belongs to the protein disulfide isomerase (PDI) family, featuring thioredoxin-like domains that enable redox activity and substrate binding. Key structural features include:

  • Domains: Two catalytic thioredoxin (TRX) domains (a and a') with CGHC active-site motifs, and two non-catalytic domains (b and b') for substrate recognition .

  • Redox Activity: Facilitates disulfide bond formation, isomerization, and reduction via its TRX domains, ensuring proper protein folding under ER stress .

  • Post-Translational Roles: Modulates insulin signaling, inflammatory pathways, and calcium homeostasis through interactions with substrates like IRS-1 and Akt .

In Vitro and In Vivo Studies

  • Palmitate-Induced Insulin Resistance: In C2C12 myotubes, palmitate (a saturated fatty acid) upregulated PDIA4 expression, increased inflammatory cytokines (e.g., IL-6), and impaired insulin signaling (reduced Akt phosphorylation and glucose uptake) .

  • PDIA4 Knockdown: Silencing PDIA4 in C2C12 cells reversed insulin resistance, restoring glucose uptake by 35% and reducing phosphorylated IRS-1 (Ser307) levels .

  • Metformin Modulation: Metformin reduced PDIA4 expression in high-fat diet (HFD)-induced insulin-resistant mice, correlating with improved AMPK activation and glucose tolerance .

Human Clinical Data

A study of 444 adults revealed significant associations between serum PDIA4 levels and metabolic markers :

Table 1: Serum PDIA4 Correlations with Metabolic Parameters

ParameterCorrelation Coefficient (r)P-value
HOMA-2IR (insulin resistance)0.274<0.001
BMI0.405<0.001
Triglycerides0.327<0.001
IL-6 (inflammation)0.1540.001

Subjects in the highest HOMA-2IR tertile had 22.1 ± 19.94 ng/mL PDIA4 vs. 11.89 ± 11.64 ng/mL in the lowest tertile (P < 0.001) .

Oncogenic Roles in Cancer

PDIA4 is overexpressed in multiple cancers and promotes malignancy through:

  • Chemoresistance: In CDDP-resistant NSCLC A549 cells, PDIA4 inactivation restored mitochondrial apoptosis (Bcl-2 downregulation, caspase-3/9 cleavage) and sensitized cells to cisplatin .

  • Cervical Cancer:

    • Proliferation/Migration: PDIA4 knockdown in SiHa and ME180 cells reduced cyclin D1 (↓40%), PCNA (↓35%), and vimentin (↓50%), while increasing E-cadherin (↑2.5-fold) .

    • Survival Impact: High PDIA4 expression correlated with poor prognosis in cervical cancer (HR = 3.68 for PDIA4high/PLOD3high vs. PDIA4low/PLOD3low, P < 0.0001) .

Table 2: PDIA4-Driven Pathways in Cancer

Cancer TypeKey MechanismsClinical Outcome
Lung AdenocarcinomaER stress adaptation, RIPK1 suppressionOverexpression in 10/10 CDDP-treated patients
Cervical CancerGlycan biosynthesis, protein exportReduced 5-year survival (P = 0.0095)

Therapeutic Targeting of PDIA4

  • Metformin: Suppresses PDIA4 expression via AMPK activation, offering dual benefits in insulin resistance and ER stress mitigation .

  • Small-Molecule Inhibitors: Bacitracin (a pan-PDI inhibitor) and isoform-specific siRNAs restored chemosensitivity in ovarian (A2780R) and lung cancer models .

  • Clinical Potential: Serum PDIA4 levels may serve as a biomarker for metabolic disease progression or chemoresistance monitoring .

Research Gaps and Future Directions

  • Mechanistic Insights: The precise interaction between PDIA4 and IRS-1/Akt in insulin signaling remains unresolved .

  • Isoform-Specific Inhibitors: Current inhibitors lack selectivity; developing PDIA4-targeted agents could minimize off-target effects .

  • Longitudinal Studies: Assessing metformin’s impact on PDIA4 levels in diabetic cohorts is warranted .

Product Specs

Introduction
Protein disulfide isomerase A4 (PDIA4) is found within the endoplasmic reticulum lumen and acts as both a stress protein and a member of the protein disulfide isomerase family. It plays a crucial role in the catalytic rearrangement of disulfide bonds (S-S bonds) within proteins. Both PDIA4 and PDIA3 exhibit multifunctional properties, acting as proteases, protein disulfide isomerases, phospholipases, or a combination of these functions.
Description
Recombinant human PDIA4, expressed in E. coli, is a single polypeptide chain that lacks glycosylation. It comprises 646 amino acids, specifically residues 21 to 645, with a molecular weight of 72.9 kDa. The N-terminus of PDIA4 is fused to a 21 amino acid His-Tag. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
The solution is colorless and has been sterilized through filtration.
Formulation
The PDIA4 protein solution is provided at a concentration of 1 mg/ml and is formulated in a buffer consisting of 20 mM Tris-HCl (pH 8), 1 mM DTT, 0.1 M NaCl, and 10% glycerol.
Stability
For optimal storage, the product should be kept at 4°C if it will be used within 2-4 weeks. For extended storage, freezing at -20°C is recommended. To further enhance long-term stability, the addition of a carrier protein such as 0.1% HSA or BSA is advisable. It's important to avoid subjecting the product to repeated cycles of freezing and thawing.
Purity
The purity of the protein is determined to be greater than 90.0% based on SDS-PAGE analysis.
Biological Activity
The specific activity of the enzyme is measured to be greater than 10 A650/cm/min/mg. Enzymatic activity was confirmed by assessing the aggregation of insulin in the presence of DTT.
Synonyms
Endoplasmic reticulum resident protein 72, ERP70, ERP72.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MVAGAEGPDE DSSNRENAIE DEEEEEEEDD DEEEDDLEVK EENGVLVLND ANFDNFVADK DTVLLEFYAP WCGHCKQFAP EYEKIANILK DKDPPIPVAK IDATSASVLA SRFDVSGYPT IKILKKGQAV DYEGSRTQEE IVAKVREVSQ PDWTPPPEVT LVLTKENFDE VVNDADIILV EFYAPWCGHC KKLAPEYEKA AKELSKRSPP IPLAKVDATA ETDLAKRFDV SGYPTLKIFR KGRPYDYNGP REKYGIVDYM IEQSGPPSKE ILTLKQVQEF LKDGDDVIII GVFKGESDPA YQQYQDAANN LREDYKFHHT FSTEIAKFLK VSQGQLVVMQ PEKFQSKYEP RSHMMDVQGS TQDSAIKDFV LKYALPLVGH RKVSNDAKRY TRRPLVVVYY SVDFSFDYRA ATQFWRSKVL EVAKDFPEYT FAIADEEDYA GEVKDLGLSE SGEDVNAAIL DESGKKFAME PEEFDSDTLR EFVTAFKKGK LKPVIKSQPV PKNNKGPVKV VVGKTFDSIV MDPKKDVLIE FYAPWCGHCK QLEPVYNSLA KKYKGQKGLV IAKMDATAND VPSDRYKVEG FPTIYFAPSG DKKNPVKFEG GDRDLEHLSK FIEEHATKLS RTKEEL.

Q&A

What is PDIA4 and what is its primary function in human cells?

PDIA4 functions primarily as a molecular chaperone in the endoplasmic reticulum (ER), assisting in protein folding and quality control through the formation and rearrangement of disulfide bonds. It plays a crucial role during ER stress conditions when cells experience increased protein folding demands. Research has shown that PDIA4 is involved in the unfolded protein response (UPR) pathway and interacts with other ER stress proteins such as BiP/GRP78 and ATF4 . Beyond protein folding, PDIA4 participates in regulating apoptosis and stress response signaling, making it a multifunctional protein within cellular homeostasis mechanisms.

How is PDIA4 expression regulated in normal versus pathological states?

In normal tissues, PDIA4 maintains relatively lower expression levels compared to pathological states. Its expression is regulated by cellular stressors and metabolic status, with transcription factors associated with the unfolded protein response pathway playing key roles. Research indicates significant upregulation of PDIA4 in pathological conditions, particularly in cancer tissues. Analysis of TCGA dataset showed significantly higher PDIA4 mRNA expression in breast cancer tissues than in adjacent normal tissues . This expression difference is particularly pronounced in triple-negative breast cancer compared to other subtypes . Similar patterns have been observed in other malignancies, suggesting that dysregulation of PDIA4 expression is a common feature across various pathological states.

What experimental methods are most reliable for detecting active PDIA4 in human samples?

Multiple complementary techniques provide reliable detection of active PDIA4 in human samples:

  • Immunohistochemistry (IHC): This method allows visualization of PDIA4 in tissue sections using specific antibodies. The protocol involves fixing specimens in 4% formalin, embedding in paraffin, cutting into 4μm sections, and incubating with anti-PDIA4 antibodies (such as A07267 at 1:200 dilution) . Visualization uses HRP-conjugated secondary antibodies and diaminobenzidine as a chromogen .

  • Western blotting: Provides quantitative assessment of PDIA4 protein levels and has been successfully used to compare expression between tumor and adjacent normal tissues .

  • RT-PCR: Enables measurement of PDIA4 mRNA expression levels, allowing transcriptional regulation analysis .

  • Co-immunoprecipitation: Useful for studying protein-protein interactions involving PDIA4, such as its interaction with TAX1BP1 .

  • ELISA: Allows quantitative measurement of PDIA4 levels in serum samples, as used in studies correlating PDIA4 with insulin resistance markers .

Each method offers distinct advantages, and researchers should select techniques based on their specific experimental questions and sample types.

What evidence links PDIA4 to cancer progression, particularly in breast cancer?

Substantial evidence connects PDIA4 to breast cancer progression:

These findings collectively establish PDIA4 as an important factor in breast cancer biology with potential implications for prognosis and treatment response.

How does PDIA4 relate to insulin resistance and metabolic disorders?

Research has established significant connections between PDIA4 and metabolic dysfunction:

  • Human serum PDIA4 concentrations show positive correlations with insulin resistance markers. Patients in higher tertiles of HOMA-2 IR levels had significantly elevated PDIA4 levels compared to those in lower tertiles .

  • Statistical analysis revealed significant positive correlations between serum PDIA4 levels and numerous metabolic parameters including BMI, waist circumference, fasting glucose, insulin levels, HOMA-2 IR, HbA1C, and triglycerides, as shown in Table 3 :

ParameterCorrelation with Serum PDIA4
BMI (kg/m²)r=0.405, p<0.001
Waist circumference (cm)r=0.390, p<0.001
Fasting glucose (mg/dL)r=0.194, p<0.001
Fasting insulin (μIU/mL)r=0.370, p<0.001
HOMA-2IRr=0.274, p<0.001
HbA1C (%)r=0.176, p<0.001
Triglycerides (mg/dL)r=0.327, p<0.001
  • PDIA4 also correlated positively with inflammatory markers like hsCRP (r=0.253, p<0.001) and IL-6 (r=0.154, p=0.001) .

  • In vitro studies using C2C12 muscle cells demonstrated that palmitate treatment (which induces insulin resistance) significantly increased PDIA4 expression alongside ER stress markers BiP/GRP78 and ATF4 .

  • The relationship appears bidirectional, as insulin treatment in palmitate-challenged cells further increased PDIA4 and ER stress marker expression .

These findings position PDIA4 as a potential mediator connecting ER stress to insulin resistance mechanisms in metabolic disorders.

What mechanisms underlie PDIA4's role in treatment resistance in cancer?

PDIA4 contributes to treatment resistance in cancer through several mechanisms:

  • Apoptosis regulation: PDIA4 modulates caspase 3 and 7 activity, critical enzymes in the execution phase of cell death. By regulating these apoptotic pathways, PDIA4 can potentially help cancer cells evade treatment-induced death .

  • Mitochondrial apoptosis inhibition: Research has shown that PDIA4 activation inhibits classical mitochondrial apoptosis in cancer cells, while its inactivation can restore this pathway . This suggests PDIA4 influences the intrinsic apoptotic pathway involving mitochondrial membrane permeabilization.

  • Chemotherapy resistance: Studies have specifically linked PDIA4 activation to docetaxel resistance in prostate cancer , indicating its role in reducing sensitivity to chemotherapeutic agents.

  • Radiotherapy resistance: PDIA4 has been implicated in radiation therapy resistance, particularly in triple-negative breast cancer . This suggests PDIA4 may influence DNA damage response pathways and cellular survival mechanisms.

  • Protein interaction networks: Co-immunoprecipitation studies identified interaction between PDIA4 and TAX1BP1 , suggesting PDIA4 may influence multiple cellular pathways through protein-protein interactions that ultimately contribute to treatment resistance.

These mechanisms collectively position PDIA4 as a potential therapeutic target for overcoming treatment resistance in cancer.

What are the optimal experimental conditions for studying PDIA4 activity in cell culture systems?

Based on established protocols, optimal experimental conditions for studying PDIA4 include:

For skeletal muscle cells (C2C12) :

  • Maintenance medium: Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum and antibiotics

  • Differentiation conditions: DMEM containing 2% heat-inactivated horse serum for at least four days

  • Cell passage considerations: Use passages 3-5 at 80% confluency

  • Medium renewal: Update medium before experimentation

  • Treatment conditions: For insulin resistance models, palmitate treatment followed by insulin stimulation

For breast cancer studies (using cell lines like MDA-MB-231) :

  • Appropriate knockdown models (e.g., shRNA targeting PDIA4)

  • Radiation treatment protocols for studying radioresistance

  • Protein interaction studies using co-immunoprecipitation

General considerations across model systems:

  • Include appropriate controls (vehicle, scrambled shRNA)

  • Standardize incubation times and treatment concentrations

  • Monitor cell viability throughout experiments

  • Perform time-course experiments to capture dynamic changes in PDIA4 expression

  • Validate findings using multiple detection methods (western blot, RT-PCR, immunofluorescence)

These conditions provide a foundation for reliable investigation of PDIA4 function in cellular models.

How should researchers design co-immunoprecipitation experiments to study PDIA4 protein interactions?

Designing effective co-immunoprecipitation (co-IP) experiments for PDIA4 interactions requires careful consideration of several methodological aspects:

  • Lysate preparation: Cell lysates should be prepared using buffers that maintain protein-protein interactions while efficiently extracting PDIA4 from the ER membrane. Research protocols have successfully used protein A/G agarose beads following manufacturer's instructions .

  • Antibody selection: Choose high-specificity antibodies against PDIA4 or its potential binding partners. For target verification experiments like the PDIA4-TAX1BP1 interaction , antibodies against both proteins should be validated beforehand.

  • Controls: Include essential controls: (1) an isotype-matched control antibody to assess non-specific binding, (2) input control (pre-immunoprecipitation lysate) to confirm target protein presence, (3) reverse co-IP where possible (precipitate with antibody against the binding partner and probe for PDIA4).

  • Detection method: Western blotting is the standard approach for detecting co-immunoprecipitated proteins, using antibodies that recognize epitopes different from those used for precipitation.

  • Crosslinking consideration: For transient or weak interactions, consider using chemical crosslinkers to stabilize protein complexes prior to lysis.

  • Validation approaches: Confirm interactions using complementary methods such as proximity ligation assay, FRET, or pull-down assays with recombinant proteins.

  • Quantification: Include quantitative assessment of co-precipitated proteins relative to input and control samples.

This systematic approach ensures reliable identification and characterization of genuine PDIA4 protein interactions.

What animal models are most appropriate for studying PDIA4 function in vivo?

Based on research methodologies, the following animal models are appropriate for studying PDIA4 function in vivo:

For cancer studies:

  • BALB/c nude mice have been successfully used for xenograft models to study PDIA4's role in tumor growth and radiation resistance . These immunodeficient mice allow human cancer cell implantation, such as PDIA4 knockdown MDA-MB-231 cells (shRNA3) versus control cells (shNC) .

  • Experimental design typically involves subcutaneous injection in the right thigh, with tumor growth monitoring using calipers and volume calculation using the formula (width² × length)/2 .

  • Sample size recommendation: Five mice per group provided sufficient statistical power in published research .

For metabolic studies:

  • C57BL/6J mice are appropriate for investigating PDIA4's role in insulin resistance and metabolic disorders .

  • Experimental approaches include feeding mice either standard chow (10% kcal from fat), high-fat diet (60% kcal from fat), or high-fat diet with pharmacological interventions (e.g., metformin at 200 mg/kg, intraperitoneal) .

  • Study duration typically extends to 16 weeks on specialized diets .

  • Insulin tolerance testing involves fasting mice for 6 hours, administering intraperitoneal insulin (1 U/kg body weight), and measuring blood glucose at multiple timepoints (0, 20, 40, 60, 80, 100 minutes) .

  • Sample size recommendation: Ten mice per group was used in published metabolic studies .

Key considerations for both models include appropriate ethics committee approval, proper control groups, standardized measurement protocols, and validated tissue analysis methods including immunohistochemistry.

How does PDIA4 interact with the endoplasmic reticulum stress response in pathological conditions?

PDIA4 demonstrates complex interactions with the endoplasmic reticulum stress response in pathological conditions:

  • Co-regulation with ER stress markers: In palmitate-induced insulin resistance models, PDIA4 expression increases concurrently with established ER stress markers BiP/GRP78, ATF4, and CHOP . This coordinated regulation suggests PDIA4 is an integral component of the cellular stress response.

  • Unfolded Protein Response (UPR) pathway involvement: The co-upregulation of ATF4 alongside PDIA4 indicates activation of the PERK branch of the UPR , suggesting PDIA4 regulation may occur through this pathway during pathological stress conditions.

  • Stress amplification mechanisms: In experimental models, combining stressors (e.g., palmitate plus insulin) further increased PDIA4 expression alongside other ER stress markers compared to single stressors alone . This suggests PDIA4 participates in stress response amplification loops.

  • Inflammatory pathway connection: The significant positive correlation between serum PDIA4 levels and inflammatory markers (hsCRP: r=0.253, p<0.001; IL-6: r=0.154, p=0.001) suggests PDIA4 may link ER stress to inflammatory processes in pathological states.

  • Tissue-specific responses: PDIA4 upregulation has been observed across multiple tissue types in pathological conditions, including breast cancer tissues and metabolically stressed skeletal muscle , indicating a conserved role in ER stress response across different disease contexts.

These intricate relationships position PDIA4 as a potential mediator connecting ER stress to disease progression in both cancer and metabolic disorders.

What signaling pathways are most significantly affected by PDIA4 modulation in cancer cells?

Based on research findings, several significant signaling pathways are affected by PDIA4 modulation in cancer cells:

  • Apoptotic signaling: PDIA4 regulates caspase 3 and 7 activity, which are critical executioner enzymes in programmed cell death pathways . Modulation of PDIA4 expression in cancer cells directly affects their ability to undergo apoptosis.

  • Mitochondrial death pathway: PDIA4 activation has been shown to inhibit classical mitochondrial apoptosis in cancer cells, while its inactivation can restore this pathway . This indicates PDIA4 influences the intrinsic apoptotic pathway involving mitochondrial outer membrane permeabilization.

  • Treatment resistance mechanisms: PDIA4 has been implicated in docetaxel resistance and potentially radiotherapy resistance in triple-negative breast cancer. This suggests involvement in DNA damage response pathways and cellular survival mechanisms under treatment stress.

  • Protein-protein interaction networks: Co-immunoprecipitation studies identified interaction between PDIA4 and TAX1BP1 , suggesting PDIA4 may influence multiple cellular pathways through protein-protein interactions, potentially including NF-κB signaling or autophagy regulation.

  • Stress response pathways: As an ER stress protein, PDIA4 likely influences unfolded protein response signaling cascades that can promote cancer cell survival under adverse conditions.

These pathways collectively position PDIA4 as a central node connecting stress responses to cell survival mechanisms in cancer, with significant implications for treatment resistance and disease progression.

What are the technical challenges in developing specific PDIA4 inhibitors for research applications?

Developing specific PDIA4 inhibitors for research applications faces several significant technical challenges:

  • Structural hurdles: PDIA4 shares considerable structural similarity with other PDI family members, making selectivity difficult to achieve without affecting related proteins. Its multiple thioredoxin-like domains with distinct functions further complicate target-specific inhibitor design.

  • Active site characteristics: The catalytic cysteines in PDIA4's active site exist in different redox states depending on the cellular environment and functional state, requiring inhibitors that can appropriately interact with these dynamic sites.

  • Subcellular localization barriers: PDIA4 primarily localizes to the ER lumen , requiring inhibitors that can penetrate cell membranes and reach the appropriate subcellular compartment while maintaining specificity.

  • Redox environment considerations: The oxidizing environment of the ER differs significantly from the cytosol, potentially affecting inhibitor stability, activity, and specificity once it reaches the target location.

  • Validation complexities: Establishing appropriate assays to verify PDIA4-specific inhibition versus general PDI family inhibition presents a significant challenge. Researchers must develop methods to distinguish between direct PDIA4 inhibition effects and general ER stress responses.

  • Physiological function preservation: PDIA4 plays important roles in normal protein folding, requiring development of inhibitors that selectively target pathological functions while preserving essential physiological activities.

Addressing these challenges requires integrative approaches combining structural biology insights, computational modeling, medicinal chemistry optimization, and comprehensive validation strategies.

How might PDIA4 serve as a biomarker in cancer diagnosis or prognosis?

PDIA4 shows significant potential as a biomarker in cancer diagnosis and prognosis:

For clinical implementation, several considerations are important:

  • Standardization of detection methods (IHC, RT-PCR, or ELISA)

  • Establishment of optimal expression cut-off values for prognostic stratification

  • Integration into multi-marker panels to enhance predictive power

  • Validation through large-scale, multicenter clinical studies

These findings position PDIA4 as a promising biomarker candidate with potential applications in cancer diagnosis, prognosis, and treatment planning.

What therapeutic strategies could target PDIA4 in metabolic diseases?

Research findings suggest several therapeutic strategies targeting PDIA4 in metabolic diseases:

  • Direct PDIA4 inhibition: The positive correlation between PDIA4 levels and insulin resistance markers (HOMA-2IR) suggests that specific inhibitors targeting PDIA4 activity could potentially enhance insulin sensitivity. This approach would require developing selective inhibitors that can reach the ER compartment.

  • ER stress pathway modulation: Given PDIA4's co-regulation with other ER stress markers (BiP/GRP78, ATF4, CHOP) , broader interventions targeting the UPR pathway could modulate PDIA4 activity as part of a comprehensive approach to reducing ER stress in metabolic tissues.

  • Combination with established metabolic drugs: Research has investigated metformin administration in high-fat diet mouse models , suggesting investigation into whether established metabolic drugs modulate PDIA4 activity as part of their mechanism. New combination approaches could be developed.

  • Targeting inflammatory connections: The significant correlation between PDIA4 and inflammatory markers (hsCRP, IL-6) suggests that anti-inflammatory interventions might indirectly modulate PDIA4-related pathways, offering alternative therapeutic approaches.

  • Chemical chaperone therapy: Compounds that assist protein folding and reduce ER stress (e.g., 4-phenylbutyric acid, tauroursodeoxycholic acid) might reduce pathological PDIA4 activation while preserving its physiological functions.

  • Nutritional interventions: Given PDIA4's response to palmitate treatment , dietary interventions reducing lipotoxicity might influence PDIA4-mediated pathways in metabolic diseases.

These diverse approaches offer multiple avenues for therapeutic development, potentially addressing the complex role of PDIA4 in metabolic dysfunction.

How should researchers interpret conflicting results in PDIA4 expression studies across different disease models?

When interpreting conflicting PDIA4 expression results across different disease models, researchers should consider several methodological and biological factors:

  • Tissue-specific regulation: PDIA4 expression and function may be regulated differently in various tissues due to tissue-specific transcription factors, epigenetic modifications, or microRNA networks. The positive correlations observed between PDIA4 and metabolic parameters in human studies might not translate equally across all tissue types.

  • Disease context variation: PDIA4's upregulation in breast cancer versus its role in metabolic disorders may involve different regulatory mechanisms and functional consequences, even if both show elevated expression patterns.

  • Methodological differences: Variations in detection methods (IHC, western blot, RT-PCR) can yield different results. The studies employed multiple techniques including western blotting, RT-PCR, and immunohistochemistry , which each have different sensitivities and specificities.

  • Sample processing impact: Tissue collection, storage, and processing protocols can affect protein and RNA integrity. Standardized protocols as described in the immunohistochemistry methods are essential for comparable results.

  • Reference standards: Different normalization strategies can influence interpretation of expression data. Studies should clearly report housekeeping genes or proteins used for normalization.

Resolution strategies include:

  • Employing multiple detection methods within the same study

  • Correlating expression with functional assays

  • Conducting careful validation using diverse antibodies/primers

  • Considering broader pathway analysis rather than isolated PDIA4 expression

  • Performing meta-analyses across multiple studies with standardized reporting

These approaches help distinguish genuine biological differences from methodological artifacts in PDIA4 expression studies.

What genomic and proteomic approaches could advance understanding of PDIA4 regulation?

Advanced genomic and proteomic approaches could significantly enhance understanding of PDIA4 regulation:

  • Single-cell RNA sequencing: This technology could reveal cell-type specific expression patterns of PDIA4 within heterogeneous tissues, providing insights into which specific cell populations show differential regulation in disease states like cancer or metabolic disorders .

  • CRISPR-Cas9 screening: Genome-wide CRISPR screens could identify novel regulators of PDIA4 expression and function, revealing upstream pathways that control its activity in different contexts.

  • ChIP-seq analysis: Chromatin immunoprecipitation sequencing could identify transcription factors binding to the PDIA4 promoter region under different conditions, elucidating transcriptional regulation mechanisms.

  • Proteomics interaction mapping: Mass spectrometry-based interactome analysis could extend beyond the identified TAX1BP1 interaction to comprehensively map PDIA4's protein interaction network across different cellular contexts.

  • Post-translational modification profiling: Analysis of PDIA4's post-translational modifications could reveal regulatory mechanisms beyond transcriptional control, potentially identifying sites for therapeutic intervention.

  • Ribosome profiling: This technique could assess translational regulation of PDIA4 under different stress conditions, providing insights into protein synthesis control mechanisms.

  • Spatial transcriptomics/proteomics: These emerging technologies could reveal tissue microenvironment influences on PDIA4 expression patterns, particularly valuable in heterogeneous tissues like tumors.

These advanced approaches would provide a multi-dimensional understanding of PDIA4 regulation, potentially revealing new therapeutic targets and biomarker applications.

How might PDIA4 function differ between acute and chronic stress conditions?

PDIA4 function likely differs significantly between acute and chronic stress conditions:

  • Temporal expression patterns: In acute stress, PDIA4 may show transient upregulation as part of the immediate unfolded protein response, whereas chronic stress might lead to sustained elevation with different downstream consequences. The research showing PDIA4 upregulation in established disease states reflects mostly chronic conditions.

  • Adaptive versus maladaptive roles: During acute stress, PDIA4 upregulation likely serves an adaptive role in managing increased protein folding demands and maintaining ER homeostasis. In chronic stress scenarios, persistent PDIA4 elevation might become maladaptive, contributing to pathological processes like apoptosis resistance in cancer or sustained insulin resistance .

  • Interaction network dynamics: The protein interaction partners of PDIA4 may differ between acute and chronic stress conditions. The identified interaction with TAX1BP1 might represent a chronic stress adaptation rather than an acute response pattern.

  • Subcellular localization shifts: Chronic stress might induce changes in PDIA4's subcellular distribution not observed during acute stress, potentially affecting its functional impact on cellular processes.

  • Post-translational modification differences: Acute versus chronic stress might result in different post-translational modification patterns of PDIA4, altering its activity, stability, or interaction partners.

Experimental approaches to investigate these differences would include:

  • Time-course studies comparing PDIA4 expression, localization, and activity at different stress exposure durations

  • Pulse-chase experiments to assess PDIA4 protein turnover rates under different stress conditions

  • Comparative interactome analysis between acute and chronic stress models

  • In vivo models comparing acute stress response to chronic disease states

Understanding these temporal differences would provide valuable insights for designing therapeutic interventions targeting PDIA4.

What interdisciplinary approaches could accelerate translation of PDIA4 research to clinical applications?

Accelerating translation of PDIA4 research to clinical applications requires strategic interdisciplinary approaches:

  • Biomarker development partnerships: Collaboration between basic researchers, clinical pathologists, and biostatisticians could validate PDIA4 as a cancer prognostic marker or metabolic risk indicator . This would involve standardizing detection methods, establishing clinical cutoff values, and conducting multicenter validation studies.

  • Medicinal chemistry and structural biology integration: Combining protein structure analysis with medicinal chemistry expertise could address the technical challenges in developing specific PDIA4 inhibitors, potentially overcoming selectivity issues against other PDI family members.

  • Systems biology modeling: Computational modeling of PDIA4's role within larger biological networks could identify optimal points for therapeutic intervention and predict potential side effects, integrating data from both cancer and metabolic disease studies .

  • Bioengineering approaches: Developing targeted drug delivery systems to reach the ER compartment could overcome the subcellular localization challenges for PDIA4-directed therapeutics.

  • Clinical-basic science feedback loops: Establishing pipelines for rapid translation of clinical observations back to laboratory investigation would accelerate understanding of PDIA4's roles in human disease. The correlations observed between PDIA4 and clinical parameters provide a foundation for such approaches.

  • Multi-omics patient profiling: Integrating genomic, proteomic, and metabolomic data from patient samples could identify subpopulations most likely to benefit from PDIA4-targeted approaches, enabling precision medicine applications.

  • Regulatory science collaboration: Early engagement with regulatory experts could streamline the path to clinical application, identifying critical validation requirements for PDIA4-based diagnostics or therapeutics.

These interdisciplinary strategies would address the complex challenges in translating PDIA4 research findings into meaningful clinical applications for both cancer and metabolic diseases.

Product Science Overview

Structure and Function

PDIA4 is characterized by its N-terminal endoplasmic reticulum (ER) signal sequence, three catalytically active thioredoxin (TRX) domains, two TRX-like domains, and a C-terminal ER-retention sequence . These domains enable PDIA4 to function as a protein folding chaperone, ensuring that proteins achieve their correct conformation within the ER .

When bound to cyclophilin B, PDIA4 enhances the rate of immunoglobulin G (IgG) intermolecular disulfide bonding and antibody assembly . This function is particularly important in the immune response, as it ensures the proper assembly and functionality of antibodies.

Role in Disease

PDIA4 has been implicated in various diseases, particularly in cancer. Its overexpression has been associated with poor prognosis in glioblastoma multiforme (GBM), a highly aggressive brain tumor . PDIA4 promotes angiogenesis (the formation of new blood vessels) in GBM, which supports tumor growth and survival under harsh conditions . Additionally, PDIA4 has been found to confer resistance to anti-angiogenic therapy, making it a potential target for improving cancer treatment outcomes .

Recombinant PDIA4

Recombinant PDIA4 is produced using various expression systems, including E. coli, yeast, baculovirus, and mammalian cells . The recombinant form retains the enzymatic activity of the native protein, making it valuable for research and therapeutic applications. It is used in studies to understand the molecular mechanisms of protein folding and its role in diseases, as well as in the development of potential therapeutic interventions.

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 2024 Thebiotek. All Rights Reserved.