Recombinant Ashbya gossypii Survival factor 1 (SVF1)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines. Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice and incurs additional charges.
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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein 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. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process. If you require a specific tag type, please inform us, and we will prioritize its development.
Synonyms
SVF1; AFL203C; Survival factor 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-385
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Ashbya gossypii (strain ATCC 10895 / CBS 109.51 / FGSC 9923 / NRRL Y-1056) (Yeast) (Eremothecium gossypii)
Target Names
SVF1
Target Protein Sequence
MLKWIQGGIS SVTGIAEPEY GPEFIHSATE RVRGKQPFHE TSRQDLAWRN ISSTHVETAT FYFTQIQTGM AGFAQIIYSK IGGLPRTAQF TFRLHHASRP ELGTWTSTKL ENFRVEGANF YADNLSLELD STATSYTLQS SVTADSVVDI TFKRLTPGVK VGEDPTTYYG DNTKEPWGTM RHVFWPRNSV NGTVVVHGEM ITLKNDYSVM ILALQGMKPH HAAKAWNFLN FHSETHSVLL MEFTTPKSYA NTKVSIGILC DKDSVLSVTI DNEVEHVKPK TDEVGWPVPK ALSMKFTGIP SSVPDKEVAS AEKLSARVDI ELKNLVERVD VMAEIPAFVK NIVSGVVGTK PYIYQYANEA LLTYAGSEYK GFAWSEVTFI SEFEN
Uniprot No.

Target Background

Function
Potentially involved in the cellular response to oxidative stress.
Database Links
Protein Families
SVF1 family
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is Ashbya gossypii Survival factor 1 (SVF1) and what is its role in cellular processes?

Survival factor 1 (SVF1) in Ashbya gossypii likely functions as a stress response protein involved in maintaining cellular viability during challenging environmental conditions. A. gossypii is a filamentous hemiascomycete with considerable biotechnological importance due to its natural ability to overproduce riboflavin (vitamin B2) . While specific SVF1 functions in A. gossypii must be experimentally determined, research on related fungi suggests SVF1 may play roles in:

  • Stress response pathways, particularly during nutrient limitation

  • Cell cycle regulation during the transition between growth phases

  • Protection against oxidative damage during metabolic processes

  • Potential involvement in the shift from trophic to productive phases

Understanding SVF1 function may provide insights into the remarkable ability of A. gossypii to survive various stressors while maintaining high production of metabolites like riboflavin.

What expression systems are most suitable for producing recombinant A. gossypii SVF1?

For laboratory-scale production of recombinant A. gossypii SVF1, several expression systems can be considered:

  • Homologous expression in A. gossypii: This approach maintains native post-translational modifications and folding environments. Strong constitutive promoters like PSED1 have proven effective for heterologous gene expression in A. gossypii .

  • Heterologous expression in S. cerevisiae: Given the close phylogenetic relationship between A. gossypii and S. cerevisiae, the latter can serve as an effective host for SVF1 expression while offering established genetic tools.

  • E. coli expression systems: For structural studies requiring high protein yields, bacterial expression may be suitable, though post-translational modifications will differ.

The optimal choice depends on research goals - homologous expression is preferred for functional studies, while bacterial systems may be more suitable for structural analysis of the protein.

How does the growth phase of A. gossypii impact SVF1 expression?

A. gossypii exhibits distinct growth phases that significantly impact gene expression patterns. Research shows that A. gossypii undergoes a trophic phase characterized by active growth followed by a productive phase where riboflavin production increases substantially .

Based on transcriptional patterns observed with other A. gossypii genes:

  • SVF1 expression may vary between trophic and productive phases

  • Transcription might be highest during stress conditions or phase transitions

  • Expression patterns could correlate with riboflavin production capacity

To determine precise SVF1 expression patterns, real-time quantitative PCR analysis across different growth phases (as performed for ADE4 and SHM2 genes) would be necessary . Understanding these temporal expression patterns would provide insights into SVF1's biological role and optimal recombinant production conditions.

What are the best methodologies for generating SVF1 knockout strains in A. gossypii?

For generating SVF1 knockout strains in A. gossypii, researchers should consider these methodological approaches:

  • Homologous recombination-based gene disruption:

    • Design a disruption cassette containing a selectable marker (such as G418 resistance) flanked by homologous regions to the SVF1 gene

    • Transform A. gossypii spores with the linearized disruption module

    • Confirm disruption by PCR analysis and Southern blotting

  • CRISPR-Cas9 system:

    • Design guide RNAs targeting SVF1

    • Co-transform with Cas9 and a repair template containing the desired modification

    • Screen transformants for successful genome editing

For verification of successful knockouts:

  • Perform analytical PCR with primers flanking the integration site

  • Conduct Southern blot analysis with appropriate probes

  • Validate the absence of SVF1 mRNA by RT-PCR or Northern blotting

This approach would mirror successful gene disruption strategies used for the BAS1 gene in A. gossypii, where integration was confirmed through both PCR and Southern blotting techniques .

How can researchers optimize purification protocols for recombinant A. gossypii SVF1?

Optimizing purification of recombinant A. gossypii SVF1 requires a systematic approach:

  • Affinity tag selection:

    • His6-tag: Most versatile for IMAC purification

    • GST-tag: Enhances solubility but adds significant size

    • FLAG or Strep-tag: Provides high specificity with milder elution conditions

  • Cell lysis optimization:

    • For A. gossypii expression: Enzymatic digestion of cell wall followed by mechanical disruption

    • Buffer composition: Include protease inhibitors and optimize pH based on SVF1's theoretical isoelectric point

  • Chromatography strategy:

    • Primary capture: Affinity chromatography based on selected tag

    • Intermediate purification: Ion exchange chromatography

    • Polishing: Size exclusion chromatography to remove aggregates

  • Quality assessment:

    • SDS-PAGE and Western blotting to confirm purity and identity

    • Mass spectrometry to verify protein integrity

    • Activity assays to confirm functional state

Testing multiple constructs with different tag positions (N-terminal versus C-terminal) is advisable, as tag placement can significantly impact protein folding and function.

What carbon sources optimize recombinant SVF1 production in A. gossypii cultures?

Optimal carbon sources for recombinant protein production in A. gossypii should be selected based on both growth promotion and gene expression considerations:

  • Glucose and xylose combinations:

    • A. gossypii strains effectively utilize mixed carbon sources containing glucose and xylose

    • Media formulations like MX2 (0.5% glucose plus 2% xylose) have proven effective for heterologous protein production

  • Complex agro-industrial substrates:

    • Corn-cob lignocellulosic hydrolysates supplemented with sugarcane or beet molasses support robust growth and production

    • These mixed formulations have enabled high yields of heterologous compounds (up to 684.5 mg/L for certain terpenes)

  • Carbon source transition strategies:

    • Initial growth on glucose followed by induction with alternative carbon sources may maximize biomass and protein production

    • Carbon source shifts could be synchronized with A. gossypii's natural transition from trophic to productive phases

Carbon source optimization should account for both biomass generation and protein expression, potentially using experimental designs that test different carbon source ratios and feeding strategies.

How does SVF1 potentially interact with the riboflavin overproduction pathway in A. gossypii?

The potential interaction between SVF1 and riboflavin overproduction pathways in A. gossypii represents an intriguing research question that connects stress response with metabolite production. Based on current understanding of A. gossypii metabolism:

  • Temporal correlation assessment:

    • Riboflavin overproduction in A. gossypii occurs during the productive phase after active growth ceases

    • SVF1, as a survival factor, may be upregulated during this transition phase

    • Temporal expression analysis using real-time quantitative PCR comparing SVF1 expression with RIB genes would reveal correlation patterns

  • Metabolic pathway interactions:

    • Riboflavin production connects to purine metabolism through GTP, a riboflavin precursor

    • SVF1 might influence purine pathway regulation, similar to the role of BAS1 transcription factor

    • Disruption or overexpression of SVF1 could alter metabolic flux through the purine and riboflavin pathways

  • Stress response connection:

    • Riboflavin overproduction has been suggested as a detoxifying and protective mechanism

    • SVF1, as a stress response protein, may coordinate this protective mechanism

    • Stress condition experiments could reveal whether SVF1 expression correlates with riboflavin production increases

This research direction could provide valuable insights into the physiological triggers for riboflavin overproduction in A. gossypii.

What post-translational modifications occur in native versus recombinant A. gossypii SVF1?

Characterizing post-translational modifications (PTMs) of native versus recombinant SVF1 is essential for understanding protein functionality and optimizing expression systems:

  • Identification of native PTMs:

    • Extraction of native SVF1 from A. gossypii cultures at different growth stages

    • Mass spectrometry analysis using techniques such as:

      • LC-MS/MS for peptide mapping and modification identification

      • Phosphoproteomics for phosphorylation site mapping

      • Glycoproteomics to identify potential glycosylation

  • Comparative analysis of recombinant versus native SVF1:

    • Expression of recombinant SVF1 in different systems (A. gossypii, S. cerevisiae, E. coli)

    • Side-by-side MS analysis to identify differences in modification patterns

    • Functional assays to determine the impact of PTM differences on protein activity

  • Engineering approaches for authentic PTM reproduction:

    • Co-expression of relevant modification enzymes in heterologous systems

    • Development of in vitro modification procedures to generate correctly modified protein

    • Assessment of modified versus unmodified protein functionality

This detailed characterization would provide valuable insights for researchers seeking to produce functionally authentic recombinant SVF1 for structural or biochemical studies.

How does genomic integration position affect recombinant SVF1 expression in A. gossypii?

The genomic context of integrated expression cassettes can significantly impact recombinant protein production in A. gossypii. Researchers should consider:

  • Targeted integration approaches:

    • Selection of genomic loci known for stable expression

    • Evaluation of different promoter-terminator combinations at the same locus

    • Assessment of chromosome position effects on expression stability

  • Comparative expression analysis:

    • Quantitative comparison of SVF1 expression levels from different integration sites

    • Evaluation of expression stability across multiple generations

    • Assessment of growth phase-dependent expression patterns at different loci

  • Design considerations:

    • Integration into native SVF1 locus (replacement) versus ectopic integration

    • Impact of nearby regulatory elements on heterologous promoter function

    • Potential for gene dosage effects through multi-copy integration

Data from similar experiments with heterologous genes in A. gossypii suggest significant variation in expression levels based on integration position . A systematic evaluation using reporter systems or direct protein quantification would provide valuable guidance for optimization of recombinant SVF1 production.

What statistical approaches are most appropriate for analyzing SVF1 expression data across different growth conditions?

For robust analysis of SVF1 expression across different growth conditions, researchers should employ these statistical approaches:

  • Normalization strategies:

    • Use multiple reference genes (e.g., ACT1, UBC6) for RT-qPCR normalization

    • Apply geNorm or NormFinder algorithms to identify the most stable reference genes under experimental conditions

    • Consider global normalization methods for RNA-seq data

  • Statistical tests for differential expression:

    • For normally distributed data: ANOVA with appropriate post-hoc tests for multi-condition comparisons

    • For non-parametric analysis: Kruskal-Wallis followed by Dunn's test

    • For time-course experiments: repeated measures ANOVA or mixed-effects models

  • Correlation analysis:

    • Pearson or Spearman correlation to associate SVF1 expression with metabolic outputs

    • Principal component analysis to identify patterns across multiple genes and conditions

    • Hierarchical clustering to identify co-regulated genes

  • Data visualization:

    • Heat maps for multi-gene, multi-condition comparisons

    • Time-course expression plots with error propagation

    • Metabolic pathway maps with expression data overlay

This approach mirrors successful expression analysis strategies used for purine biosynthesis genes in A. gossypii, where real-time quantitative PCR revealed significant regulatory patterns across growth phases .

How can researchers troubleshoot low yields of functionally active recombinant SVF1?

When confronting low yields of functionally active recombinant SVF1, researchers should implement this systematic troubleshooting approach:

  • Expression system assessment:

    • Evaluate codon optimization for the host organism

    • Test different promoter strengths and induction conditions

    • Compare homologous versus heterologous expression systems

  • Protein solubility and folding:

    • Screen buffer conditions with varying pH, salt concentration, and additives

    • Test co-expression with chaperones to improve folding

    • Evaluate fusion partners that enhance solubility (e.g., MBP, SUMO)

  • Purification optimization:

    • Modify lysis conditions to improve initial extraction

    • Test different chromatography strategies and buffer compositions

    • Implement on-column refolding for proteins recovered from inclusion bodies

  • Stability enhancement:

    • Screen stabilizing additives (glycerol, arginine, trehalose)

    • Identify and mutate protease-sensitive sites

    • Optimize storage conditions to prevent aggregation

For each intervention, functional assays should be performed to ensure that improvements in yield don't come at the expense of activity. A fractional factorial experimental design would allow efficient screening of multiple variables simultaneously.

What approaches can resolve contradictory data regarding SVF1 function in stress response versus metabolic regulation?

Resolving contradictory data regarding SVF1 function requires a comprehensive experimental strategy that bridges stress response and metabolic regulation:

  • Temporal dissection approaches:

    • High-resolution time-course experiments spanning both stress response and metabolic adaptation phases

    • Parallel monitoring of SVF1 expression, metabolic pathway activity, and stress response markers

    • Mathematical modeling to identify potential time-delayed effects

  • Conditional expression systems:

    • Develop tunable SVF1 expression systems independent of natural regulatory circuits

    • Uncouple SVF1 expression from stress conditions to examine direct metabolic effects

    • Create chimeric proteins with separable functional domains to isolate specific activities

  • Protein-protein interaction studies:

    • Conduct comprehensive interactome analysis of SVF1 under different conditions

    • Compare interacting partners during stress response versus normal growth

    • Validate key interactions through co-immunoprecipitation and functional studies

  • Multi-omics integration:

    • Combine transcriptomics, proteomics, and metabolomics data from SVF1 mutants

    • Apply network analysis to identify condition-specific regulatory patterns

    • Develop testable hypotheses that explain apparently contradictory observations

This approach, similar to studies resolving dual functions of transcription factors like BAS1 in A. gossypii , would help determine whether SVF1 has distinct or interconnected roles in stress response and metabolic regulation.

How might CRISPR-Cas9 genome editing enhance functional studies of SVF1 in A. gossypii?

CRISPR-Cas9 technology offers transformative opportunities for SVF1 functional studies in A. gossypii:

  • Precise genetic modifications:

    • Generation of point mutations to alter specific functional domains without disrupting the entire gene

    • Creation of tagged versions at the endogenous locus to study localization and interactions

    • Development of conditional alleles through insertion of regulatable elements

  • High-throughput functional genomics:

    • Systematic mutagenesis of SVF1 to create comprehensive variant libraries

    • Parallel screening of multiple genetic backgrounds with SVF1 modifications

    • Combinatorial editing of SVF1 with related genes to map genetic interactions

  • Regulatory element characterization:

    • Precise editing of promoter elements to dissect transcriptional regulation

    • Engineering of synthetic regulatory circuits to control SVF1 expression

    • Creation of reporter fusions at the endogenous locus

  • Metabolic engineering applications:

    • Optimization of SVF1 expression to enhance stress tolerance during industrial fermentation

    • Integration with other genetic modifications to improve metabolite production

    • Development of biosensor systems linked to SVF1 stress-responsive elements

This technology would significantly accelerate understanding of SVF1 function compared to traditional genetic approaches that have been used for studying genes like BAS1 in A. gossypii .

What potential roles might SVF1 play in regulating the transition between trophic and productive phases in A. gossypii?

SVF1 may function as a key regulator in the critical transition between trophic (growth) and productive (riboflavin synthesis) phases in A. gossypii:

  • Temporal expression analysis:

    • SVF1 expression patterns could be analyzed across the growth curve using real-time quantitative PCR

    • Comparison with known phase-transition markers would establish correlation with phase shifts

    • Protein levels and post-translational modifications should be monitored throughout the transition

  • Metabolic coordination:

    • SVF1 might integrate nutrient sensing with metabolic reprogramming

    • Similar to BAS1's role in regulating purine biosynthesis genes differentially during growth phases

    • SVF1 could influence the decline in purine gene transcription observed during the transition to productive phase

  • Physiological impacts of SVF1 modulation:

    • SVF1 overexpression or deletion might alter the timing of phase transition

    • Effects on the duration of trophic phase (which is prolonged in bas1 mutants)

    • Influence on metabolite accumulation patterns during productive phase

  • Stress response connection:

    • The transition to productive phase might represent a stress response

    • SVF1 could coordinate survival mechanisms including riboflavin overproduction

    • Experimental stress conditions could be used to test whether SVF1 accelerates phase transition

Understanding this regulatory role would provide valuable insights for biotechnological applications seeking to optimize growth and production phases in A. gossypii.

How can structural biology approaches inform the development of SVF1 variants with enhanced stability or function?

Structural biology offers powerful tools for engineering improved SVF1 variants:

  • Structure determination approaches:

    • X-ray crystallography of purified recombinant SVF1

    • Cryo-EM analysis for flexible regions or complexes

    • NMR spectroscopy for dynamic elements and ligand interactions

    • Integrative modeling combining experimental data with computational prediction

  • Structure-guided engineering strategies:

    • Identification of stability-limiting regions through analysis of B-factors and molecular dynamics

    • Rational design of disulfide bonds or salt bridges to enhance thermostability

    • Modification of solvent-exposed hydrophobic patches to improve solubility

    • Engineering of substrate binding sites or interaction interfaces for altered function

  • Experimental validation pipeline:

    • High-throughput thermal shift assays to screen stability-enhanced variants

    • Functional assays to ensure improved stability doesn't compromise activity

    • In vivo testing in A. gossypii to confirm enhanced properties in the native context

  • Application to industrial contexts:

    • Development of SVF1 variants optimized for different stress conditions

    • Engineering of variants with altered regulatory properties for biotechnological applications

    • Creation of biosensors based on SVF1 conformational changes

This approach would mirror successful protein engineering strategies employed for industrial enzymes, providing variants with enhanced properties for both research and biotechnological applications.

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.