ERO1B Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ERO1B and Its Antibodies

ERO1B is a flavoprotein critical for disulfide bond formation during protein folding, working in concert with Protein Disulfide Isomerase (PDI) . It is highly expressed in pancreatic β-cells and plays roles in insulin biosynthesis, ER stress responses, and cancer progression . ERO1B antibodies are essential for:

  • Localization studies: Mapping ERO1B expression in tissues (e.g., pancreatic islets, cancer cells) .

  • Functional assays: Assessing impacts on insulin content, ER stress adaptation, and tumor metastasis .

  • Diagnostic/therapeutic research: Identifying ERO1B as a biomarker in cancers and metabolic disorders .

Validated ERO1B Antibodies

The table below lists top validated ERO1B antibodies, their suppliers, and applications based on experimental data :

SupplierCatalog NumberHostApplicationsReferences
Proteintech Group11261-2-APRabbitWB, ELISA, ICC, IHC6
LSBioLS-C399028RabbitWB, ELISA, IHC-
Invitrogen AntibodiesPA5-25142RabbitWB, IHC, Flow Cytometry1
NovoPro Bioscience Inc.131455RabbitWB, ELISA, IHC, FC-
Abcamab197290RabbitWB, IHC-P-

Role in Pancreatic β-Cell Function

  • Insulin biosynthesis: ERO1B maintains insulin content by facilitating proinsulin folding. Silencing ERO1B reduces insulin levels by ~30% in MIN6 cells .

  • ER stress adaptation: ERO1B deficiency increases JNK phosphorylation and susceptibility to ER stress-induced apoptosis .

  • Regulation by PDX1: PDX1 directly binds the Ero1lβ promoter, and Pdx1 haploinsufficiency reduces ERO1B expression by 57% in mouse islets .

Implications in Cancer

  • Metastasis promotion: ERO1B knockdown reduces migration in MDA-MB-231 breast cancer cells by 40% and synergizes with anti-angiogenic therapies to decrease lung metastases .

  • Hypoxia response: ERO1B is upregulated in hypoxic tumor microenvironments, correlating with aggressive basal breast cancers .

  • Angiogenesis modulation: ERO1B-deficient tumors show 30% lower microvessel density (CD31 staining) and impaired VEGF-A secretion .

Tissue Distribution

ERO1B exhibits enriched expression in pancreatic islets, liver, and endocrine tissues, with moderate levels in the brain and gastrointestinal tract .

Transcriptional Control

  • Glucose induction: High glucose upregulates ERO1B in β-cells to manage oxidative folding demands .

  • Chemical modulation: Sodium arsenite and acrylamide alter ERO1B expression, linking it to environmental stress responses .

Therapeutic and Diagnostic Potential

  • Cancer biomarkers: Elevated ERO1B correlates with shorter metastasis-free survival in breast cancer patients (HR = 1.8) .

  • Drug targeting: Inhibiting ERO1B enhances the efficacy of anti-VEGF therapies, reducing tumor growth by 50% in preclinical models .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary based on your location and the method of purchase. For specific delivery information, please consult your local distributor.
Synonyms
Endoplasmic oxidoreductin-1-like protein B antibody; Endoplasmic reticulum oxidoreductin-1-like protein B antibody; ERO1; S. cerevisiae; homolog of; beta antibody; ERO1-L-beta antibody; ERO1-like beta (S. cerevisiae) antibody; ERO1-like protein beta antibody; ERO1B_HUMAN antibody; Ero1lb antibody; Oxidoreductin-1-L-beta antibody
Target Names
Uniprot No.

Target Background

Function
ERO1B is an oxidoreductase enzyme involved in disulfide bond formation within the endoplasmic reticulum. It efficiently reoxidizes P4HB/PDI, an enzyme crucial for protein disulfide formation, allowing P4HB to continue facilitating disulfide bond formation in subsequent cycles. While ERO1B can also reoxidize other members of the protein disulfide isomerase family, it does so at lower rates compared to P4HB. This includes PDIA2 (50% of P4HB reoxidation rate), as well as PDIA3, PDIA4, PDIA6, and NXNDC12 (<10%). Following P4HB reoxidation, ERO1B transfers its electrons to molecular oxygen via FAD, leading to the production of reactive oxygen species (ROS) within the cell. ERO1B may participate in oxidative proinsulin folding in pancreatic cells, suggesting a potential role in glucose homeostasis.
Gene References Into Functions
  1. The peptide Ac-VDTTD-AFC was effectively cleaved by purified caspase-2 and auto-activating caspase-2 in mammalian cells. This peptide demonstrated improved selectivity for caspase-2 compared to caspase-3 in relation to currently available reagents. PMID: 27919037
  2. GJB2 and ERO1LB are implicated in pancreatic cancer progression and can be utilized as predictive markers for patient survival. PMID: 28177904
  3. ERO1B is upregulated in pancreatic neuroendocrine tumors. PMID: 26765469
  4. In recent research, it was demonstrated that recombinant human Ero1beta exhibits twice the activity of Ero1alpha in enzymatic assays. PMID: 21091435
  5. Glutathione plays a role in limiting Ero1-dependent oxidation within the endoplasmic reticulum. PMID: 15161913
  6. Research has focused on analyzing Ero1beta tissue distribution and dimerization. PMID: 16012172
  7. Ero1alpha and Ero1beta are retained within the endoplasmic reticulum through interactions with PDI and ERp44. PMID: 16677073
  8. The FAD binding domain of Ero proteins is critical for their conformational stability, enabling them to withstand stressful conditions that cause client proteins to misfold. PMID: 16822866
  9. The lack of correlation between changes in SAT adiponectin gene and protein expression and its circulating levels suggests that adipose tissue synthesis and release of adiponectin are highly regulated processes. PMID: 18996753

Show More

Hide All

Database Links

HGNC: 14355

OMIM: 615437

KEGG: hsa:56605

STRING: 9606.ENSP00000346635

UniGene: Hs.558519

Protein Families
EROs family
Subcellular Location
Endoplasmic reticulum membrane; Peripheral membrane protein; Lumenal side. Note=The association with ERP44 may be essential for its retention in the endoplasmic reticulum.
Tissue Specificity
Highly expressed in the digestive tract, including the duodenum and lower digestive tract. In the stomach, highly expressed in enzyme-producing chief cells (at protein level). In the pancreas, expressed in islets of Langerhans and, at lower levels, in enz

Q&A

What is ERO1B and why is it important in cellular function?

ERO1B (ERO1-like protein beta) is an endoplasmic reticulum oxidoreductin responsible for maintaining the oxidative environment necessary for proper disulfide bond formation in proteins within the ER. Unlike its ubiquitously expressed counterpart ERO1α, ERO1B shows enriched expression in pancreatic tissue . It functions by transferring electrons from protein disulfide isomerase (PDI) to molecular oxygen, enabling PDI to facilitate disulfide bond formation in cargo proteins . This process is particularly critical in cells with high secretory capacity, such as pancreatic β-cells, where ERO1B plays a vital role in insulin biosynthesis and folding. Research demonstrates that ERO1B deficiency leads to delayed proinsulin folding to its native disulfide state, reducing insulin content and impairing insulin secretion . Furthermore, ERO1B is directly regulated by PDX1 (pancreatic and duodenal homeobox 1) and is induced by high glucose concentrations, indicating its importance in the β-cell response to increased insulin demand .

What are the key considerations when selecting an ERO1B antibody for research?

When selecting an ERO1B antibody for research applications, several critical factors should be considered to ensure reliable and reproducible results. First, evaluate the antibody's validated applications (Western blot, immunohistochemistry, immunofluorescence, etc.) to confirm it will work in your experimental system . For instance, the anti-ERO1B antibody from Boster Bio has been validated for Western blot, immunohistochemistry, immunocytochemistry, immunofluorescence, and ELISA applications .

Second, consider the species reactivity of the antibody. The antibody should recognize ERO1B in your experimental model organism. Some antibodies may only react with human ERO1B, while others may have cross-reactivity with mouse or other species . Third, examine the immunogen used to generate the antibody. Antibodies raised against full-length proteins or large domains may provide different results than those raised against small peptides. For example, the Boster antibody utilizes E.coli-derived human ERO1B recombinant protein (Position: E170-R467) as its immunogen .

Finally, consider the clonality (monoclonal vs. polyclonal) based on your research needs. Polyclonal antibodies like the one described in the search results recognize multiple epitopes and may provide higher sensitivity but potentially lower specificity compared to monoclonals .

How can I distinguish between ERO1B and the closely related ERO1α in my experiments?

Distinguishing between ERO1B and ERO1α in experiments requires careful consideration of their differences in expression patterns, molecular characteristics, and the selection of specific antibodies. ERO1α is ubiquitously expressed across tissues, whereas ERO1B expression is enriched in pancreatic tissue, particularly in β-cells . This differential expression pattern can be leveraged when designing experiments.

For immunoblotting or immunostaining experiments, utilize antibodies that specifically target unique epitopes of ERO1B not shared with ERO1α. Verify antibody specificity by examining the immunogen sequence used for antibody production and confirming it doesn't share significant homology with ERO1α. For instance, antibodies generated against the C-terminal region may provide better specificity as this region often differs between protein isoforms .

At the mRNA level, design primers that target unique regions of ERO1B transcripts for RT-PCR or qPCR analysis. The search results mention primers for mouse Ero1lβ and Ero1lα that were previously described in the literature, suggesting researchers have successfully developed tools to distinguish between these paralogs .

When analyzing protein function, consider the potential for compensatory upregulation of ERO1α in response to ERO1B knockdown. Measuring both proteins simultaneously may provide insights into their functional relationship.

What are the optimal conditions for using ERO1B antibodies in Western blotting?

For optimal Western blot results with ERO1B antibodies, several technical considerations must be addressed. First, sample preparation should preserve the native structure of ERO1B, which has a calculated molecular weight of approximately 53.5 kDa . Cells or tissues should be lysed in a buffer containing appropriate protease inhibitors to prevent degradation, and samples should be processed promptly.

For SDS-PAGE separation, a 10% or 12% polyacrylamide gel is typically suitable for resolving proteins in the 50-60 kDa range. Based on the search results, the recommended dilution range for Western blot applications is 1:500-2000 . Researchers should optimize this range for their specific experimental conditions and antibody lot.

For the primary antibody incubation, overnight incubation at 4°C with gentle agitation typically yields the best results. As described in the search results, this was the method used with a 1:1000 dilution in PBS plus 0.05% Tween . For detection, appropriate HRP-conjugated secondary antibodies (goat anti-rabbit if using a rabbit primary antibody) should be used at the manufacturer's recommended dilution, with incubation for approximately 1 hour at room temperature .

Always include appropriate controls in your Western blot experiments, including positive controls (tissues or cells known to express ERO1B, such as pancreatic tissue), negative controls (tissues with minimal ERO1B expression), and loading controls (cyclophilin A or β-actin have been successfully used as shown in the search results) .

How should I optimize ERO1B antibody staining for immunohistochemistry and immunofluorescence?

For tissue sections, proper fixation is crucial. Paraformaldehyde (4%) is commonly used, but the fixation time should be optimized to preserve antigenicity while maintaining tissue morphology. Antigen retrieval methods may be necessary, especially for formalin-fixed, paraffin-embedded tissues. Heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) is often effective for exposing masked epitopes.

For cell lines, such as the MIN6 cells mentioned in the search results, fixation with 4% paraformaldehyde for 15-20 minutes at room temperature is typically sufficient . Permeabilization with 0.1-0.3% Triton X-100 may be necessary to allow antibody access to intracellular ERO1B, which is predominantly localized to the endoplasmic reticulum.

Blocking steps are critical to reduce background signal. A solution of 1-5% normal serum (from the same species as the secondary antibody) or BSA in PBS is commonly used for 1-2 hours at room temperature. For fluorescence detection, select secondary antibodies with appropriate fluorophores, and include DAPI counterstaining to visualize nuclei. Always include negative controls (primary antibody omitted) and positive controls (tissues known to express ERO1B, such as pancreatic islets) to validate staining specificity.

What methods can be used to validate ERO1B antibody specificity?

Validating antibody specificity is critical for ensuring reliable experimental results. Several complementary approaches should be employed to confirm ERO1B antibody specificity. First, RNA interference experiments can provide strong evidence for antibody specificity. As described in the search results, small interfering RNA (siRNA)-mediated silencing of Ero1lβ was used to decrease ERO1B expression in MIN6 cells . Following siRNA treatment, the reduction in ERO1B protein levels detected by the antibody confirms its specificity for the target.

Second, overexpression studies using vectors containing the ERO1B coding sequence can demonstrate antibody specificity by showing increased signal intensity in transfected versus non-transfected cells. Third, peptide competition assays, where the antibody is pre-incubated with the immunizing peptide or recombinant protein before application to samples, can validate specificity by demonstrating signal reduction.

Western blot analysis should show a single band at the expected molecular weight (approximately 53.5 kDa for ERO1B) , while additional bands may indicate cross-reactivity with other proteins. Comparative analysis across multiple tissues with known differential expression of ERO1B (high in pancreatic tissue, lower in other tissues) can further validate specificity.

Finally, using multiple antibodies targeting different epitopes of ERO1B and observing consistent results provides additional confidence in antibody specificity. The search results indicate that blocking peptides can be purchased for the anti-ERO1B antibody, which would be valuable for competition assays to confirm specificity .

How does ERO1B expression change under ER stress conditions, and what are the implications for experimental design?

ERO1B expression demonstrates dynamic regulation under ER stress conditions, which has significant implications for experimental design. Research indicates that ERO1B levels are induced by reducing agents such as dithiothreitol (DTT), suggesting a role in adaptation to increased oxidative protein folding load in the β-cell ER . This induction likely represents a cellular response to restore the oxidative environment necessary for proper disulfide bond formation when disrupted by reducing conditions.

High glucose concentrations also induce ERO1B expression, particularly in pancreatic β-cells, indicating its role in managing increased insulin production demands . When designing experiments to study ERO1B under stress conditions, researchers should consider time-course experiments to capture the dynamic changes in expression. For instance, in the research described, MIN6 cells were treated with 1 mM DTT for various timepoints (0, 1, 2, 4, and 6 hours) before harvesting for RNA and protein analysis .

Experimental designs should include appropriate controls and multiple stress inducers to comprehensively characterize ERO1B responses. Combining transcriptional analysis (RT-PCR) with protein-level analysis (Western blot) provides a more complete picture of ERO1B regulation. Additionally, researchers should consider measuring markers of ER stress response pathways (such as XBP1 splicing, CHOP induction, and JNK phosphorylation) alongside ERO1B to contextualize its role within the broader stress response .

For knockdown or overexpression studies, timing is critical since cellular responses to ER stress are highly dynamic. The search results indicate that for siRNA experiments in MIN6 cells, harvesting occurred 72 or 96 hours after nucleofection to measure silencing efficiency .

What is the relationship between ERO1B and cancer progression, and how can antibodies help investigate this connection?

While the search results focus primarily on ERO1α's role in cancer progression rather than ERO1B, the information provides valuable insights for investigating potential ERO1B involvement in cancer. ERO1α overexpression correlates with worse prognosis in multiple cancer indications, including multiple myeloma, breast cancer, hepatocellular carcinoma, lung cancer, esophageal cancer, and diffuse B-cell lymphoma according to The Cancer Genome Atlas (TCGA) and The Protein Atlas . ERO1α promotes immune escape through up-regulation of PD-L1 in breast cancer, mediating both oxidative folding of PD-L1 and augmenting PD-L1 mRNA expression through HIF-1α .

Given the functional similarity between ERO1α and ERO1B in oxidative protein folding, researchers might hypothesize parallel or complementary roles for ERO1B in cancer biology, particularly in pancreatic cancers where ERO1B is highly expressed. ERO1B antibodies can help investigate this potential connection through several approaches:

  • Tissue microarray analysis: Using ERO1B antibodies for immunohistochemical staining of cancer tissue arrays to correlate expression levels with clinical outcomes, tumor stage, and patient survival.

  • Cell line characterization: Profiling ERO1B expression across cancer cell lines, particularly those derived from pancreatic origin, using Western blot and immunofluorescence with validated ERO1B antibodies.

  • Functional studies: Employing ERO1B antibodies to monitor protein levels following experimental manipulation (knockdown or overexpression) to assess effects on cancer hallmarks like proliferation, invasion, and resistance to apoptosis.

  • Co-immunoprecipitation: Using ERO1B antibodies to identify protein-protein interactions that might reveal novel signaling pathways in cancer cells.

  • Monitoring treatment responses: Analyzing changes in ERO1B expression following various cancer treatments to identify potential biomarker applications.

How does ERO1B function influence insulin production in pancreatic β-cells, and what experimental approaches can be used to study this?

ERO1B plays a crucial role in insulin production in pancreatic β-cells through its function in maintaining the oxidative environment necessary for proper disulfide bond formation. Insulin contains three disulfide bonds that are essential for its proper folding, stability, and function. The search results demonstrate that ERO1B deficiency in MIN6 cells (a mouse insulinoma cell line) delays proinsulin folding to its native disulfide state and reduces insulin content, leading to impaired insulin secretion .

Research has shown that ERO1B is a direct target of PDX1 (pancreatic and duodenal homeobox 1), a key transcription factor in pancreatic β-cell development and function. PDX1 deficiency reduced ERO1B transcript levels in mouse islets and MIN6 cells, and PDX1 was found to occupy the ERO1B promoter in β-cells . Additionally, ERO1B expression is induced by high glucose concentrations, suggesting its role in adaptation to increased insulin production demands .

To study the relationship between ERO1B and insulin production, researchers can employ several experimental approaches:

  • siRNA-mediated knockdown: As described in the search results, silencing ERO1B expression using siRNA provides insights into its necessity for insulin content and secretion .

  • Proinsulin folding analysis: Non-reducing SDS-PAGE and immunoblotting with anti-insulin antibodies can be used to monitor the formation of native disulfide bonds in proinsulin following ERO1B manipulation .

  • Glucose-stimulated insulin secretion assays: Following ERO1B knockdown or overexpression, measuring insulin secretion in response to glucose challenges can reveal functional consequences.

  • ER stress analysis: Monitoring markers of ER stress (XBP1 splicing, CHOP expression, JNK phosphorylation) alongside ERO1B manipulation can elucidate the connection between ERO1B, insulin production, and ER homeostasis .

  • In vivo models: Tissue-specific knockout or overexpression of ERO1B in pancreatic β-cells of mice can provide insights into its physiological role in insulin production and glucose homeostasis.

What are the common technical challenges when working with ERO1B antibodies, and how can they be addressed?

Researchers working with ERO1B antibodies may encounter several technical challenges that require troubleshooting. One common issue is weak or absent signal in Western blot or immunostaining applications. This may result from insufficient protein expression, antibody degradation, or suboptimal experimental conditions. To address this, researchers should:

  • Verify ERO1B expression in their experimental system using RT-PCR before proceeding to protein-level analysis.

  • Use positive controls known to express ERO1B (such as pancreatic tissue or MIN6 cells) .

  • Optimize antibody concentration through titration experiments.

  • Extend primary antibody incubation time (overnight at 4°C as described in the search results) .

  • Consider alternative detection methods or more sensitive substrates.

Another challenge is high background or non-specific staining. This can be mitigated by:

  • Increasing blocking time or concentration (using 5% milk or BSA) .

  • Diluting antibody in fresh blocking solution.

  • Increasing wash steps in duration and number.

  • Using more stringent washing conditions (higher salt concentration or mild detergents).

  • Pre-absorbing the antibody with non-specific proteins from the species being studied.

For immunoprecipitation applications, low efficiency may occur. Researchers can:

  • Cross-link the antibody to beads to prevent co-elution with the target protein.

  • Optimize lysis conditions to ensure ERO1B remains soluble.

  • Extend incubation time for antigen-antibody binding.

Multiple bands in Western blot may indicate degradation, post-translational modifications, or non-specific binding. This can be addressed by:

  • Adding additional protease inhibitors to lysis buffer.

  • Validating bands using siRNA knockdown as described in the search results .

  • Running non-reducing and reducing conditions in parallel to understand disulfide-dependent banding patterns.

How can ERO1B antibodies be used in combination with other markers to study ER stress responses?

ERO1B antibodies can be effectively combined with other ER stress markers to provide comprehensive insights into cellular stress responses. Based on the search results, several key combinations and approaches emerge.

For immunofluorescence co-staining experiments, ERO1B antibodies can be paired with antibodies against ER resident proteins such as PDI (protein disulfide isomerase) to examine co-localization and potential functional interactions during ER stress. Additionally, co-staining with UPR (unfolded protein response) markers like BiP/GRP78 can reveal the relationship between ERO1B expression and ER stress intensity.

In Western blot analyses, researchers can probe the same samples for ERO1B alongside established ER stress markers. The search results indicate that phospho-JNK, total JNK, and CHOP antibodies have been successfully used in studies examining ERO1B function . This multi-protein analysis approach can reveal temporal relationships between ERO1B regulation and activation of specific ER stress pathways.

Protein MarkerFunctionDilution UsedReference
ERO1BER oxidoreductin1:1000
Phospho-JNKStress kinase activation1:1000
Total JNKStress kinase expression1:1000
CHOPPro-apoptotic transcription factor1:500
Cyclophilin ALoading control1:50,000
β-actinLoading control1:5000

For gene expression studies, quantitative RT-PCR for ERO1B can be performed alongside analysis of UPR-regulated genes. The search results mention primers for mouse Ero1lβ, sXbp1 (spliced XBP1), tXbp1 (total XBP1), and Chop , which together provide a comprehensive view of UPR activation across different branches (IRE1, PERK, and ATF6 pathways).

Flow cytometry can also be employed to simultaneously measure ERO1B levels and apoptotic markers in response to ER stress inducers, providing single-cell resolution of the relationship between ERO1B expression and cell fate decisions.

What considerations are important when designing experiments to study ERO1B interactions with other proteins?

Designing experiments to study ERO1B interactions with other proteins requires careful consideration of several factors due to the unique properties of this ER-resident oxidoreductase. Based on the search results and understanding of ERO1B biology, researchers should address the following considerations:

  • Maintaining native conditions: ERO1B functions through thiol-disulfide exchange reactions, which are sensitive to redox conditions. When preparing samples for interaction studies, avoid reducing agents in buffers that would disrupt native disulfide bonds. The search results mention non-reducing conditions for insulin immunoblots , suggesting similar approaches may be valuable for interaction studies.

  • Subcellular localization: ERO1B is primarily localized to the ER, though the search results mention it has been found in the Golgi apparatus and exosomes . Interaction studies should account for this compartmentalization by using appropriate fractionation methods to isolate ER-enriched fractions or by employing proximity-based techniques like proximity ligation assays (PLA).

  • Potential interaction partners: Based on its function, ERO1B likely interacts with PDI family members and possibly other components of the oxidative folding machinery. The search results indicate that ERO1 transfers electrons from PDI to molecular oxygen , suggesting PDI as a key interaction partner. Co-immunoprecipitation experiments should include PDI as a positive control.

  • Transient interactions: The enzymatic nature of ERO1B suggests its interactions may be transient. Consider using crosslinking agents before immunoprecipitation to capture these interactions. Mild crosslinkers like DSP (dithiobis[succinimidyl propionate]) that can be reversed may be particularly useful.

  • Expression level considerations: Overexpression systems may alter the stoichiometry of protein complexes. The search results indicate that both endogenous expression in appropriate cell types (like MIN6 cells) and regulated expression systems have been used to study ERO1B . Ideally, interaction studies should be performed at endogenous expression levels.

  • Validation strategies: Multiple orthogonal approaches should be used to validate interactions, including reciprocal co-immunoprecipitation, proximity ligation assays, FRET/BRET, and functional assays that assess the consequence of disrupting the interaction.

How might ERO1B antibodies be utilized in developing therapeutic approaches for diseases related to ER stress?

ERO1B antibodies could play important roles in developing therapeutic approaches for diseases related to ER stress, particularly those affecting tissues with high secretory demands like the pancreas. Based on the search results, ERO1B is critical for maintaining insulin content and regulating β-cell survival during ER stress , suggesting several potential therapeutic applications.

For diagnostic applications, ERO1B antibodies could be used to develop immunohistochemical assays to assess ERO1B expression levels in patient samples, potentially serving as biomarkers for ER stress severity in diseases like diabetes. The validated applications of ERO1B antibodies in immunohistochemistry support this potential use.

In drug discovery pipelines, ERO1B antibodies could be instrumental in high-throughput screening assays to identify compounds that modulate ERO1B activity or expression. Compounds that enhance ERO1B function might protect against ER stress-induced cell death in conditions like diabetes, while inhibitors might be relevant in contexts where excessive ERO1B activity contributes to pathology.

For targeted therapy development, the search results indicate that ERO1α (related to ERO1B) could be a tractable target for cancer treatment . Similar approaches might be considered for ERO1B in specific contexts. Antibodies would be essential tools for validating target engagement and efficacy in preclinical models.

In cell-based therapies, such as islet transplantation for diabetes, ERO1B antibodies could help select cell populations with optimal ERO1B expression or monitor stress responses post-transplantation. Additionally, gene therapy approaches aiming to modulate ERO1B expression would require antibodies to verify successful intervention.

Interestingly, the connection between ERO1 proteins and immune regulation suggested by the search results opens possibilities for immunotherapeutic approaches, with ERO1B antibodies serving crucial roles in understanding and manipulating these pathways.

What emerging technologies might enhance the utility of ERO1B antibodies in research?

Emerging technologies have the potential to significantly enhance the utility of ERO1B antibodies in research, enabling more precise spatial, temporal, and functional analysis. Super-resolution microscopy techniques such as STORM (Stochastic Optical Reconstruction Microscopy) and STED (Stimulated Emission Depletion) can overcome the diffraction limit, allowing researchers to visualize ERO1B localization within the ER with unprecedented detail. This could reveal previously undetectable microdomains or co-localization patterns with interaction partners.

Mass cytometry (CyTOF) combined with metal-conjugated ERO1B antibodies would enable simultaneous measurement of ERO1B expression alongside dozens of other cellular markers at single-cell resolution. This approach could be particularly valuable for analyzing heterogeneous responses to ER stress in mixed cell populations or tissues.

CRISPR-Cas9 genome editing can be paired with ERO1B antibodies to create and validate knockin cell lines expressing tagged versions of ERO1B (such as FLAG or HA tags). These systems would enable cleaner immunoprecipitation experiments and live-cell imaging when combined with fluorescent protein fusions.

Proximity-dependent biotinylation approaches like BioID or TurboID, paired with ERO1B antibodies for validation, could revolutionize the identification of ERO1B protein-protein interactions within the native cellular environment. By fusing ERO1B to a biotin ligase, researchers could identify proteins in close proximity to ERO1B during different conditions, such as ER stress or high glucose stimulation mentioned in the search results .

Antibody-based biosensors that detect conformational changes in ERO1B could provide real-time readouts of its activity in living cells. Such tools would be invaluable for understanding the dynamics of ERO1B function during normal physiology and disease states.

Organoid technology, combined with ERO1B antibody staining, could provide insights into ERO1B function in more physiologically relevant 3D models, particularly for pancreatic tissue where ERO1B plays important roles in insulin production .

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.