ERO1L Human

ERO1-Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

ERO1L Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 468 amino acids (24-468) and having a molecular mass of 54.4 kDa.
ERO1L is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
ERO1-like protein alpha (ERO1L) is a crucial enzyme that facilitates protein oxidation and is indispensable for the proper folding of immunoglobulins. This protein interacts with protein disulfide-isomerase (PDI) to form disulfide bonds within proteins in the endoplasmic reticulum. Hypoxia stimulates ERO1L, suggesting its regulation by the hypoxia-inducible transcription factor (HIF) pathway. ERO1L expression is ubiquitous at low levels but elevates in the upper digestive tract and esophagus. Notably, ERO1L participates in releasing unfolded cholera toxin from reduced P4HB/PDI during V.cholerae infection, thereby playing a role in toxin retrotranslocation. Moreover, ERO1L has a critical role in ER stress-induced, CHOP-dependent apoptosis by activating the inositol 1,4,5-trisphosphate receptor IP3R1.
Description
This product consists of a recombinant human ERO1L protein produced in E.coli. It is a single, non-glycosylated polypeptide chain containing 468 amino acids (residues 24-468) with a molecular weight of 54.4 kDa. The protein includes a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
This product appears as a clear, colorless solution that has been sterilized by filtration.
Formulation
The ERO1L protein is provided at a concentration of 1mg/ml in a solution containing 20mM Tris-HCl buffer (pH 8.0) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), store the product at 4°C. For longer storage, freeze the product at -20°C. To maximize long-term stability, adding a carrier protein such as HSA or BSA (0.1%) is recommended. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of this protein is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms
ERO1-Like (S. Cerevisiae), Endoplasmic Oxidoreductin-1-Like Protein, Oxidoreductin-1-L-Alpha, ERO1-L-Alpha, ERO1LA, ERO1-L, ERO1 (S. Cerevisiae)-Like, ERO1-Like Protein Alpha, ERO1-Alpha, EC 1.8.4.-, EC 1.8.4, ERO1A, ERO1L.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEEQPPET AAQRCFCQVS GYLDDCTCDV ETIDRFNNYR LFPRLQKLLE SDYFRYYKVN LKRPCPFWND ISQCGRRDCA VKPCQSDEVP DGIKSASYKY SEEANNLIEE CEQAERLGAV DESLSEETQK AVLQWTKHDD SSDNFCEADD IQSPEAEYVD LLLNPERYTG YKGPDAWKIW NVIYEENCFK PQTIKRPLNP LASGQGTSEE NTFYSWLEGL CVEKRAFYRL ISGLHASINV HLSARYLLQE TWLEKKWGHN ITEFQQRFDG ILTEGEGPRR LKNLYFLYLI ELRALSKVLP FFERPDFQLF TGNKIQDEEN KMLLLEILHE IKSFPLHFDE NSFFAGDKKE AHKLKEDFRL HFRNISRIMD CVGCFKCRLW GKLQTQGLGT ALKILFSEKL IANMPESGPS YEFHLTRQEI VSLFNAFGRI STSVKELENF RNLLQNIH.

Q&A

What is ERO1L and what is its primary function in human cells?

ERO1L, also known as ERO1A or ERO1-like protein alpha, is an endoplasmic reticulum (ER) resident glycoprotein that functions as a thiol oxidoreductase. It contains flavin adenosine dinucleotide (FAD) and plays a crucial role in oxidative protein folding by catalyzing disulfide bond formation in nascent polypeptides . This process is essential for proper protein folding and maturation within the ER. ERO1L works in conjunction with protein disulfide isomerase (PDI) to facilitate the transfer of electrons, ultimately creating disulfide bonds in substrate proteins . In humans, the ERO1L gene is located on chromosome 14 .

Methodologically, to study ERO1L's function, researchers typically employ knockdown or overexpression systems using lentiviral vectors containing shRNAs or full-length ERO1L constructs, respectively. For instance, the inactive mutant form ERO1L-C394A is often used as a control to demonstrate the requirement of ERO1L's oxidoreductase activity for its function .

How does ERO1L differ from its paralog ERO1β in terms of expression and function?

In mammals, there are two ERO1 isoforms: ERO1α (ERO1L) and ERO1β. While both are involved in oxidative protein folding, they differ significantly in their expression patterns. ERO1α is expressed ubiquitously across all cell types, highlighting its fundamental role in oxidative protein folding . In contrast, ERO1β expression is more restricted, being selectively expressed in pancreatic and stomach cells, suggesting a specialized role in insulin and glucose metabolism .

From a research methodology perspective, distinguishing the functions of these paralogs requires tissue-specific knockout models or selective inhibition strategies. When studying ERO1L specifically, it's critical to account for potential compensatory mechanisms from ERO1β, especially in pancreatic tissues where both isoforms are expressed .

How is ERO1L expression altered in different cancer types and what are the prognostic implications?

ERO1L is significantly upregulated in multiple cancer types compared to normal tissues. According to comprehensive analyses across various tumor types, elevated ERO1L expression has been documented in:

  • Pancreatic ductal adenocarcinoma (PDAC) and its precursor pancreatic intraepithelial neoplasia

  • Lung adenocarcinoma (LUAD)

  • Bile duct cancer

  • Cervical cancer

  • Breast cancer

  • Liver cancer

  • Gastric cancer

Notably, prostate cancer appears to be an exception, showing no significant difference in ERO1L expression between tumor and normal tissues .

Prognostically, elevated ERO1L expression correlates with poor clinical outcomes in several cancers. In PDAC, it serves as an independent prognostic factor for patient survival . Similarly, in LUAD, high ERO1L expression is associated with poor prognosis and correlates with lymph node metastasis, TNM stage, and tumor size .

Methodologically, researchers assess ERO1L expression through techniques including immunohistochemistry of tissue microarrays, quantitative PCR, and mining public databases such as TCGA, GTEx, and Oncomine for pan-cancer expression profiles .

What molecular mechanisms underlie ERO1L's role in cancer progression?

ERO1L contributes to cancer progression through multiple molecular mechanisms:

  • Warburg Effect Regulation: ERO1L plays a critical role in cancer cell metabolism by promoting the Warburg effect (aerobic glycolysis). In PDAC, ERO1L enhances glycolysis, as evidenced by increased extracellular acidification rate (ECAR) and decreased oxygen consumption rate (OCR) . This metabolic reprogramming supports rapid tumor growth.

  • ROS Generation: ERO1L-mediated reactive oxygen species (ROS) generation is essential for its oncogenic activities. During oxidative protein folding, ERO1L produces hydrogen peroxide as a byproduct, which contributes to oxidative stress within cancer cells .

  • Signaling Pathway Activation: In LUAD, ERO1L promotes proliferation and metastasis by activating the Wnt2/β-catenin signaling pathway. ERO1L expression positively correlates with Wnt2 expression and regulates Wnt2 ubiquitination .

  • Angiogenesis Promotion: ERO1L facilitates tumor angiogenesis by enhancing oxidative protein folding of vascular endothelial growth factor (VEGF) and upregulating VEGF mRNA expression .

  • Immune Evasion: ERO1L inhibits T cell responses by recruiting myeloid-derived suppressor cells through regulation of granulocyte-colony stimulating factor and CXCL1/2 .

Methodologically, these mechanisms are typically studied using a combination of loss-of-function (shRNA, siRNA, pharmacological inhibition) and gain-of-function (overexpression of wild-type and mutant forms) approaches, followed by functional assays specific to each mechanism .

What are the most effective techniques for modulating ERO1L expression in experimental settings?

Several approaches can be employed to effectively modulate ERO1L expression in experimental settings:

For ERO1L Knockdown:

  • RNA interference: shRNAs delivered via lentiviral vectors provide stable long-term knockdown. For example, studies have successfully used puromycin selection to generate stable cell lines with ERO1L knockdown .

  • siRNA transfection: For transient knockdown, siRNAs can be transfected using reagents like Lipofectamine RNAiMAX. This approach is particularly useful for short-term experiments .

  • Pharmacological inhibition: The compound EN460 has been used as an ERO1L inhibitor in both in vitro and in vivo experiments .

  • CRISPR-Cas9 genome editing: For complete knockout studies, though this wasn't explicitly mentioned in the search results but represents a current standard approach.

For ERO1L Overexpression:

  • Lentiviral expression systems: Full-length ERO1L can be cloned into vectors like pCDH-CMV-MCS-EF1-Puro for stable overexpression .

  • Mutant forms: The inactive mutant ERO1L-C394A serves as an important control to distinguish between enzymatic and scaffolding functions of ERO1L .

The selection of approach depends on experimental goals, with stable modulation preferred for in vivo studies and long-term in vitro experiments, while transient modulation may be sufficient for short-term mechanistic studies.

How can researchers accurately measure ERO1L-mediated effects on cellular metabolism and oxidative stress?

To measure ERO1L's effects on metabolism and oxidative stress, researchers can employ several complementary techniques:

For Metabolism Assessment:

  • Seahorse XF Analyzer: This platform measures both extracellular acidification rate (ECAR, an indicator of glycolysis) and oxygen consumption rate (OCR, reflecting mitochondrial respiration) in real-time. The protocol typically involves sequential injection of glucose, oligomycin, and 2-DG for ECAR, and oligomycin, FCCP, and antimycin A/rotenone for OCR .

  • Glucose Uptake Assays: Using fluorescent glucose analogs like 2-NBDG or radiolabeled glucose (3H-2-deoxyglucose).

  • Lactate Production Measurement: Quantifying lactate in cell culture media as an indicator of glycolytic activity.

  • 18F-FDG PET/CT Imaging: In clinical samples, correlation of ERO1L expression with maximum standard uptake value (SUVmax) provides in vivo evidence of ERO1L's impact on glucose metabolism .

For Oxidative Stress Measurement:

  • ROS Detection Probes: Fluorescent dyes such as DCFDA (2',7'-dichlorofluorescin diacetate) for general ROS detection or more specific probes for hydrogen peroxide.

  • Glutathione Levels: Measuring reduced (GSH) and oxidized (GSSG) glutathione ratios as indicators of cellular redox state.

  • Protein Carbonylation Assays: To assess oxidative damage to proteins.

  • Lipid Peroxidation Measurement: Using techniques like TBARS (thiobarbituric acid reactive substances) assay.

For comprehensive analyses, these measurements should be performed in both ERO1L knockdown/inhibition and overexpression conditions, with appropriate controls including the enzymatically inactive ERO1L-C394A mutant .

How does the interplay between ERO1L and hypoxia/ER stress influence cancer cell adaptation to hostile microenvironments?

The relationship between ERO1L, hypoxia, and ER stress represents a sophisticated adaptive mechanism in cancer cell survival:

ERO1L expression is upregulated under both hypoxic conditions and ER stress , creating a regulatory feedback loop. Under hypoxia, HIF1α (Hypoxia-Inducible Factor 1-alpha) activation leads to increased ERO1L expression. Experimentally, this can be validated using siRNAs targeting HIF1α (e.g., si-HIF1α-1: GAGGAAGAACUAAAUCCAAdTdT; si-HIF1α-2: UGAUACCAACAGUAACCAAdTdT) to demonstrate the dependency of ERO1L upregulation on HIF1α .

This hypoxia-induced ERO1L expression helps cancer cells adapt to low oxygen environments by:

  • Enhancing glycolysis to maintain ATP production without requiring oxygen

  • Optimizing protein folding under stress conditions

  • Modulating redox signaling through controlled ROS production

Simultaneously, ER stress activates the Unfolded Protein Response (UPR), which also increases ERO1L expression. Analysis of TCGA cohort has revealed a specific glycolysis gene expression signature that strongly correlates with unfolded protein response-related gene signature , suggesting coordinated regulation of these pathways.

Methodologically, researchers can study this interplay using hypoxia chambers with controlled O2 levels (typically 1% O2), chemical inducers of hypoxia (e.g., cobalt chloride), and ER stress inducers (e.g., tunicamycin, thapsigargin). The resulting adaptations can be measured through cell viability assays under stress conditions, metabolic profiling, and analysis of ER stress markers (BiP/GRP78, XBP1 splicing, CHOP).

What is the relationship between ERO1L and cancer-specific protein folding requirements?

Cancer cells frequently experience elevated protein synthesis rates and unique folding challenges due to their rapid proliferation and genetic alterations. ERO1L plays a central role in addressing these cancer-specific protein folding requirements:

  • Handling Secretory Pathway Overload: Cancer cells often overexpress growth factors, cytokines, and extracellular matrix proteins that require proper disulfide bond formation. ERO1L's enhanced activity in cancer helps process this increased load of secretory proteins .

  • Folding of Cancer-Specific Proteins: ERO1L facilitates the proper folding of proteins critical for malignant phenotypes, such as VEGF for angiogenesis and possibly proteins involved in the Wnt signaling pathway .

  • Oxidative Protein Folding Network: ERO1L works in conjunction with PDI family members to maintain disulfide bond formation capacity. The ERO1L/PDI axis orchestrates proper protein folding under the increased demands of cancer cells .

  • Adaptive Response to Misfolded Proteins: Cancer-associated mutations often increase the propensity for protein misfolding. ERO1L's activity helps mitigate the accumulation of misfolded proteins that could trigger apoptosis.

To study these relationships experimentally, researchers can employ techniques such as:

  • Pulse-chase experiments with radiolabeled amino acids to track protein folding kinetics

  • Analysis of disulfide bond formation in specific cancer-related proteins

  • Protein-protein interaction studies (co-immunoprecipitation, proximity ligation assays) to identify cancer-specific ERO1L substrates

  • Proteomic analysis of the secretome in ERO1L-modulated cancer cells

How might targeting ERO1L translate into novel cancer therapeutic approaches?

ERO1L represents a promising target for cancer therapy based on multiple lines of evidence:

  • Differential Expression Pattern: ERO1L is highly expressed in various cancers compared to normal tissues, potentially providing a therapeutic window .

  • Validated Oncogenic Functions: Knockdown or pharmacological inhibition of ERO1L suppresses cancer cell proliferation in vitro and tumor growth in vivo .

  • Multiple Mechanistic Pathways: ERO1L influences cancer through various mechanisms (metabolism, ROS, signaling pathways), suggesting potential for broad anti-cancer effects .

Potential therapeutic approaches include:

Direct ERO1L Inhibition:

  • Small molecule inhibitors: EN460 has shown efficacy in preclinical models

  • Development of more specific and potent inhibitors

  • Structure-based drug design targeting the FAD-binding domain or catalytic cysteine residues

Targeting ERO1L-Dependent Pathways:

  • Combining ERO1L inhibition with glycolysis inhibitors, as inhibition of tumor glycolysis partially abrogates the growth-promoting activity of ERO1L

  • Exploiting synthetic lethality with ROS-modulating agents

  • Targeting the Wnt2/β-catenin pathway in combination with ERO1L inhibition in lung cancer

Translational Challenges:

  • The unavailability of highly specific inhibitors for ERO1L

  • Potential complications from ERO1L's interconnections with its paralog ERO1β

  • The need to carefully monitor normal tissue toxicity, particularly in pancreatic tissue where ERO1L function may be essential

Clinically, ERO1L expression could serve as a biomarker for patient stratification, potentially identifying those most likely to benefit from metabolism-targeting therapies or specific pathway inhibitors.

What are the most significant unresolved questions about ERO1L biology that require further investigation?

Despite growing understanding of ERO1L's role in cancer, several significant questions remain unresolved:

  • Substrate Specificity: Which specific proteins depend on ERO1L for proper folding in cancer cells? Comprehensive identification of ERO1L substrates would provide insight into its cancer-promoting mechanisms and potential therapeutic vulnerabilities.

  • Post-Translational Regulation: How is ERO1L activity regulated post-translationally in different cancer contexts? While we know hypoxia and ER stress upregulate ERO1L expression , less is understood about how its enzymatic activity is modulated.

  • Metabolic Network Integration: How does ERO1L coordinate with other metabolic pathways beyond glycolysis? Understanding its integration with lipid metabolism, glutaminolysis, and mitochondrial function could reveal new therapeutic opportunities.

  • Immune Modulation Mechanisms: The detailed mechanisms by which ERO1L regulates immune responses in the tumor microenvironment require further elucidation. How might ERO1L inhibition combine with immunotherapy?

  • Isoform-Specific Functions: The distinct roles of ERO1L versus ERO1β in cancer biology, particularly in pancreatic cancer where both are expressed, remain incompletely understood .

  • Resistance Mechanisms: What mechanisms might confer resistance to ERO1L inhibition? Identifying these preemptively could inform combination therapy strategies.

  • Biomarker Development: Can ERO1L expression or activity serve as a predictive biomarker for response to specific therapies? Development of robust assays for clinical use is needed.

  • Normal Tissue Function: A deeper understanding of ERO1L's role in normal physiology is essential to anticipate potential toxicities from therapeutic targeting.

To address these questions, interdisciplinary approaches combining molecular biology, biochemistry, structural biology, systems biology, and translational research will be necessary.

Product Science Overview

Structure and Function

ERO1L is an FAD-dependent protein disulfide oxidase. It works in conjunction with Protein Disulfide Isomerase (PDI) to catalyze the formation of disulfide bonds in nascent proteins. Disulfide bonds are critical for the structural integrity and function of many proteins. The activity of ERO1L is tightly regulated to ensure that the oxidative environment within the ER is maintained without causing excessive oxidative stress .

Mechanism of Action

The primary function of ERO1L is to reoxidize PDI, which in turn catalyzes the formation of disulfide bonds in substrate proteins. ERO1L transfers electrons from PDI to molecular oxygen, producing hydrogen peroxide (H₂O₂) as a byproduct. This process is vital for the proper folding of secretory and membrane proteins .

Recombinant ERO1L

Recombinant human ERO1L is produced using baculovirus expression systems in insect cells. The recombinant protein is typically tagged with a His-tag to facilitate purification. It is supplied as a carrier-free formulation, meaning it does not contain Bovine Serum Albumin (BSA), which can interfere with certain applications .

Applications

Recombinant ERO1L is used in various research applications, including:

  • Protein Folding Studies: Understanding the mechanisms of protein folding and the role of disulfide bond formation.
  • Oxidative Stress Research: Investigating the effects of oxidative stress and the role of ERO1L in maintaining ER homeostasis.
  • Drug Development: Screening for compounds that can modulate the activity of ERO1L and PDI, which may have therapeutic potential for diseases related to protein misfolding .
Storage and Stability

Recombinant ERO1L is typically stored at -20 to -70°C to maintain its stability. It is important to avoid repeated freeze-thaw cycles to prevent degradation. The protein is supplied in a solution containing Tris, NaCl, and TCEP, which helps maintain its stability during storage .

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.