Recombinant Neurospora crassa Endoplasmic oxidoreductin-1 (ero-1), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you require a specific format, please specify this in your order notes.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested and agreed upon in advance. Additional charges will apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a reference for your own protocols.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
ero-1; NCU02074Endoplasmic reticulum oxidoreductin-1; EC 1.8.4.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987)
Target Names
ero-1
Uniprot No.

Target Background

Function
Essential oxidoreductase responsible for protein oxidation in the endoplasmic reticulum, forming disulfide bonds. It acts by directly oxidizing PDI1 isomerase via disulfide exchange. It doesn't directly oxidize folding substrates but relies on PDI1 to transfer oxidizing equivalents. Its selectivity for PDI1 over related proteins indicates its ability to discriminate between substrates. Reoxidation likely involves electron transfer to molecular oxygen through FAD, independent of glutathione. It may contribute significantly to cellular reactive oxygen species (ROS), potentially inducing oxidative stress.
Database Links

KEGG: ncr:NCU02074

Protein Families
EROs family
Subcellular Location
Endoplasmic reticulum membrane; Peripheral membrane protein; Lumenal side.

Q&A

What is the function of ERO-1 in Neurospora crassa?

ERO-1 is a flavin adenine dinucleotide (FAD)-containing endoplasmic reticulum (ER) oxidoreductase that catalyzes the formation of disulfide bonds in nascent polypeptides. In N. crassa, ERO-1 works in conjunction with protein disulfide isomerase (PDI) to facilitate proper protein folding. The oxidative protein folding process in the ER involves ERO-1 transferring electrons from PDI to molecular oxygen, generating hydrogen peroxide as an intermediate .

Research has shown that ERO-1's enzymatic function involves a cycle where:

  • Reduced ERO-1 utilizes molecular oxygen

  • FAD becomes reduced through electron transfer

  • Hydrogen peroxide is produced as an intermediate

  • This process enables disulfide bond formation during protein folding

ERO-1 in N. crassa is crucial for maintaining ER homeostasis, especially under conditions of high protein secretion capacity that require an efficient system for protein synthesis, folding, and transport .

How does ERO-1 relate to the unfolded protein response (UPR) in N. crassa?

The unfolded protein response (UPR) is activated when the folding capacity of the ER is exceeded. In N. crassa, ERO-1 plays a crucial role in this process:

  • Under ER stress conditions, the bZIP transcription factor HAC-1 undergoes mRNA splicing that removes a 23 nt intron, changing its open reading frame

  • This activated HAC-1 upregulates genes including ERO-1, PDI, and molecular chaperones like GRP78/BiP

  • ERO-1's activity is essential for maintaining oxidative protein folding during ER stress

Experimental evidence shows that N. crassa strains with disrupted UPR signaling (Δhac-1) fail to upregulate ERO-1 and other ER stress response genes, leading to growth defects when exposed to ER stress-inducing agents like tunicamycin . This indicates that ERO-1 is a critical downstream effector of the UPR pathway in N. crassa.

How is ERO-1 conserved across species from yeast to mammals?

ERO-1 represents a highly conserved protein family involved in ER oxidative folding:

SpeciesERO-1 HomologsDistinctive Features
Saccharomyces cerevisiaeSingle ERO1 geneEssential for viability
Neurospora crassaSingle ERO1 geneFunctions in UPR response
MammalsERO1α and ERO1βERO1α is ubiquitous; ERO1β is tissue-specific

N. crassa ERO-1 shares the core enzymatic mechanism with its homologs in other organisms, utilizing molecular oxygen as a terminal electron acceptor and generating hydrogen peroxide as an intermediate . The conservation of ERO-1 across fungal and mammalian species underscores its fundamental importance in eukaryotic protein folding mechanisms .

How does N. crassa ERO-1 coordinate with the RIP (Repeat-Induced Point mutation) defense mechanism?

N. crassa possesses a unique genome defense mechanism called Repeat-Induced Point mutation (RIP) that mutates duplicated DNA sequences during the sexual cycle. The relationship between ERO-1 and RIP involves several complex interactions:

RIP affects nearly all duplicated sequences in the N. crassa genome, creating C→T mutations . This process has profound implications for ERO-1 and other proteins:

  • RIP has blocked productive gene duplication in N. crassa, limiting the diversification of protein families including oxidoreductases

  • The extreme paucity of gene families in N. crassa (as opposed to other fungi) means that ERO-1 likely faces less functional redundancy

  • This places heightened importance on the single ERO-1 gene for maintaining ER oxidative folding capacity

Experimental evidence suggests that approximately 10,000 protein-coding genes exist in N. crassa, with RIP leaving a clear signature on the genome by preventing the fixation of duplicated genes . This evolutionary constraint may explain why N. crassa relies on a single ERO-1 enzyme, while mammals have evolved specialized isoforms (ERO1α and ERO1β) .

What are the structural and mechanistic differences between N. crassa ERO-1 and mammalian ERO1α/ERO1β?

While sharing core catalytic functions, N. crassa ERO-1 differs from mammalian ERO1 isoforms in several important aspects:

FeatureN. crassa ERO-1Mammalian ERO1αMammalian ERO1β
ExpressionUbiquitousUbiquitousTissue-specific (pancreas, stomach)
RegulationPrimarily transcriptional through HAC-1Includes post-translational phosphorylation and regulatory disulfide bridgingSimilar to ERO1α but with tissue-specific control
Cysteine arrangementContains conserved catalytic cysteinesRegulatory disulfide bridging between Cys94-Cys99 and Cys394-Cys397Similar to ERO1α with some variations

The structural differences impact inhibitor specificity. For example, the mammalian ERO1α inhibitor EN-460 forms a covalent adduct with FAD, showing an IC₅₀ of approximately 16.46 ± 3.47 μM . The efficacy of such inhibitors against N. crassa ERO-1 would likely differ due to structural variations, though both share the conserved FAD-binding domain that is crucial for oxidoreductase activity .

How does ERO-1 activity impact N. crassa's ability to degrade cellulose and adapt to its natural environment?

N. crassa is naturally found growing on dead plant material composed primarily of lignocellulose. Research has revealed a critical relationship between ERO-1/UPR activity and cellulose degradation:

  • Growth on cellulose requires secretion of numerous enzymes, imposing major demands on ER function

  • HAC-1-deficient strains (which cannot properly regulate ERO-1) show dramatically impaired growth on cellulose

  • This suggests ERO-1's oxidative folding capacity is essential for the secretion of functional cellulases

Interestingly, growth on hemicellulose, another carbon source requiring enzyme secretion, is not impaired in HAC-1 mutants, nor is the amount of protein secreted on this substrate . This suggests differential requirements for ERO-1 activity depending on the specific secretory load, with cellulose degradation being particularly dependent on optimal ER oxidative capacity.

These findings highlight ERO-1's physiological importance beyond basic cellular functions, extending to N. crassa's ecological role in plant biomass decomposition .

How can CRISPR/Cas9 be effectively used to generate N. crassa ERO-1 mutants?

Recent advances in CRISPR/Cas9 technology have facilitated genetic manipulation in N. crassa. A user-friendly system has been developed that can be adapted for ERO-1 mutagenesis:

Step-by-Step CRISPR/Cas9 Mutagenesis Protocol:

  • System Setup:

    • Utilize a strain with genomically integrated Cas9 under the control of the ccg1 promoter

    • Design guide RNAs targeting specific regions of the ERO-1 gene

    • Use cyclosporin-resistant-1 (csr-1) as a selectable marker gene

  • gRNA Design Considerations for ERO-1:

    • Target conserved regions encoding catalytic cysteines

    • Design gRNAs with minimal off-target effects using N. crassa genome databases

    • Include appropriate PAM sequences (NGG for SpCas9)

  • Efficiency Enhancement:

    • Combining gRNAs targeting ERO-1 with gRNAs targeting csr-1 can increase detection of desired mutations by up to 10-fold

    • This co-targeting approach allows for selection on cyclosporin A-containing media

This CRISPR/Cas9 system eliminates the need for constructing multiple vectors, significantly speeding up the mutagenesis process for ERO-1 .

What assays can be used to measure the enzymatic activity of N. crassa ERO-1 in vitro?

Several complementary approaches can be used to assess ERO-1 activity:

1. Oxygen Consumption Assay:

  • Measures rate of O₂ utilization using an oxygen electrode

  • Reaction mixture contains purified ERO-1, reduced PDI, and buffer

  • Rate of oxygen consumption directly correlates with ERO-1 activity

2. Hydrogen Peroxide Production Assay:

  • Uses Amplex Red reagent to detect H₂O₂ generation

  • Fluorescence increase (excitation 530 nm, emission 590 nm) indicates ERO-1 activity

  • Allows quantification of the hydrogen peroxide byproduct of ERO-1 catalysis

3. PDI Re-oxidation Assay:

  • Monitors the rate at which ERO-1 re-oxidizes reduced PDI

  • Can be tracked by following the change in fluorescence of PDI-bound fluorophores

  • Provides direct measurement of ERO-1's physiological function

4. Inhibitor Screening Method:
For testing potential ERO-1 inhibitors, a cellular thermal shift assay (CETSA) can be employed to determine isozyme specificity, similar to the approach used for mammalian ERO1α . This method can differentiate between specific binding to ERO-1 versus other FAD-containing enzymes.

These complementary assays provide a comprehensive assessment of N. crassa ERO-1 activity and can be used to evaluate the effects of mutations or potential inhibitors.

How should researchers interpret ERO-1 activity in N. crassa mutants with defects in the unfolded protein response?

When analyzing ERO-1 activity in UPR-deficient N. crassa strains, several key patterns should be considered:

Expected Observations in Δhac-1 Mutants:

ParameterNormal ConditionER Stress Condition
ERO-1 expressionBaseline levelsNo induction (unlike wild-type)
PDI oxidation statePartially reducedAbnormally reduced
Growth on celluloseMinimal growthSevere growth defect
Growth on hemicelluloseNear normalNear normal

Research has demonstrated that HAC-1 is essential for ERO-1 induction during ER stress . In Δhac-1 mutants, the absence of this transcription factor prevents upregulation of ERO-1 and other UPR target genes like grp78/bip and pdi, leading to impaired growth under ER stress conditions.

When interpreting experimental results:

  • Compare relative expression levels of ERO-1 mRNA and protein in wild-type vs. Δhac-1 strains under both normal and stress conditions

  • Assess oxidative folding capacity by measuring the ratio of reduced to oxidized PDI

  • Evaluate phenotypic consequences through growth assays on different carbon sources

These analyses can distinguish between direct effects on ERO-1 function versus indirect effects through the broader UPR pathway .

How does the genetic background of N. crassa strains affect studies of ERO-1 function?

The genetic background of N. crassa strains can significantly impact ERO-1 studies in ways that researchers must carefully address:

Genetic Background Considerations:

  • RIP Effects: Different laboratory strains may have varying degrees of RIP activity, potentially affecting ERO-1 if duplicated constructs are introduced. The paucity of gene families in N. crassa due to RIP has been extensively documented .

  • meiotic silencing effects: Unpaired DNA during meiosis can trigger silencing mechanisms in N. crassa. When introducing tagged or modified ERO-1 constructs, researchers should be aware that:

    • If the modified ERO-1 sequences don't pair properly during meiosis, they may be silenced

    • This silencing operates independently of canonical recombination pathways

    • It could lead to misinterpretation of genetic crosses involving ERO-1 mutations

To address these variables, researchers should:

  • Always report the full genetic background of strains

  • Include appropriate isogenic controls

  • Consider backcrossing mutations into a standard genetic background

  • Be aware of potential meiotic silencing effects when analyzing genetic crosses

What are the potential pitfalls when comparing ERO-1 function between N. crassa and other model systems?

When comparing ERO-1 function across species, researchers should be mindful of several important differences:

Interspecies Comparison Challenges:

  • Evolutionary Context Differences:

    • N. crassa has undergone unique evolutionary pressures due to RIP, resulting in minimal gene family redundancy

    • In contrast, organisms like S. cerevisiae and mammals have multiple oxidoreductases with overlapping functions

    • These differences can lead to misinterpretation of phenotypic severity when ERO-1 is disrupted

  • Physiological Adaptations:

    • N. crassa naturally grows on cellulose-rich materials, requiring robust secretory capacity

    • This ecological niche has shaped the relationship between ERO-1 and the UPR

    • Direct comparisons with organisms adapted to different environments may be misleading

  • Technical Considerations:

    ParameterN. crassaS. cerevisiaeMammalian Cells
    Growth temperature25-30°C30°C37°C
    Optimal pH5.85.57.4
    Redox environmentMore oxidizingLess oxidizingVariable by compartment
    Growth ratesHoursHoursDays
  • Methodological Differences:

    • Protein extraction from N. crassa often requires stronger disruption methods

    • Buffer systems optimized for yeast may be suboptimal for N. crassa proteins

    • Antibodies developed against mammalian ERO1 may show poor cross-reactivity

To overcome these challenges, researchers should:

  • Develop and use N. crassa-specific reagents when possible

  • Validate interspecies functional complementation experimentally

  • Consider the broader cellular context when interpreting phenotypes

  • Acknowledge the limitations of direct comparisons in publications

What are the promising applications of N. crassa ERO-1 research for understanding protein folding diseases?

N. crassa ERO-1 research offers unique insights into protein folding diseases through several avenues:

  • Model for Misfolding Disorders:

    • N. crassa can serve as a simplified model for studying fundamental aspects of protein misfolding

    • The absence of gene family redundancy due to RIP makes phenotypes more interpretable

    • Connection between ERO-1, UPR, and specific substrates (e.g., cellulases) provides a clear readout of folding efficiency

  • Drug Discovery Platform:

    • N. crassa ERO-1 inhibitors could serve as starting points for therapeutic development

    • Compounds like the sulfuretin derivative T151742 that inhibit mammalian ERO1α could be tested against N. crassa ERO-1

    • This would help establish evolutionary conservation of binding sites and mechanism

  • Biotechnological Applications:

    • Enhancing ERO-1 function could improve production of recombinant proteins in fungal systems

    • Understanding how N. crassa balances ER stress during high secretory load could inform bioprocessing strategies

    • The links between HAC-1, ERO-1, and cellulose utilization have direct implications for biofuel production

Future research should focus on developing genetic tools to tag and track ERO-1 in living cells, creating conditional ERO-1 mutants, and establishing high-throughput screening methods using N. crassa as a model system.

How might computational approaches advance our understanding of N. crassa ERO-1 structure and function?

Computational approaches offer powerful tools for ERO-1 research:

1. Structure Prediction and Analysis:

  • AlphaFold2 and RoseTTAFold can predict N. crassa ERO-1 structure with high confidence

2. Systems Biology Approaches:

  • Genome-scale metabolic models of N. crassa have been developed and validated

  • These models can predict the metabolic impacts of ERO-1 dysfunction

  • Integration of transcriptomic, proteomic, and metabolomic data can provide a holistic view of ERO-1's role

3. Evolutionary Analysis:

  • Comparative genomics across fungal species with different RIP activities can illuminate ERO-1 evolution

  • Ancestral sequence reconstruction methods can reveal how ERO-1 adapted to different ecological niches

  • Analysis of selection pressure on ERO-1 across lineages may identify functionally important residues

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.