Recombinant Ashbya gossypii Succinate dehydrogenase [ubiquinone] cytochrome b small subunit, mitochondrial (SDH4)

Shipped with Ice Packs
In Stock

Description

Production and Purification

The recombinant SDH4 is produced via heterologous expression in E. coli . Key protocols include:

  • Expression System: E. coli BL21(DE3) or similar strains .

  • Purification: Affinity chromatography using Ni-NTA resin for His-tag isolation, followed by size-exclusion chromatography .

  • Purity: >90% as verified by SDS-PAGE .

  • Storage: Lyophilized powder in Tris/PBS buffer with 6% trehalose (pH 8.0); stable at -20°C/-80°C .

Reconstitution Guidelines:

  • Centrifuge lyophilized protein before reconstitution in sterile water (0.1–1.0 mg/mL) .

  • Glycerol (5–50%) is recommended for long-term storage .

Functional and Biochemical Properties

SDH4 anchors the SDH complex to the mitochondrial inner membrane, enabling electron transfer from succinate to ubiquinone . Comparative studies highlight its structural homology to SDH4 subunits in Saccharomyces cerevisiae and Schizosaccharomyces pombe, with conserved transmembrane domains critical for ubiquinone binding .

Key Functional Domains:

  • Two transmembrane helices (residues 15–37 and 44–61) .

  • Conserved heme-binding motifs (absent in A. gossypii, unlike bacterial homologs) .

Metabolic Engineering

  • SDH4 is a target for optimizing A. gossypii’s industrial applications, such as riboflavin and terpene production . Disruption of SDH-related genes alters metabolic flux toward target metabolites .

  • Engineered A. gossypii strains with modified SDH activity show enhanced carbon source utilization, including lignocellulosic waste .

Comparative Analysis with Other Systems

FeatureA. gossypii SDH4S. cerevisiae SDH4Schizosaccharomyces pombe SDH4
Length (aa)61 120 105
His TagN-terminal None C-terminal
Expression HostE. coli Pichia pastoris E. coli
Purity>90% >85% >85%

Research Significance

  • Biotechnological Relevance: A. gossypii is a model organism for riboflavin overproduction, and SDH4 engineering could enhance its efficiency in utilizing non-conventional carbon sources (e.g., glycerol, xylose) .

  • Evolutionary Insights: SDH4’s minimalistic structure in A. gossypii provides clues about fungal mitochondrial evolution .

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them when placing your order, and we will accommodate your needs.
Lead Time
Delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Note: All our proteins are shipped with standard blue ice packs by default. If dry ice shipping is required, please communicate this beforehand, as additional fees will apply.
Notes
Repeated freezing and thawing is discouraged. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging this vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile 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 final glycerol concentration is 50%, which can serve as a reference for your own preparations.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein itself.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. Lyophilized forms have a shelf life of 12 months 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 will be determined during the manufacturing process.
The tag type is determined during production. If you have a specific tag type in mind, please inform us, and we will prioritize its development.
Synonyms
SDH4; AGL137W; Succinate dehydrogenase [ubiquinone] cytochrome b small subunit, mitochondrial; CybS; Succinate-ubiquinone reductase membrane anchor subunit
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-61
Protein Length
full length protein
Species
Ashbya gossypii (strain ATCC 10895 / CBS 109.51 / FGSC 9923 / NRRL Y-1056) (Yeast) (Eremothecium gossypii)
Target Names
SDH4
Target Protein Sequence
MQGIKMASPVIRAACKRSLHQSAVRAITIPFLPTLPQNPGGVKGDVNEANPVPPANKMHG S
Uniprot No.

Target Background

Function
This protein serves as the membrane-anchoring subunit of succinate dehydrogenase (SDH), which is a component of complex II within the mitochondrial electron transport chain. Its primary function is to facilitate the transfer of electrons from succinate to ubiquinone (coenzyme Q).
Database Links
Protein Families
CybS family
Subcellular Location
Mitochondrion inner membrane; Multi-pass membrane protein.

Q&A

What is the biological role of SDH4 in Ashbya gossypii metabolism?

SDH4 in Ashbya gossypii functions as the membrane anchor subunit of succinate dehydrogenase (Complex II), a crucial enzyme that participates in both the tricarboxylic acid (TCA) cycle and the electron transport chain in mitochondria. This dual functionality places SDH4 at a critical junction in cellular respiration, where it contributes to both energy production through oxidative phosphorylation and intermediate metabolism. In A. gossypii, which is a natural riboflavin overproducer, SDH4 plays a particularly important role in maintaining redox balance and providing metabolic precursors for various biosynthetic pathways, including those involved in riboflavin production. Studies of A. gossypii metabolism suggest that proper functioning of the TCA cycle components, including SDH4, is essential for normal hyphal growth and riboflavin biosynthesis .

How does the SDH4 protein structure in A. gossypii compare to homologous proteins in other model organisms?

The SDH4 protein in A. gossypii shares significant structural homology with its counterparts in other fungi, particularly Saccharomyces cerevisiae, with which A. gossypii has "remarkable similarities at the synteny level" . The protein contains a single transmembrane domain that anchors the SDH complex to the inner mitochondrial membrane. Comparative structural analyses reveal that the A. gossypii SDH4 contains highly conserved histidine residues that coordinate the heme group, which is essential for electron transfer during oxidative phosphorylation. Unlike S. cerevisiae, which underwent whole genome duplication, A. gossypii lacks sequence duplications, potentially making it a simpler model for studying the fundamental properties of this protein . The evolutionary relationship between these organisms provides valuable insights into the conservation of essential mitochondrial proteins across fungal species and can inform structural studies aimed at understanding protein function.

What are the optimal heterologous expression systems for producing recombinant A. gossypii SDH4?

For the heterologous expression of recombinant A. gossypii SDH4, several expression systems have been evaluated, with the following considerations for optimal production:

Expression SystemAdvantagesChallengesYield (mg/L culture)Recommended Tags
E. coli (BL21 DE3)Fast growth, high yield, economicalPotential improper folding, lacks post-translational modifications5-10 (with optimization)N-terminal His6-tag with TEV cleavage site
Pichia pastorisEukaryotic processing, high-density culturesLonger cultivation time, more complex media8-15C-terminal His6-tag
S. cerevisiaeSimilar cellular machinery to A. gossypiiLower yields than P. pastoris3-7Dual His6-FLAG tag

For membrane proteins like SDH4, expression in yeast systems generally produces more properly folded protein with correct post-translational modifications. When using E. coli, incorporation of the protein into inclusion bodies is a common issue, requiring refolding protocols. The addition of 0.5-1% Triton X-100 or n-dodecyl-β-D-maltoside (DDM) during cell lysis significantly improves protein solubilization. Additionally, co-expression with other succinate dehydrogenase subunits has been shown to enhance stability and proper folding of recombinant SDH4. For optimal expression, induction conditions should be carefully controlled with temperature reduced to 16-18°C during induction phase to minimize aggregation.

How should researchers optimize codon usage for expressing A. gossypii SDH4 in heterologous systems?

Codon optimization is crucial for efficient heterologous expression of A. gossypii SDH4, particularly given the significant differences in codon bias between A. gossypii and common expression hosts. A methodological approach should include:

  • Codon Adaptation Index (CAI) analysis: Compare the native A. gossypii SDH4 sequence CAI with the expression host. For E. coli expression, codons with usage frequency below 20% in the host should be replaced.

  • GC content adjustment: The natural A. gossypii genome has a GC content of approximately 52% , which may need adjustment depending on the expression host. For E. coli expression, maintaining a GC content between 40-60% in the optimized sequence is recommended.

  • Removal of problematic sequence elements: Eliminate internal Shine-Dalgarno-like sequences, cryptic splice sites, and premature polyadenylation signals that could interfere with proper expression.

  • mRNA secondary structure prediction: Using algorithms like Mfold to predict and minimize stable secondary structures in the 5' region of the mRNA, particularly in the first 40-50 nucleotides, which can impede translation initiation.

What are the most effective chromatography techniques for purifying recombinant A. gossypii SDH4?

Purification of recombinant A. gossypii SDH4 presents unique challenges due to its membrane-associated nature. A multi-step chromatography approach is recommended:

  • Initial Detergent Solubilization: Prior to chromatography, solubilize mitochondrial membranes containing SDH4 in a buffer containing 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 1% DDM or 1% digitonin. Incubate for 1 hour at 4°C with gentle rotation.

  • Immobilized Metal Affinity Chromatography (IMAC): For His-tagged SDH4, use Ni-NTA resin equilibrated with buffer containing 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, and 0.1% DDM. After binding, wash with increasing imidazole concentrations (10-40 mM) to remove non-specific binding proteins before elution with 250-300 mM imidazole.

  • Size Exclusion Chromatography (SEC): Apply IMAC-purified protein to a Superdex 200 column equilibrated with 20 mM HEPES (pH 7.0), 150 mM NaCl, and 0.05% DDM to separate monomeric SDH4 from aggregates and potential contaminating proteins.

  • Ion Exchange Chromatography (Optional): For samples requiring higher purity, a final polishing step using anion exchange (Q Sepharose) can be performed.

The choice of detergent is critical throughout the purification process. DDM has been widely successful for SDH4 purification, but LMNG (lauryl maltose neopentyl glycol) at 0.01-0.05% provides enhanced stability for long-term storage and crystallization attempts. Throughout purification, maintain temperature at 4°C and include protease inhibitors to prevent degradation. Typical yields from optimized purification protocols range from 0.5-2 mg of pure SDH4 per liter of yeast culture, with protein purity >95% as assessed by SDS-PAGE and protein-specific Western blotting.

How can researchers assess the functional integrity of purified recombinant A. gossypii SDH4?

Assessing the functional integrity of purified recombinant A. gossypii SDH4 requires multiple complementary approaches:

  • Succinate:Ubiquinone Oxidoreductase Activity Assay: Measure the rate of 2,6-dichlorophenolindophenol (DCPIP) reduction in the presence of phenazine methosulfate (PMS), succinate, and purified SDH4 reconstituted with other SDH subunits. A functionally intact SDH complex shows activity in the range of 0.5-2 μmol DCPIP reduced/min/mg protein at 30°C.

  • Spectroscopic Characterization: UV-visible spectroscopy can verify the presence of properly incorporated heme b in SDH4, with characteristic absorption peaks at approximately 560 nm and 430 nm in the reduced state. The reduced/oxidized spectral ratio provides a quantitative measure of heme incorporation.

  • Thermal Shift Assays: Using differential scanning fluorimetry with SYPRO Orange to determine protein stability. Properly folded A. gossypii SDH4 typically exhibits a melting temperature (Tm) between 45-55°C, with higher values indicating better stability.

  • Lipid Reconstitution and Membrane Potential Measurements: Incorporate purified SDH4 into liposomes and measure proton pumping activity using pH-sensitive fluorescent dyes or membrane potential-sensitive probes.

  • Co-purification Analysis: Verify the ability of recombinant SDH4 to associate with other SDH subunits through pull-down assays followed by Western blotting or mass spectrometry.

A combination of these methods provides comprehensive assessment of structural integrity and functional activity. When optimizing purification conditions, researchers should monitor activity retention after each purification step, with specific activities typically declining by 10-15% after IMAC and an additional 5-10% after SEC. The presence of 10% glycerol and 1 mM DTT in storage buffers can help maintain functional integrity during storage at -80°C for up to 6 months.

What are the most efficient methods for generating A. gossypii SDH4 gene knockouts or conditional mutants?

For generating A. gossypii SDH4 gene knockouts or conditional mutants, several approaches have proven effective with varying advantages depending on research objectives:

  • PCR-Based Gene Targeting: This method utilizes homologous recombination with PCR-amplified cassettes containing a selection marker flanked by sequences homologous to regions upstream and downstream of the SDH4 gene. For A. gossypii, 45-60 bp homology regions typically yield transformation efficiencies of 15-30%, which is significantly higher than in S. cerevisiae due to A. gossypii's efficient homologous recombination machinery. Selection markers such as GEN3 (G418 resistance) or NAT1 (nourseothricin resistance) work effectively in A. gossypii .

  • CRISPR-Cas9 System: More recently adapted for A. gossypii, this system allows precise genome editing. Design sgRNAs targeting the SDH4 locus and co-transform with a Cas9 expression vector and a repair template. This method increases editing efficiency to 40-60% and reduces off-target effects.

  • Conditional Expression Systems: For essential genes like SDH4, conditional systems are crucial:

    • Tetracycline-repressible promoter system: Replace the native SDH4 promoter with a tetO7-CYC1 promoter, allowing gene expression to be turned off by adding doxycycline

    • Temperature-sensitive degron tags: Fuse an N-terminal temperature-sensitive degron to SDH4, allowing protein degradation at elevated temperatures (37°C)

For phenotypic analysis of SDH4 mutants in A. gossypii, it's essential to monitor both growth characteristics and riboflavin production, as SDH4 disruption typically affects both processes due to altered mitochondrial function. Growth should be assessed at different temperatures (typically 16°C, 30°C, and 37°C) and on various carbon sources (glucose, glycerol, ethanol) to fully characterize respiratory capacity .

How can researchers perform site-directed mutagenesis to study specific residues in A. gossypii SDH4?

Site-directed mutagenesis of A. gossypii SDH4 requires careful methodological planning to ensure efficient mutation incorporation and subsequent functional analysis:

  • Selection of Target Residues: Priority should be given to residues involved in:

    • Heme binding (conserved histidine residues)

    • Membrane anchoring (hydrophobic transmembrane regions)

    • Interaction interfaces with other SDH subunits

    • Ubiquinone binding sites

  • Mutagenesis Strategy:

    • For plasmid-based mutagenesis: Use overlap extension PCR with mutagenic primers or commercial kits like QuikChange (Agilent) or Q5 Site-Directed Mutagenesis (NEB)

    • For genomic integration: Generate a complete SDH4 cassette containing the desired mutation(s) and homology regions for integration at the native locus

  • Verification of Mutations:

    • Sequence the entire SDH4 gene to confirm the intended mutation and absence of secondary mutations

    • Verify expression levels of mutant protein are comparable to wild-type using Western blotting

  • Functional Impact Assessment:

    • Enzymatic activity measurements comparing wild-type and mutant proteins

    • Growth complementation assays in SDH4-deficient strains

    • Mitochondrial membrane potential measurements using fluorescent dyes like JC-1 or TMRM

    • Reactive oxygen species (ROS) production quantification, as SDH4 mutations often affect ROS generation

When studying heme-binding residues, the double histidine mutant (typically H56A/H89A, using A. gossypii numbering) serves as an important negative control as it abolishes heme binding. For suspected ubiquinone-binding residues, complementary approaches including isothermal titration calorimetry (ITC) with ubiquinone analogs can provide binding affinity data. Mutations in the transmembrane domain may require more extensive analysis including lipid bilayer incorporation studies to assess membrane integration efficiency.

What analytical techniques are most informative for studying the interaction between recombinant A. gossypii SDH4 and other subunits of the succinate dehydrogenase complex?

Several complementary analytical techniques provide valuable insights into the interactions between recombinant A. gossypii SDH4 and other SDH subunits:

  • Blue Native Polyacrylamide Gel Electrophoresis (BN-PAGE): This technique separates intact protein complexes under native conditions. When combined with Western blotting using subunit-specific antibodies, it can reveal the assembly state of the SDH complex containing recombinant SDH4. For optimal results, solubilize mitochondrial membranes in 1% digitonin at a detergent:protein ratio of 4:1, and use a 4-16% gradient gel.

  • Co-immunoprecipitation (Co-IP): Using antibodies against epitope-tagged SDH4 to pull down associated proteins, followed by mass spectrometry or Western blotting. This approach can identify not only the canonical SDH subunits but also potential assembly factors or novel interacting partners.

  • Surface Plasmon Resonance (SPR): For quantitative measurement of binding kinetics between SDH4 and other subunits. Immobilize purified SDH4 on a sensor chip and measure real-time association/dissociation of other purified SDH subunits. This provides affinity constants (KD) typically in the nanomolar range for proper subunit interactions.

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): This technique maps the protein regions involved in subunit interactions by measuring the rate of hydrogen/deuterium exchange in peptides, which is reduced at protein-protein interfaces. When comparing exchange rates of isolated SDH4 versus the assembled complex, regions showing reduced exchange identify interaction interfaces.

  • Cryo-Electron Microscopy (Cryo-EM): For structural characterization of the assembled SDH complex containing recombinant SDH4. Recent advances allow resolution of 3-4 Å, sufficient to identify key interaction residues.

  • Crosslinking Mass Spectrometry (XL-MS): Using chemical crosslinkers like BS3 or DSS to covalently link interacting regions, followed by protease digestion and mass spectrometry to identify crosslinked peptides. This provides spatial constraints for modeling subunit arrangements.

When applying these techniques to A. gossypii SDH4, it's important to consider that optimal detergent choice varies by method. While digitonin preserves native interactions for BN-PAGE, DDM often works better for Co-IP studies. For Cryo-EM, newer amphipathic polymers like amphipols or nanodiscs have shown superior results in maintaining complex integrity during grid preparation. The integration of data from multiple techniques provides the most complete understanding of complex assembly and subunit interactions.

How can researchers determine the effect of mutations in A. gossypii SDH4 on enzyme kinetics and electron transport?

To comprehensively evaluate the functional impact of mutations in A. gossypii SDH4 on enzyme kinetics and electron transport, researchers should employ a multi-faceted approach:

  • Steady-State Enzyme Kinetics: Compare wild-type and mutant SDH complex activity using:

    • Succinate oxidation assay: Measure the rate of succinate-dependent reduction of artificial electron acceptors like DCPIP or ferricyanide

    • Determine Km for succinate (typically 0.1-0.5 mM for wild-type) and Vmax values

    • Calculate catalytic efficiency (kcat/Km) for quantitative comparison

    • Measure kinetic parameters at different pH values (5.5-8.0) and temperatures (25-40°C) to identify condition-dependent effects

  • Quinone Binding and Reduction Kinetics:

    • Use a series of ubiquinone analogs with varying side chain lengths (UQ1-UQ10) to probe the quinone binding site

    • Measure the reduction rates with different quinones to determine specificity changes in mutants

    • For mutations suspected to affect quinone binding, determine IC50 values for competitive quinone-site inhibitors like thenoyltrifluoroacetone (TTFA) or atpenin A5

  • Electron Paramagnetic Resonance (EPR) Spectroscopy:

    • Characterize the [2Fe-2S], [4Fe-4S], and [3Fe-4S] clusters in the assembled complex containing mutant SDH4

    • Compare signal intensities and g-values between wild-type and mutant proteins

    • Perform temperature-dependent EPR (10-40K) to distinguish between different iron-sulfur clusters

  • Membrane Potential Measurements in Reconstituted Systems:

    • Incorporate purified wild-type or mutant SDH complexes into liposomes

    • Use potential-sensitive fluorescent dyes like Oxonol VI or TMRM to quantify succinate-dependent membrane potential generation

    • Calculate the H⁺/e⁻ ratio to assess proton pumping efficiency

  • Reactive Oxygen Species (ROS) Production:

    • Quantify superoxide and hydrogen peroxide production using fluorescent probes like MitoSOX or Amplex Red

    • Compare ROS production rates at different succinate concentrations (0.1-10 mM)

    • Particularly important for mutations near the ubiquinone binding site, as these often affect ROS generation

SDH4 Mutation TypeExpected Enzymatic EffectDiagnostic AssayTypical Results in Mutants
Heme coordinationReduced electron transfer rateCytochrome b spectrum analysisLoss of characteristic 560/430 nm peaks
Quinone binding siteAltered Km for ubiquinoneDCPIP reduction kinetics with variable ubiquinone3-10 fold increase in Km for ubiquinone
Membrane anchor residuesDisrupted complex assemblyMembrane fractionation analysisDecreased stability in detergent solutions
Inter-subunit contactsReduced complex stabilityThermal inactivation assaysLower melting temperature by 5-10°C

For all kinetic measurements, proper control experiments are essential. These include parallel analysis of equivalent mutations in the well-characterized S. cerevisiae Sdh4p and comparison with known SDH inhibitors to distinguish between specific mechanistic effects and general perturbation of enzyme structure.

How can researchers utilize A. gossypii SDH4 studies to better understand mitochondrial disease mechanisms?

A. gossypii SDH4 provides a valuable model for understanding mitochondrial diseases associated with succinate dehydrogenase dysfunction for several research-relevant reasons:

  • Evolutionary conservation: The high degree of conservation between A. gossypii SDH4 and human SDHD allows for functional studies of disease-associated mutations. Researchers can introduce equivalent human SDHD mutations into A. gossypii SDH4 and evaluate their phenotypic effects in a simplified eukaryotic system. This approach has several advantages:

    • A. gossypii can survive with compromised mitochondrial function by utilizing alternative metabolic pathways, allowing characterization of mutations that would be lethal in higher organisms.

    • The filamentous growth pattern provides visible phenotypes (hyphal extension rates, branching patterns) that correlate with mitochondrial function .

    • A. gossypii's haploid nature eliminates complications from heterozygosity when interpreting mutation effects.

  • Systematic mutation analysis: Create a library of A. gossypii strains carrying SDH4 mutations corresponding to human SDHD variants associated with:

    • Hereditary paraganglioma

    • Pheochromocytoma

    • Gastrointestinal stromal tumors

    • Carney-Stratakis syndrome

  • Metabolomic profiling: Quantify metabolic changes resulting from SDH4 mutations:

    • Measure succinate accumulation and succinate/fumarate ratios using LC-MS/MS

    • Detect compensatory metabolic pathway activation

    • Identify novel metabolic biomarkers of SDH dysfunction

  • Mitochondrial dynamics assessment: Investigate how SDH4 mutations affect:

    • Mitochondrial network morphology using fluorescent proteins targeted to mitochondria

    • Mitochondrial membrane potential measured with JC-1 or TMRM dyes

    • Mitochondrial turnover through autophagy (mitophagy) processes

  • ROS signaling and damage: Quantify oxidative stress parameters:

    • Measure lipid peroxidation products (MDA, 4-HNE)

    • Assess protein carbonylation levels

    • Quantify mitochondrial DNA damage

The resulting data can be compiled into a comprehensive phenotypic database correlating specific SDH4 mutations with cellular consequences, providing valuable insights for predicting clinical outcomes of equivalent mutations in humans. This approach is particularly valuable for cataloging variants of uncertain significance (VUS) found in human patients, as functional studies in A. gossypii can help classify their pathogenicity.

What are the most promising approaches for using A. gossypii SDH4 in biotechnological applications related to metabolic engineering?

The strategic manipulation of A. gossypii SDH4 presents several promising avenues for metabolic engineering applications, particularly in redirecting carbon flux and optimizing riboflavin production:

  • Fine-tuning TCA cycle flux: Modulating SDH4 expression or activity can redirect carbon flow:

    • Controlled downregulation of SDH4 causes succinate accumulation, which can be channeled toward commercially valuable succinate production

    • Partial inhibition of SDH activity increases NADH:NAD+ ratio, potentially enhancing riboflavin yields by increasing the availability of reducing equivalents required for biosynthesis

  • Engineering electron transport chain efficiency:

    • Site-directed mutagenesis of SDH4 ubiquinone binding site can alter the enzyme's affinity for electron acceptors

    • Optimizing the ratio of electrons flowing through Complex II versus other respiratory complexes can balance ATP production against biosynthetic needs

  • Enhancing cellular redox state management:

    • Creating SDH4 variants with reduced propensity for reactive oxygen species generation

    • Engineering A. gossypii strains with modified SDH4 that maintain optimal NADH/FADH2 ratios under industrial fermentation conditions

  • Improving stress tolerance for industrial applications:

    • SDH4 mutations affecting mitochondrial membrane composition can enhance resistance to osmotic stress

    • Modified SDH4 proteins with increased thermostability can improve strain performance in high-temperature industrial processes

Modification ApproachTarget PathwayExpected OutcomeIndustrial Relevance
SDH4 promoter replacement with controllable promoterTCA cycle fluxTunable succinate:fumarate ratioEnhanced riboflavin or organic acid production
SDH4 heme-binding site engineeringElectron transport chain efficiencyAltered ROS productionImproved strain longevity in industrial fermentations
SDH4 transmembrane domain modificationsMitochondrial membrane propertiesEnhanced stress toleranceBetter performance in non-optimal industrial conditions
Chimeric SDH4 with domains from thermophilic organismsProtein thermostabilityFunctional mitochondria at higher temperaturesReduced cooling costs in industrial processes

For implementing these approaches, modern synthetic biology tools are essential. These include:

  • CRISPR-Cas9 genome editing for precise modification of the native SDH4 locus

  • Inducible promoter systems that respond to affordable industrial inducers

  • Biosensor-based selection systems that can detect desired metabolic states

  • Proteomic analysis to verify the impact of SDH4 modifications on the entire mitochondrial proteome

The most successful strategies will likely involve combinatorial approaches, simultaneously targeting SDH4 along with other key enzymes in central carbon metabolism. When designing such strategies, researchers should consider the pH dependence of A. gossypii growth, as the organism shows "substantial growth at pH 4.5 only on complex medium" and grows better at higher pH values (6.5) with defined nitrogen sources like ammonium .

What are the critical factors to consider when designing experiments to study the role of A. gossypii SDH4 in riboflavin production?

When investigating the relationship between A. gossypii SDH4 and riboflavin production, researchers should carefully consider several critical experimental design factors:

  • Strain selection and validation:

    • Use the widely studied A. gossypii ATCC 10895 strain as a reference point for comparison with industrial overproducing strains

    • Verify SDH4 sequence integrity in all experimental strains through sequencing

    • Confirm consistent mitochondrial copy number across experimental and control strains, as variations can confound interpretation of SDH4-specific effects

  • Media composition optimization:

    • Define precise carbon and nitrogen sources, as A. gossypii shows variable growth on different substrates

    • Maintain pH control with appropriate buffers, as A. gossypii growth is pH-dependent with optimal growth at pH 6.5 for defined media

    • Consider trace element supplementation, particularly iron, which affects both SDH4 function (heme coordination) and riboflavin biosynthesis pathways

  • Experimental design for causality determination:

    • Implement genetic complementation studies with wild-type SDH4 to confirm phenotype specificity

    • Create a titration series of SDH4 expression levels using regulatable promoters to establish dose-response relationships

    • Develop parallel experiments with specific SDH inhibitors to distinguish enzymatic from structural roles of SDH4

  • Comprehensive phenotypic assessment:

    • Monitor growth parameters (biomass accumulation, hyphal extension rate, branching frequency) alongside riboflavin production

    • Track dissolved oxygen levels continuously, as oxygen availability affects both respiration and riboflavin biosynthesis

    • Measure key TCA cycle intermediates (particularly succinate, fumarate, and malate) at multiple time points during cultivation

  • Analytical methodology considerations:

    • Standardize riboflavin extraction and quantification methods (preferably HPLC with fluorescence detection)

    • Implement appropriate internal standards to account for extraction efficiency variations

    • Consider analyzing both intracellular and extracellular riboflavin pools separately

A particularly effective experimental approach is to combine SDH4 modifications with isotopic flux analysis using 13C-labeled substrates. This provides direct evidence of how carbon flow through the TCA cycle relates to riboflavin production capacity. When designing such experiments, researchers should include controls for:

  • Potential pleiotropic effects of SDH4 modification on other metabolic pathways

  • Changes in gene expression of riboflavin biosynthetic enzymes (rib genes) in response to altered SDH4 function

  • Effects of growth stage on the relationship between SDH4 activity and riboflavin production

The use of bioreactors with precise control of pH, dissolved oxygen, and nutrient feeding is strongly recommended for reproducible results, as shake flask cultures often show high variability in both growth and riboflavin production.

How should researchers approach the integration of proteomics and metabolomics data when studying the impact of SDH4 mutations on A. gossypii metabolism?

The integration of proteomics and metabolomics provides powerful insights into the systemic effects of SDH4 mutations on A. gossypii metabolism. A methodical approach should include:

  • Coordinated sample collection and preparation:

    • Harvest cells at multiple defined growth phases (early exponential, mid-exponential, early stationary)

    • Process parallel samples for both proteomics and metabolomics from the same cultures

    • Include subcellular fractionation to specifically analyze mitochondrial proteome changes

    • Implement rapid quenching procedures (e.g., cold methanol at -40°C) to capture accurate metabolic snapshots

  • Comprehensive analytical platforms:

    • Proteomics:

      • Use both shotgun proteomics for global protein identification and targeted approaches for low-abundance mitochondrial proteins

      • Implement SILAC or TMT labeling for accurate quantitative comparison between wild-type and mutant strains

      • Include phosphoproteomic analysis to detect regulatory changes in metabolic enzymes

    • Metabolomics:

      • Deploy both targeted and untargeted approaches using LC-MS/MS

      • Implement methods optimized for different metabolite classes (organic acids, amino acids, nucleotides)

      • Measure metabolite turnover rates using isotope pulse-chase experiments

  • Data integration strategies:

    • Map identified proteins and metabolites onto known A. gossypii metabolic pathways

    • Develop correlation networks between protein abundance changes and metabolite level alterations

    • Implement multivariate statistical approaches (PCA, PLS-DA) to identify patterns in the combined datasets

    • Use genome-scale metabolic models of A. gossypii to predict the systemic effects of observed changes

  • Validation experiments:

    • Confirm key findings with targeted enzyme activity assays

    • Verify metabolic flux changes using 13C metabolic flux analysis

    • Test predictions from integrated analyses using specific metabolic inhibitors or genetic modifications

A particularly valuable approach is to analyze the temporal sequence of changes following SDH4 mutation or expression modulation. This reveals:

Time PointProteomic FocusMetabolomic FocusIntegration Goal
Immediate response (0-2h)Post-translational modificationsRapid metabolite pool changesIdentify direct regulatory responses
Short-term adaptation (2-12h)Protein expression changes in mitochondriaCentral carbon metabolism shiftsMap compensatory metabolic pathways
Long-term adaptation (>12h)Global proteome remodelingSecondary metabolite profile changesUnderstand systemic adaptation strategies

When implementing this framework, researchers should be mindful of the unique physiological characteristics of A. gossypii. The filamentous growth pattern creates heterogeneity within cultures, with older hyphal segments potentially showing different metabolic profiles than actively growing tips. Single-cell or single-hypha approaches, while technically challenging, may reveal important spatial aspects of metabolic adaptation to SDH4 mutations that bulk analyses would miss.

What are the most effective solutions for troubleshooting heterologous expression issues with recombinant A. gossypii SDH4?

When encountering challenges with heterologous expression of recombinant A. gossypii SDH4, researchers can implement the following systematic troubleshooting strategies:

  • Low expression levels:

    • Optimize codon usage for the expression host, particularly focusing on the first 15-20 codons after the start codon

    • Screen multiple promoter-terminator combinations to identify optimal expression control elements

    • Test different fusion tags (MBP, SUMO, GST) that can enhance solubility and expression levels

    • Implement low-temperature expression protocols (16-20°C) with extended induction times

    • For E. coli expression, co-express molecular chaperones (GroEL/ES, DnaK/J) to assist proper folding

  • Protein aggregation/inclusion body formation:

    • Include mild solubilizing agents (0.5-1% Triton X-100, 5-10% glycerol) in lysis buffers

    • Test multiple detergents for membrane protein solubilization (DDM, LMNG, digitonin) at various concentrations

    • Express SDH4 as part of a larger fusion with a highly soluble partner protein

    • For extreme cases, develop refolding protocols from solubilized inclusion bodies using gradual detergent dialysis

  • Proteolytic degradation:

    • Identify and remove predicted protease recognition sites through conservative mutagenesis

    • Include multiple protease inhibitors (PMSF, EDTA, leupeptin, pepstatin A) during purification

    • Test multiple expression hosts to identify systems with lower proteolytic activity

    • Optimize cell lysis and protein purification to minimize processing time

  • Poor function/misfolding:

    • Co-express SDH4 with other SDH subunits to promote proper complex assembly

    • Include heme precursors (δ-aminolevulinic acid) in the growth medium to ensure adequate heme availability

    • Test expression in eukaryotic hosts (Pichia pastoris, S. cerevisiae) that provide more appropriate folding environments

    • Implement in vitro transcription/translation systems supplemented with appropriate membrane mimetics

IssueDiagnostic TestSolution StrategySuccess Indicators
Low expressionWestern blot with anti-tag antibodiesOptimize expression vector elements≥3-fold increase in band intensity
Inclusion bodiesFractionation and SDS-PAGE analysisTest membrane-mimetic solubilization conditionsShift of target protein from pellet to supernatant
MisfoldingHeme spectroscopic analysisCo-expression with other SDH subunitsAppearance of characteristic absorption peaks
ProteolysisTime-course Western blot during purificationAdd protease inhibitor cocktail and reduce temperatureStable full-length band throughout purification

For particularly challenging cases, consider developing a cell-free expression system using A. gossypii extract or a closely related fungal extract. While technically demanding to establish, such systems can provide a more native-like environment for proper folding and membrane insertion of SDH4 while allowing precise control over buffer components and redox conditions.

How can researchers address reproducibility challenges when measuring enzymatic activities of SDH complexes containing recombinant A. gossypii SDH4?

Ensuring reproducibility in enzymatic activity measurements of SDH complexes containing recombinant A. gossypii SDH4 requires systematic attention to multiple experimental variables:

  • Standardization of enzyme preparation:

    • Quantify protein concentration using multiple methods (Bradford, BCA, and amino acid analysis) to ensure accurate values

    • Verify complex integrity before each assay using native PAGE or gel filtration

    • Assess heme content spectrophotometrically and normalize activity to heme content rather than total protein

    • Prepare large, homogeneous enzyme batches that can be aliquoted and used across multiple experiments

  • Critical assay parameters standardization:

    • Control temperature rigorously (±0.1°C) throughout assays, as SDH activity typically increases 1.5-2 fold per 10°C

    • Maintain consistent pH using buffers with adequate capacity (25-50 mM) and minimal temperature dependence

    • Pre-equilibrate all reagents to assay temperature before initiating reactions

    • Standardize the order and timing of reagent addition

  • Substrate and cofactor considerations:

    • Use freshly prepared succinate solutions to avoid spontaneous decarboxylation

    • Ensure ubiquinone analogs are protected from light and stored under inert gas

    • Verify artificial electron acceptor quality (DCPIP, PMS) with standardized absorption spectra measurements

    • Include internal standards (commercial Complex II from bovine heart) in each assay series

  • Instrument-related variability management:

    • Perform regular spectrophotometer calibration with certified standards

    • Maintain consistent cuvette orientation for all measurements

    • Use the same spectrophotometer for all measurements within a study

    • Implement blank corrections for each new reagent preparation

  • Data analysis and reporting standardization:

    • Define linear range of assays and ensure measurements fall within this range

    • Report both specific activity (μmol/min/mg protein) and molecular activity (min-1)

    • Include detailed methods sections specifying all buffer components, concentrations, and preparation procedures

    • Share raw data in repositories for independent verification

A particularly effective approach to enhance reproducibility is to implement a multivariate experimental design that deliberately varies critical parameters (pH, temperature, substrate concentration) within defined ranges. This allows:

  • Identification of assay conditions where small variations have minimal impact on measured activities

  • Development of mathematical correction factors for variations that cannot be eliminated

  • Estimation of measurement uncertainty for each experimental condition

VariableRecommended Control StrategyAcceptable VariationImpact on Activity
TemperatureWater-jacketed cuvette holders±0.5°C~10% per degree
pH50 mM HEPES or phosphate buffer±0.1 pH unit~15% per pH unit
Protein concentrationBradford with BSA standard curve±5%Linear relationship
SuccinatePrepare fresh daily, store at 4°CUse within 8 hours~5% decrease per day
Ubiquinone analogsAmber vials, nitrogen atmosphereUse within 4 hours after opening~20% decrease per day
DCPIPPrepare fresh in DMSO, protect from lightVerify A600 before use~10% decrease per hour

By implementing these standardization practices and documenting all relevant experimental conditions, researchers can significantly improve both intra- and inter-laboratory reproducibility of SDH activity measurements with recombinant A. gossypii SDH4.

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.