PDIA3 Human, Active

Protein Disulfide Isomerase A3 Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

PDIA3 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 518 amino acids (25-505 a.a.) and having a molecular wieght of 58.5 kDa. The PDIA3 is fused to 37 a.a. His-Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Protein disulfide-isomerase A3 (PDIA3), also known as ERp57, is an enzyme primarily located in the endoplasmic reticulum (ER). It plays a crucial role in protein folding and quality control by catalyzing the formation, rearrangement, and breakage of disulfide bonds in newly synthesized glycoproteins. PDIA3 interacts with lectin chaperones calreticulin and calnexin to facilitate proper protein folding. It is involved in various cellular processes, including MHC class I antigen presentation, calcium homeostasis, and response to cellular stress. Dysregulation of PDIA3 has been implicated in several diseases, including cancer and neurodegenerative disorders.
Description
Recombinant human PDIA3 protein was expressed in E. coli and purified to a high degree. This protein is a single, non-glycosylated polypeptide chain consisting of 518 amino acids (residues 25-505) with a His-tag fused at the N-terminus. The molecular weight of the protein is approximately 58.5 kDa.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation
The PDIA3 protein is supplied in a solution containing 20mM Tris-HCl buffer (pH 8.0), 1mM DTT, 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the PDIA3 protein should be stored at 4°C. For long-term storage, it is recommended to store the protein at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of the PDIA3 protein is greater than 95% as determined by SDS-PAGE analysis.
Biological Activity
The PDIA3 protein exhibits a specific activity exceeding 20 A650/cm/min/mg. This activity was validated by assessing the protein's ability to catalyze insulin aggregation in the presence of DTT.
Synonyms
ERp57, ERp60, ERp61, GRP57, GRP58, HsT17083, P58, PI-PLC, ER60, Protein disulfide-isomerase A3, Disulfide isomerase ER-60, Endoplasmic reticulum resident protein 60, ER protein 60, 58 kDa microsomal protein, Endoplasmic reticulum resident protein 57, ER protein 57, 58 kDa glucose-regulated protein, PDIA3.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMSDV LELTDDNFES RISDTGSAGL MLVEFFAPWC GHCKRLAPEY EAAATRLKGI VPLAKVDCTA NTNTCNKYGV SGYPTLKIFR DGEEAGAYDG PRTADGIVSH LKKQAGPASV PLRTEEEFKK FISDKDASIV GFFDDSFSEA HSEFLKAASN LRDNYRFAHT NVESLVNEYD DNGEGIILFR PSHLTNKFED KTVAYTEQKM TSGKIKKFIQ ENIFGICPHM TEDNKDLIQG KDLLIAYYDV DYEKNAKGSN YWRNRVMMVA KKFLDAGHKL NFAVASRKTF SHELSDFGLE STAGEIPVVA IRTAKGEKFV MQEEFSRDGK ALERFLQDYF DGNLKRYLKS EPIPESNDGP VKVVVAENFD EIVNNENKDV LIEFYAPWCG HCKNLEPKYK ELGEKLSKDP NIVIAKMDAT ANDVPSPYEV RGFPTIYFSP ANKKLNPKKY EGGRELSDFI SYLQREATNP PVIQEEKPKK KKKAQEDL.

Q&A

What is PDIA3 and what are its fundamental biological functions?

PDIA3, also known as protein disulfide-isomerase A3, is a 58.5 kDa (518 amino acids) protein primarily localized in the endoplasmic reticulum. Its fundamental functions include:

  • Protein folding assistance through interactions with lectin chaperones calreticulin and calnexin to modulate folding of newly synthesized glycoproteins

  • Enzymatic activity as a protein disulfide isomerase, catalyzing formation, reduction, and isomerization of disulfide bonds during protein folding

  • Critical participation in the major histocompatibility complex (MHC) class I peptide-loading complex (TAP1), essential for formation of the final antigen conformation and export from the endoplasmic reticulum to cell surface

  • Involvement in immunological processes including antigen presentation and immune cell function regulation

The protein contains active sites with CGHCK motifs characteristic of the thioredoxin family and demonstrates specific activity of >20 A650/cm/min/mg when measuring insulin aggregation in the presence of DTT .

How can researchers accurately measure PDIA3 enzymatic activity?

Accurate measurement of PDIA3 enzymatic activity requires consideration of several methodological approaches:

  • Insulin turbidity assay:

    • The standard approach measures PDIA3's ability to catalyze insulin disulfide bond reduction in the presence of DTT

    • Specific activity should exceed 20 A650/cm/min/mg for properly folded active protein

    • Monitor aggregation spectrophotometrically at 650 nm with time-course measurements

  • Storage and handling considerations:

    • Use appropriate buffer conditions: 20 mM Tris-HCl buffer (pH 8.0) containing 1mM DTT, 0.1 M NaCl, and 10% glycerol

    • For short-term use (1-2 weeks), store at 4°C

    • For long-term storage, aliquot and maintain at -20°C or -70°C

    • Avoid repeated freeze-thaw cycles that can dramatically reduce activity

  • Quality control parameters:

    • Confirm purity >95% by SDS-PAGE before activity measurements

    • Verify molecular mass (58.5 kDa) by techniques such as MALDI-TOF

    • Consider the redox state of the protein preparation, as this significantly impacts measured activity

What molecular mechanisms underlie PDIA3's role in normal physiology versus pathological conditions?

PDIA3 functions through several distinct molecular mechanisms that can become dysregulated in pathological conditions:

  • In normal physiology:

    • Interacts with calreticulin and calnexin to form a quality control complex for glycoprotein folding in the ER

    • Facilitates the formation of the final antigen conformation in MHC class I peptide-loading complex

    • Maintains appropriate oxidative folding of proteins in the secretory pathway

  • In pathological conditions:

    • Becomes involved in tumor microenvironment modulation through interactions with immune and stromal cells

    • Correlates with genomic alterations including loss of PTEN and amplification of EGFR in high-expression gliomas

    • Associates with IDH mutations in approximately 80% of low PDIA3 expression gliomas

    • Participates in immune regulatory processes that can suppress anti-tumor immunity through multiple pathways

    • Drives immune autoreactivity in inflammatory liver conditions, promoting TH1- and TH17-polarized responses

    • Contributes to the generation of pathogenic antibodies that can exacerbate tissue damage in autoimmune contexts

These mechanisms make PDIA3 a potential biomarker and therapeutic target in various diseases, particularly those involving immune dysregulation or cancer progression.

How should researchers analyze relationships between PDIA3 expression and immune infiltration in tumor microenvironments?

Analysis of PDIA3's relationship with immune infiltration requires a comprehensive multi-method approach:

In glioma research, this approach revealed PDIA3 positively correlates with diverse infiltrating immune cell types, with particularly strong associations with macrophages and T cells as confirmed by single-cell sequencing analysis .

What methodologies are optimal for investigating PDIA3's role in T cell-mediated immune responses?

Investigating PDIA3's role in T cell-mediated immunity requires specialized approaches:

  • Gene Set Variation Analysis (GSVA):

    • Apply GSVA to identify correlations between PDIA3 and specific T cell functions, including:

      • Negative regulation of T cell mediated cytotoxicity

      • Negative regulation of T cell proliferation

      • Antigen processing and presentation via MHC class I molecules

      • Regulation of T cell activation and tolerance induction

      • T helper cell differentiation and cytokine production

    • Validate findings across multiple independent cohorts (e.g., TCGA and CGGA databases)

  • Functional validation studies:

    • Isolate T cell populations from relevant tissues for ex vivo analysis

    • Perform flow cytometry to characterize PDIA3 expression in different T cell subsets

    • Assess correlation with activation/exhaustion markers and cytokine production profiles

  • Experimental manipulation:

    • Conduct gain/loss-of-function experiments with PDIA3 in T cells

    • Evaluate changes in proliferation, cytotoxicity, and cytokine production

    • Test impact on antigen recognition and immune synapse formation

  • Translational applications:

    • Correlate findings with response to immunotherapies targeting T cell function

    • Evaluate PDIA3 as a predictive biomarker for immunotherapy response

    • Consider targeting PDIA3-mediated pathways to enhance anti-tumor immunity

These methods have revealed that PDIA3 may serve as a crucial mediator in suppressing T cell-related anti-tumor immune response in the glioma microenvironment while simultaneously affecting processes like antigen presentation and T cell differentiation .

How can researchers effectively use single-cell sequencing to understand PDIA3 function in heterogeneous tissue samples?

Single-cell sequencing offers powerful insights into PDIA3's cell type-specific roles in complex tissues:

  • Experimental design considerations:

    • Use fresh tissue dissociation protocols optimized to preserve cell viability

    • Include controls to account for batch effects and patient-specific variations

    • Achieve sufficient depth (>50,000 reads per cell) for reliable PDIA3 detection

  • Analysis workflow:

    • Implement clustering algorithms to identify distinct cell populations

    • Regress out patient effects to focus on biological variation

    • Create expression landscapes showing PDIA3 distribution across cell types

    • Quantify the proportion of PDIA3-high cells within each identified cluster

  • Validation and extension:

    • Confirm key findings using orthogonal methods (immunohistochemistry, flow cytometry)

    • Integrate with spatial transcriptomics to preserve tissue context information

    • Compare expression patterns between healthy and diseased tissues

  • Data interpretation:

    • Examine correlation with known cell type-specific markers

    • Assess potential functional implications based on the cell types enriched for PDIA3

    • Consider developmental lineage relationships between PDIA3-expressing populations

In glioma research, this approach successfully identified eight distinct cell clusters, revealing that high PDIA3 expression was particularly enriched in macrophages and T cells . This cell type-specific information provides crucial context for understanding PDIA3's functional roles in the tumor microenvironment.

How does PDIA3 expression correlate with clinical features and genomic alterations in gliomas?

PDIA3 expression demonstrates significant correlations with multiple clinical and genomic features in gliomas:

  • Clinical correlations:

    • Associated with higher grade gliomas and worse clinical outcomes

    • Functions as a potential prognostic biomarker in glioma patients

    • Correlates with inflammation signatures in the tumor microenvironment

  • Genomic alteration patterns:

    • High PDIA3 expression gliomas show distinctive mutation patterns:

      • Loss of tumor suppressor PTEN

      • Amplification of oncogenic driver EGFR

      • Chromosomal instability signatures

    • Low PDIA3 expression gliomas demonstrate:

      • Enrichment for IDH mutations (approximately 80% of cases)

      • Generally less genomic instability

      • Different prognostic trajectory

  • Molecular pathway associations:

    • Involvement in immune regulatory processes

    • Correlation with other immune checkpoint inhibitors including HAVCR2, CD274, CD276, CD80, IDO1, PDCD1, CTLA-4, and PDCD1LG2

    • Participation in inflammation-related signaling networks

These correlations suggest PDIA3 may be involved in fundamental tumorigenic processes and could represent a molecular target for prognosis and immune-related treatment strategies in gliomas .

What is the evidence for PDIA3's involvement in autoimmune and inflammatory conditions?

PDIA3 demonstrates significant involvement in autoimmune and inflammatory conditions through several mechanisms:

  • Epitope-driven immune autoreactivity:

    • Specific PDIA3 epitopes can drive TH1- and TH17-polarized immune responses in the liver

    • These polarized responses contribute to tissue damage and disease progression

    • The autoreactive response shows specificity for particular PDIA3 regions

  • Pathogenic antibody development:

    • Generation of anti-PDIA3 antibodies that undergo isotype switching from IgM to IgG3

    • This isotype switch indicates a mature, T cell-dependent antibody response

    • These antibodies can exacerbate hepatocyte death when transferred to susceptible hosts

  • Clinical evidence from human patients:

    • Increased humoral responses to PDIA3 observed in patients with:

      • Autoimmune hepatitis

      • Primary biliary cholangitis

      • Type 2 diabetes with hepatic manifestations

    • These findings support the clinical relevance of PDIA3 autoreactivity

  • Mechanistic insights from experimental models:

    • Metabolic insults (e.g., high-fat, high-fructose diet) can trigger PDIA3-directed autoimmunity

    • Passive transfer of PDIA3-specific T cells exacerbates metabolic liver damage

    • This provides a causative link between PDIA3 autoreactivity and disease progression

These findings suggest PDIA3 may function as both a biomarker and a mechanistic contributor to autoimmune and inflammatory pathology, particularly in metabolic and liver diseases .

How might PDIA3 serve as a therapeutic target in cancer and inflammatory diseases?

PDIA3's diverse functions position it as a promising therapeutic target through several potential mechanisms:

  • In cancer therapy approaches:

    • Targeting PDIA3's role in the tumor microenvironment:

      • Modulation of interactions with infiltrating immune and stromal cells

      • Combination with other immune checkpoint inhibitors for enhanced efficacy

    • Exploiting PDIA3's association with specific genomic alterations:

      • Developing synthetic lethal approaches with PTEN loss or EGFR amplification

      • Stratifying patients based on PDIA3 expression for personalized therapy

    • Addressing PDIA3's immunomodulatory functions:

      • Enhancing T cell-mediated anti-tumor responses by blocking PDIA3's suppressive effects

      • Manipulating antigen presentation pathways dependent on PDIA3 function

  • In inflammatory disease contexts:

    • Targeting epitope-specific autoimmune responses:

      • Developing epitope-specific tolerization strategies

      • Blocking TH1/TH17 polarization driven by PDIA3 autoreactivity

    • Neutralizing pathogenic anti-PDIA3 antibodies:

      • Developing decoy molecules to sequester harmful antibodies

      • Blocking antibody-dependent cellular cytotoxicity mechanisms

    • Addressing PDIA3's enzymatic functions:

      • Developing selective inhibitors of PDIA3's disulfide isomerase activity

      • Modulating protein folding stress responses in affected tissues

  • Translational considerations:

    • Development of biomarker assays to identify patients likely to benefit from PDIA3-targeted therapies

    • Design of combinatorial approaches with established treatment modalities

    • Careful assessment of potential off-target effects given PDIA3's fundamental cellular roles

These approaches acknowledge PDIA3's multifaceted roles across different disease contexts and suggest tailored therapeutic strategies based on specific pathophysiological mechanisms .

What are the optimal conditions for producing and maintaining active recombinant human PDIA3 protein?

Production and maintenance of active recombinant human PDIA3 requires careful attention to several technical parameters:

  • Expression system optimization:

    • E. coli expression systems can produce functional human PDIA3 with appropriate yields

    • N-terminal His-tag fusion enables efficient purification while preserving activity

    • Purification through conventional chromatography techniques produces high purity (>95%)

  • Buffer composition for maximal stability:

    • Optimal buffer: 20 mM Tris-HCl (pH 8.0) containing:

      • 1 mM DTT (to maintain reduced active site cysteines)

      • 0.1 M NaCl (for ionic strength)

      • 10% glycerol (as a stabilizing agent)

    • This composition maintains enzyme activity while preventing aggregation

  • Storage conditions:

    • Short-term storage (1-2 weeks): 4°C

    • Long-term storage: Aliquot and maintain at -20°C or -70°C

    • Avoid repeated freeze-thaw cycles that significantly reduce enzymatic activity

  • Quality control parameters:

    • Confirm molecular mass (58.5 kDa) using techniques such as MALDI-TOF

    • Verify enzymatic activity: specific activity >20 A650/cm/min/mg using insulin aggregation assay

    • Assess purity through SDS-PAGE (should exceed 95%)

    • Validate protein folding through circular dichroism or thermal stability assays

These optimized conditions ensure that recombinant PDIA3 maintains its native conformation and enzymatic function, critical for reliable experimental results in both biochemical and cellular studies .

What experimental controls are essential when studying PDIA3 function in complex biological systems?

Robust investigation of PDIA3 function requires implementation of several critical experimental controls:

  • Specificity controls:

    • Include other PDI family members as comparators to establish PDIA3-specific effects

    • Use siRNA/shRNA with validated specificity or CRISPR/Cas9 knockout systems

    • Implement rescue experiments with wild-type PDIA3 to confirm phenotype causality

    • Employ catalytically inactive mutants (C57S/C60S) to distinguish enzymatic from structural functions

  • System-appropriate controls:

    • For tumor microenvironment studies:

      • Include multiple cancer types/grades to account for heterogeneity

      • Compare with matched normal tissue from the same patient

      • Stratify analyses by known molecular subtypes (e.g., IDH-mutant vs. wild-type gliomas)

    • For autoimmune models:

      • Include appropriate diet controls in metabolic disease models

      • Compare multiple inflammatory conditions to distinguish disease-specific effects

      • Include time-course analyses to capture disease progression

  • Technical validation approaches:

    • Employ multiple detection methods for PDIA3 (antibody-based and mRNA-based)

    • Validate key findings across independent patient cohorts (e.g., TCGA and CGGA)

    • Use orthogonal techniques to confirm observations (e.g., validate RNAseq with qPCR)

  • Translational validation:

    • Confirm findings from model systems in human patient samples

    • Stratify analyses based on clinical parameters and treatment history

    • Consider potential confounding factors like age, sex, and comorbidities

These comprehensive controls ensure that experimental observations are specifically attributable to PDIA3 function rather than experimental artifacts or broader PDI family effects, critical for accurate interpretation of results in complex biological contexts .

How can researchers effectively analyze seemingly contradictory findings about PDIA3 function across different experimental models?

Analyzing contradictory findings about PDIA3 requires a systematic approach to data reconciliation:

  • Context-specific evaluation framework:

    • Stratify analyses by:

      • Cell/tissue type (considering native PDIA3 expression levels)

      • Disease context (cancer vs. inflammatory vs. normal physiology)

      • Experimental model (in vitro, animal models, human samples)

    • Consider microenvironmental differences that might alter PDIA3 function

    • Examine cell type-specific effects rather than relying solely on bulk tissue data

  • Mechanistic reconciliation approaches:

    • Dissect PDIA3's multiple functional domains and activities

    • Distinguish between enzymatic (oxidoreductase) and chaperone functions

    • Consider post-translational modifications that might regulate activity

    • Examine protein-protein interactions that vary between experimental contexts

  • Technical considerations:

    • Evaluate methodological differences between contradictory studies

    • Assess specificity of detection methods (antibodies, activity assays)

    • Consider temporal dynamics of observed effects

    • Verify protein localization, as PDIA3 may function differently in distinct cellular compartments

  • Integrative analysis strategies:

    • Perform meta-analyses across published datasets

    • Use systems biology approaches to model context-dependent functions

    • Apply machine learning to identify variables that predict functional differences

For example, in glioma research, PDIA3 was found to upregulate MHC-I, which contradicted findings in other tumor types. Researchers attributed this to tumor heterogeneity, suggesting this discrepancy represents a productive direction for future glioma research rather than an experimental error . This approach acknowledges that biological complexity often manifests as seemingly contradictory results across different experimental systems.

What are the most promising avenues for translating PDIA3 research into clinical applications?

Several high-potential translational pathways exist for PDIA3 research:

  • Diagnostic and prognostic applications:

    • Development of PDIA3 expression as a prognostic biomarker in gliomas

    • Detection of anti-PDIA3 antibodies as diagnostic markers in autoimmune conditions

    • Integration of PDIA3 status into molecular classification systems for cancer

    • Creation of companion diagnostics for therapies targeting PDIA3-related pathways

  • Therapeutic targeting strategies:

    • Development of small molecule inhibitors specific to PDIA3

    • Engineering of biologics that modulate PDIA3-dependent immune processes

    • Design of combination approaches with existing immunotherapies

    • Creation of epitope-specific immunomodulation for autoimmune conditions

  • Personalized medicine applications:

    • Stratification of patients based on PDIA3 expression patterns

    • Identification of synthetic lethal interactions with common cancer mutations:

      • PTEN loss in high PDIA3 expression tumors

      • IDH mutations in low PDIA3 expression contexts

    • Development of treatment algorithms incorporating PDIA3 status

  • Drug repurposing opportunities:

    • Screening of approved drugs for PDIA3-modulating activity

    • Evaluation of existing immunomodulators for effects on PDIA3-dependent pathways

    • Identification of natural products with PDIA3-targeting properties

These translational avenues acknowledge PDIA3's multifaceted roles in disease pathophysiology and leverage growing understanding of its mechanisms to develop clinically relevant applications in both cancer and inflammatory disease contexts .

What technological advances would most benefit future PDIA3 research?

Several technological advances would significantly accelerate PDIA3 research:

  • Structural biology innovations:

    • Cryo-EM structures of PDIA3 in complex with binding partners

    • Time-resolved structural studies to capture dynamic conformational changes

    • Computational modeling of PDIA3's interactions within the peptide loading complex

    • Development of conformation-specific antibodies for functional studies

  • Single-cell and spatial technologies:

    • Higher-throughput single-cell proteomics to measure PDIA3 protein levels

    • Spatial transcriptomics to map PDIA3 expression within tissue architecture

    • In situ protein interaction mapping to visualize PDIA3 complexes in native context

    • Live-cell imaging of PDIA3 trafficking between cellular compartments

  • Functional genomics tools:

    • Domain-specific CRISPR screens to map functional regions of PDIA3

    • Base editing approaches for precise modification of PDIA3 active sites

    • Inducible/reversible perturbation systems for temporal control

    • Single-cell CRISPR screens to capture cell type-specific functions

  • Translational research platforms:

    • Patient-derived organoids to study PDIA3 in personalized models

    • Humanized mouse models for immune interaction studies

    • High-throughput screening platforms for PDIA3-targeting compounds

    • Sensitive assays for detecting anti-PDIA3 antibodies in patient samples

These technological advances would overcome current limitations in understanding PDIA3's complex functions across different cellular contexts and disease states, enabling more precise targeting of PDIA3-dependent pathways for therapeutic benefit .

What are the most critical unanswered questions regarding PDIA3 biology and pathophysiology?

Several fundamental questions remain unanswered about PDIA3 biology:

  • Regulatory mechanisms:

    • How is PDIA3 expression and activity regulated in different cell types?

    • What post-translational modifications control PDIA3 function?

    • How do cells regulate PDIA3's distribution between different subcellular compartments?

    • What determines substrate specificity among PDI family members?

  • Immune system interactions:

    • What specific mechanisms underlie PDIA3's effects on T cell function?

    • How does PDIA3 contribute to epitope selection during antigen presentation?

    • What drives the development of pathogenic anti-PDIA3 antibodies?

    • How does PDIA3 influence the balance between immunity and tolerance?

  • Disease pathogenesis contributions:

    • Is PDIA3 dysregulation a cause or consequence of disease processes?

    • What explains the seemingly contradictory roles of PDIA3 across different cancer types?

    • How do metabolic insults trigger PDIA3 autoreactivity in liver conditions?

    • Can targeting PDIA3 overcome immunotherapy resistance mechanisms?

  • Translational research priorities:

    • Which patient populations would benefit most from PDIA3-targeted therapies?

    • What biomarkers best predict response to PDIA3 modulation?

    • How can PDIA3-specific interventions be delivered to appropriate cellular compartments?

    • What combination approaches might synergize with PDIA3-targeted strategies?

Addressing these questions will require interdisciplinary approaches combining structural biology, immunology, cancer biology, and translational research. The answers will illuminate not only fundamental aspects of cellular protein folding and quality control but also open new therapeutic avenues for conditions ranging from cancer to autoimmune and metabolic diseases .

Product Science Overview

Localization and Function

PDIA3 is primarily localized in the endoplasmic reticulum (ER), where it interacts with lectin chaperones such as calreticulin and calnexin . These interactions are essential for modulating the folding of glycoproteins by promoting the formation of disulfide bonds . The enzyme’s activity is vital for maintaining cellular homeostasis and ensuring the correct conformation of proteins, which is critical for their functionality .

Preparation Methods

The recombinant form of PDIA3 is produced using recombinant DNA technology. This involves cloning the PDIA3 gene into an expression vector, which is then introduced into a suitable host cell, such as Escherichia coli or yeast. The host cells are cultured under conditions that promote the expression of the PDIA3 protein. Once expressed, the protein is purified using various chromatographic techniques to obtain the active recombinant enzyme .

Chemical Reactions and Analysis

PDIA3 catalyzes several important chemical reactions involving disulfide bonds. These reactions include:

  1. Formation of Disulfide Bonds: PDIA3 facilitates the formation of disulfide bonds between cysteine residues in nascent polypeptides, which is essential for the structural integrity of proteins.
  2. Isomerization of Disulfide Bonds: The enzyme can rearrange incorrect disulfide bonds to ensure the proper folding of proteins.
  3. Reduction or Oxidation of Disulfide Bonds: PDIA3 can also reduce or oxidize disulfide bonds, depending on the cellular redox state .

The activity of PDIA3 is typically analyzed using biochemical assays that measure its ability to catalyze these reactions. These assays often involve substrates that mimic the natural substrates of PDIA3 and can be monitored using spectroscopic or chromatographic methods .

Biological Significance

PDIA3 is involved in various biological processes, including the unfolded protein response (UPR), which is activated in response to the accumulation of misfolded proteins in the ER. By ensuring the proper folding of glycoproteins, PDIA3 helps maintain protein homeostasis and prevent cellular stress .

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.