An ERP29 antibody pair typically includes:
Capture antibody: Binds ERP29 and immobilizes it on a solid phase.
Detection antibody: Recognizes a different epitope and generates a measurable signal.
This configuration enhances specificity and sensitivity in detecting ERP29, particularly in complex biological samples .
The table below lists validated ERP29 antibodies from diverse sources that could form functional pairs:
Epitope diversity: Antibodies targeting non-overlapping regions (e.g., N-terminal vs. C-terminal) minimize cross-interference.
Application compatibility: For ELISA, pair antibodies validated for capture (e.g., 24344-1-AP) and detection (e.g., ab137670) .
Species reactivity: Cross-species pairs (e.g., human/mouse) enable translational studies .
Role in metastasis: ERP29 promotes CRC cell proliferation and migration by inhibiting apoptosis via Bcl-2-mediated suppression of caspase-3 and PARP .
Regulatory loop: ERP29 expression negatively correlates with miR-135a-5p (R = −0.3, P = 0.021), suggesting a feedback mechanism in cancer progression .
Capture: Coat plates with Proteintech 24344-1-AP (1:500 dilution in PBS).
Block: Use 5% BSA for 1 hour.
Incubate: Add sample (e.g., cell lysate or serum).
Detect: Apply biotinylated ab137670 (1:1000 dilution) with streptavidin-HRP.
ERP29 (Endoplasmic Reticulum Protein 29) is a chaperone protein primarily localized in the endoplasmic reticulum that plays a critical role in protein folding and secretion. It is widely expressed in various tissues and is involved in early protein processing events within the ER .
ERP29 has gained significant research attention due to its:
Involvement in ER stress responses and protein quality control mechanisms
Tissue-specific roles in cancer progression
Participation in multiple signaling pathways including WNT, MAPK, and PI3K/AKT
Association with drug resistance mechanisms, particularly to cisplatin (CDDP) in cancer
Studies have demonstrated that ERP29 expression varies across different disease states, making it a valuable biomarker and potential therapeutic target in conditions ranging from cancer to age-related degenerative disorders .
When optimizing Western blotting for ERP29 detection, consider these methodological approaches:
Sample preparation:
Gel conditions:
Transfer and blocking:
Transfer to PVDF or nitrocellulose membranes (PVDF may provide better signal-to-noise ratio)
Block with 5% non-fat milk or BSA in TBST
Antibody incubation:
Detection and troubleshooting:
Use enhanced chemiluminescence for visualization
If background is high, increase washing steps or decrease antibody concentration
For weak signals, consider longer exposure times or signal enhancement systems
Research has shown that ERP29 expression may vary significantly between different cell types and physiological conditions, so optimization may be required for specific experimental contexts .
When validating ERP29 antibody specificity, both positive and negative controls should be incorporated:
Positive Controls:
Cell lines with known ERP29 expression: HEK-293, L02, HeLa cells
Tissue samples: Mouse liver, kidney, lung; rat testis tissue
Recombinant ERP29 protein as a reference standard
Negative Controls:
Primary antibody omission control
Use of non-specific IgG of the same host species
ERp29-knockout or silenced cells (via siRNA or CRISPR systems)
Peptide competition assay using the immunizing peptide that was used to generate the antibody
Validation Methods:
Cross-reactivity assessment: Test reactivity with closely related proteins
Molecular weight verification: Confirm the observed band is at the expected 29 kDa
Multiple antibody approach: Use antibodies targeting different epitopes of ERP29
Orthogonal validation: Corroborate protein expression with mRNA levels
Research has demonstrated that rabbit polyclonal antibodies against ERP29 show cross-reactivity with human, mouse, rat, and other species including dog, cow, sheep, and guinea pig samples , providing versatility for comparative studies across species.
ERP29 functions as an important regulator in the endoplasmic reticulum stress (ERS) response pathway:
Protective role: ERP29 attenuates ERS-induced apoptosis in various cellular contexts
Temporal dynamics: ERP29 shows distinct expression patterns in response to stress
Methods to study ERP29 in ER stress include:
Basic approaches:
Western blotting to quantify ERP29 and ERS markers (GRP78, CHOP, p-eIF2α)
RT-qPCR to measure transcript levels
Immunofluorescence to examine subcellular localization changes
Advanced approaches:
Proximity ligation assay to detect in situ protein-protein interactions
CRISPR-Cas9 gene editing to create ERP29-knockout cell lines
Mass spectrometry to identify ERP29 interacting partners during ERS
Polysome profiling to examine effects on translation
Live-cell imaging with fluorescently tagged ERP29
In a study using retinal pigment epithelial (RPE) cells exposed to cigarette smoke extract (CSE), researchers found that overexpression of ERP29 protected cells from CSE-induced apoptosis, while knockdown of ERP29 exacerbated cell death, demonstrating its critical role in stress adaptation .
ERP29 functionality depends on its dimerization state, making this an important aspect to investigate:
Techniques to detect and characterize ERP29 dimerization:
Chemical cross-linking:
Size exclusion chromatography:
Separate proteins based on size
Compare elution profiles with known standards
Western blot fractions to detect ERP29
Native PAGE:
Perform electrophoresis under non-denaturing conditions
Western blot to detect native complexes
Fluorescence techniques:
FRET (Förster Resonance Energy Transfer) with fluorescently tagged ERP29
Fluorescence Correlation Spectroscopy to determine diffusion rates
Studying functional significance:
Mutagenesis approaches:
Domain-based studies:
Research has shown that the N-terminal domain of ERP29 is responsible for dimerization, while the C-terminal domain is likely involved in substrate binding. The D42A mutation specifically disrupts dimerization and significantly impairs ERp29 function, highlighting the critical nature of this structural feature .
Recent studies have revealed a regulatory feedback loop between ERP29 and microRNAs, particularly miR-135a-5p and miR-4421, which can be studied using these approaches:
Experimental strategies:
Expression correlation studies:
Direct targeting validation:
Luciferase reporter assays with wild-type and mutated ERP29 3'UTR
miRNA mimic and inhibitor transfections followed by ERP29 quantification
RISC immunoprecipitation to confirm physical interaction
Feedback mechanism investigation:
Functional impact assessment:
miRNA modulation followed by phenotypic assays
Combined manipulation of miRNA and ERP29 to determine epistatic relationships
In vivo models testing therapeutic potential of miRNA inhibitors
Research findings:
In colorectal cancer, miR-135a-5p directly targets ERP29 mRNA
Conversely, ERP29 suppresses IL-1β-elicited methylation of the miR-135a-5p promoter region
This creates a homeostatic feedback loop that maintains appropriate levels of both molecules
In cancer, this balance is disrupted, contributing to disease progression
Lower ERP29 and higher miR-4421 expressions correlate with poor survival in patients
These findings suggest potential therapeutic applications targeting the ERP29-miRNA regulatory axis in diseases like colorectal and pharyngeal squamous cell carcinomas.
Optimizing immunohistochemistry (IHC) protocols for ERP29 detection requires attention to several critical factors:
Tissue preparation and fixation:
Formalin-fixed paraffin-embedded (FFPE) sections typically yield good results
Optimal section thickness: 4-6 μm
Freshly cut sections are preferable to stored slides
Antigen retrieval:
Heat-induced epitope retrieval (HIER) is recommended
Two effective buffer options:
Microwave or pressure cooker for 10-20 minutes
Antibody selection and optimization:
Rabbit polyclonal antibodies show good reactivity across multiple species
Incubation conditions: 4°C overnight or 1-2 hours at room temperature
Both DAB (chromogenic) and fluorescence detection systems work well
Controls and validation:
Positive tissue controls: Human liver cancer, breast cancer, pancreas; mouse liver, kidney
Negative controls: Primary antibody omission, isotype control
Counterstain: Hematoxylin for nuclear visualization
Consider dual staining with ER markers (calnexin, PDI) for co-localization
Analysis considerations:
ERP29 shows primarily cytoplasmic/ER localization pattern
Expression levels vary significantly between cell types within the same tissue
ERp29 expression profile largely parallels that of protein disulfide isomerase (PDI) but with strikingly different ERp29/PDI ratios in various cell types
Research has identified 35 functionally distinct cell types in rat tissues that express ERP29, establishing it as a general ER marker but with tissue-specific expression patterns that may correlate with secretory activity .
ERP29 has emerged as an important factor in cisplatin (CDDP) resistance mechanisms, particularly in pharyngeal squamous cell carcinoma (PSCC). Researchers can investigate this relationship using these methodological approaches:
Cell model establishment:
Develop cisplatin-sensitive, cisplatin-treated, and cisplatin-resistant cell lines:
Verify resistance phenotype through dose-response curves and IC50 determination
ERP29 expression analysis:
Quantify baseline ERP29 expression across cell models using:
Western blotting (protein level)
qRT-PCR (mRNA level)
Flow cytometry (per-cell analysis)
Key finding: FaDu-R cells show decreased ERP29 expression compared to sensitive and treated cells
Functional studies:
Modify ERP29 expression:
Overexpression using expression vectors
Silencing using siRNA or shRNA
Assess cellular behaviors:
Cell proliferation and cell cycle analysis
Necrosis and apoptosis detection
Migration assays
3D spheroid formation with E-cadherin/vimentin assessment
Evaluate cisplatin sensitivity changes through:
MTT/MTS viability assays at varying drug concentrations
Colony formation assays
Apoptosis markers (Annexin V, PARP cleavage)
Mechanistic investigations:
Gene expression analysis:
Protein interaction studies:
Co-immunoprecipitation with ERP29 antibodies
Immunofluorescence co-localization
miRNA-based approaches:
Assess miR-4421 effects:
Survival correlation analysis in patient samples
Recent research found that ERP29 silencing decreases necrotic cell death and increases migration in cisplatin-sensitive, treated, and resistant cells. During cisplatin treatment, ERP29 silencing enhances cell proliferation and alters expression of genes involved in key signaling pathways, suggesting its important role in drug resistance mechanisms .
Co-immunoprecipitation (Co-IP) using ERP29 antibodies can reveal novel protein interactions and complexes, but requires careful optimization:
Experimental design considerations:
Lysis buffer optimization:
Use mild non-denaturing buffers to preserve protein-protein interactions
Include appropriate protease/phosphatase inhibitors
Consider specialized buffers for ER proteins (e.g., containing low concentrations of detergents like digitonin or NP-40)
Antibody selection:
Controls to include:
Input control (5-10% of starting material)
IgG control (same species/amount as ERP29 antibody)
Reciprocal IP (IP with antibodies against suspected interacting proteins)
Negative control samples (ERP29-knockdown cells)
Detection strategies:
Western blot with antibodies against potential interacting partners
Mass spectrometry for unbiased identification of protein complexes
Functional validation of identified interactions
Known ERP29 interactions to investigate:
PDI family members (protein folding)
Components of WNT, MAPK, and PI3K/AKT pathways
miRNA processing machinery
Advanced technical considerations:
Crosslinking approaches:
Subcellular fractionation:
Enrich for ER fractions before IP to increase specificity
Enables detection of compartment-specific interactions
Proximity-dependent labeling:
BioID or APEX2 fusions with ERP29 for in vivo proximity labeling
Identifies spatial protein networks around ERP29
Research has demonstrated that ERP29 forms homodimers that are essential for its function, and that the N-terminal domain is responsible for this dimerization. Co-IP experiments have successfully identified interactions between ERP29 and other ER proteins involved in protein folding and secretion .
ERP29's involvement in epithelial-mesenchymal transition (EMT) can be studied through these methodological approaches:
Cellular models and markers:
3D spheroid cultures:
EMT marker profiling:
Western blot/IF analysis of canonical markers:
Epithelial: E-cadherin, ZO-1, PAR3
Mesenchymal: Vimentin, N-cadherin, Fibronectin
qRT-PCR for transcriptional regulators (SNAIL, SLUG, ZEB1/2, TWIST)
Functional assays:
Migration and invasion:
Cell adhesion:
Matrix adhesion assays
Cell-cell aggregation assays
E-cadherin function assays (calcium switch)
Morphological analysis:
Phase-contrast microscopy for morphological changes
Cytoskeletal reorganization (F-actin staining)
Molecular mechanisms:
Signal pathway analysis:
WNT pathway activation (β-catenin localization)
MAPK pathway (ERK1/2 phosphorylation)
PI3K/AKT signaling (AKT phosphorylation)
TGF-β signaling (SMAD activation)
Gene expression regulation:
ChIP assays for promoter binding
3'UTR reporter assays for miRNA regulation
RNA-seq for global expression changes
Translational approach:
Clinical correlations:
IHC analysis of patient samples for ERP29 and EMT markers
Correlation with prognostic indicators and survival outcomes
Metastasis association analysis