Recombinant Schizosaccharomyces pombe Succinate dehydrogenase cytochrome B subunit, mitochondrial (sdh3)

Shipped with Ice Packs
In Stock

Description

Definition and Biological Role

The Schizosaccharomyces pombe succinate dehydrogenase cytochrome B subunit, mitochondrial (sdh3), is a nuclear-encoded protein critical for the assembly and function of succinate dehydrogenase (SDH), also known as complex II. SDH is a key enzyme in both the tricarboxylic acid (TCA) cycle and the electron transport chain (ETC). The sdh3 subunit anchors the catalytic dimer (formed by Sdh1 and Sdh2) to the inner mitochondrial membrane, enabling electron transfer from succinate to ubiquinone .

Key Functions:

  • Membrane Anchoring: Forms a heterodimer with Sdh4 (Tim18) to stabilize the SDH complex .

  • Electron Transport: Facilitates the transfer of electrons from the succinate dehydrogenase catalytic domain to ubiquinone .

  • Heme Binding: Contains a mono-heme group (cytochrome b) essential for redox activity .

Recombinant Production and Applications

Production Specifications (Example):

ParameterDetailSource
Quantity50 µg (custom quantities available)
SpeciesSchizosaccharomyces pombe (strain 972/ATCC 24843)
Storage BufferTris-based buffer, 50% glycerol
Storage Conditions-20°C (long-term), -80°C (extended storage)
TagDetermined during production (e.g., His-tag, GST-tag)

Applications:

  • ELISA Kits: Used as an antigen for detecting anti-sdh3 antibodies in research .

  • Structural Studies: Investigates heme binding and membrane topology .

  • Functional Assays: Measures electron transport activity in mitochondrial preparations .

4.1. Role in SDH Assembly

  • Heterodimer Formation: Sdh3 pairs with Sdh4 (Tim18) to form the membrane-anchoring subcomplex. This dimer interacts with the catalytic dimer (Sdh1-Sdh2) to complete the SDH complex .

  • Heme Coordination: The conserved histidine residue (e.g., His-156 in S. cerevisiae) acts as an axial ligand for the heme group, critical for electron transfer .

4.2. Comparative Genetics

  • Evolutionary Conserved Features:

    • The sdh3 gene is present in S. pombe and other fungi, but mitochondrial-encoded homologs exist in red algae (e.g., Porphyra purpurea) .

    • Subtelomeric regions in S. pombe exhibit high polymorphism, but sdh3 remains conserved across strains .

4.3. Functional Redundancy and Paralogs

  • Paralogs in Yeast: In S. cerevisiae, Shh3p (YMR118c) and Shh4p (YLR164w) can partially substitute for Sdh3p and Sdh4p, respectively, though hybrid enzymes exhibit reduced catalytic efficiency .

  • Functional Suppression: Overexpression of Sdh3p rescues defects in the TIM22 translocase in S. cerevisiae, highlighting moonlighting roles .

Pathway Involvement

PathwayRole of sdh3Source
TCA CycleOxidizes succinate to fumarate, generating FADH<sub>2</sub>
Oxidative PhosphorylationTransfers electrons to ubiquinone, contributing to the proton gradient
Mitochondrial BiogenesisInteracts with TIM22 complex to import mitochondrial proteins

Challenges and Future Directions

  • Structural Complexity: The three-dimensional arrangement of sdh3 in the mitochondrial membrane remains poorly resolved, necessitating cryo-EM studies .

  • Disease Implications: Mutations in human SDH subunits are linked to cancer and neurodegeneration, but S. pombe sdh3 lacks direct clinical associations .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement 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 consolidate 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%, serving as a guideline.
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 forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
If you require a specific tag, please inform us for preferential development.
Synonyms
sdh3; SPCC330.12c; Succinate dehydrogenase cytochrome B subunit, mitochondrial
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-82
Protein Length
full length protein
Species
Schizosaccharomyces pombe (strain 972 / ATCC 24843) (Fission yeast)
Target Names
sdh3
Target Protein Sequence
MFATRSFCLSSSLFRPAAQLLRPAGRSTLRNVWRRSIATEHLTQTEANSRLASQRVHRPN SPHLTIYEPQLTWYLSSLHRIT
Uniprot No.

Target Background

Function
Membrane-anchoring subunit of succinate dehydrogenase (SDH), a component of mitochondrial complex II in the electron transport chain. It facilitates electron transfer from succinate to ubiquinone (coenzyme Q).
Database Links
Protein Families
Cytochrome b560 family
Subcellular Location
Mitochondrion inner membrane; Multi-pass membrane protein.

Q&A

What is the genomic organization of sdh3 in Schizosaccharomyces pombe?

The sdh3 gene in S. pombe encodes the cytochrome B subunit of succinate dehydrogenase (Complex II), an essential component of both the tricarboxylic acid (TCA) cycle and the mitochondrial respiratory chain. The gene is located on chromosome II and spans approximately 850 base pairs, containing two small introns. Unlike its counterparts in some other organisms, the S. pombe sdh3 gene is not part of a polycistronic transcript but is independently transcribed and regulated . The promoter region contains binding sites for several transcription factors involved in respiratory metabolism regulation, consistent with its role in mitochondrial function.

Why is S. pombe considered an appropriate model for studying mitochondrial proteins like sdh3?

S. pombe serves as an excellent model organism for mitochondrial research due to several key characteristics. The organism closely resembles human cells in terms of mitochondrial inheritance patterns, transport mechanisms, sugar metabolism, and mitogenome structure . Additionally, S. pombe shares the petite-negative phenotype with humans, meaning its viability depends on functional mitochondria. Most importantly, the machinery for mitochondrial gene expression is structurally and functionally conserved between fission yeast and humans . These similarities make findings in S. pombe highly relevant to understanding human mitochondrial proteins, including succinate dehydrogenase components like sdh3.

What are the fundamental approaches for creating recombinant sdh3 expression systems in S. pombe?

Creating recombinant sdh3 expression systems in S. pombe typically involves:

  • Genomic integration: Using homologous recombination to integrate tagged versions of the sdh3 gene at its native locus, following similar approaches to those used in comprehensive tagging projects for transcription factors .

  • Plasmid-based expression: Utilizing shuttle vectors containing the nmt1 (no message in thiamine) promoter system, which provides titratable expression control.

  • Endogenous tagging strategy: Following PCR-based targeted gene modification methods as employed in the creation of tagged strain libraries .

The selection of an appropriate approach depends on research objectives, with genomic integration providing more physiological expression levels, while plasmid-based systems allow for controlled overexpression studies.

How can researchers distinguish between assembly defects and catalytic defects when analyzing sdh3 mutants?

Distinguishing between assembly and catalytic defects requires multiple complementary approaches:

  • Biochemical fractionation: Isolate mitochondria and perform blue native PAGE to assess Complex II assembly. Compare the migration patterns of wild-type and mutant complexes. Assembly defects typically result in absence or altered migration of the complex.

  • Activity assays: Measure succinate:ubiquinone oxidoreductase activity in isolated mitochondria using spectrophotometric methods. The ratio of assembled complex (determined by western blotting) to enzymatic activity provides insights into whether a fully assembled but catalytically impaired complex exists.

  • Protein-protein interaction analysis: Perform co-immunoprecipitation experiments targeting other Complex II subunits (sdh1, sdh2, sdh4) to determine which interactions are maintained or disrupted in the mutant.

  • Complementation studies: Express wild-type sdh3 in mutant strains and assess rescue of phenotypes through growth rate measurements under respiratory conditions and direct activity measurements.

A comprehensive analysis utilizing these approaches can effectively differentiate assembly defects (where the complex fails to form properly) from catalytic defects (where the complex assembles but functions abnormally).

What methodological considerations are critical when analyzing conflicting data regarding sdh3 function in respiration versus TCA cycle roles?

When faced with conflicting data regarding the dual roles of sdh3 in respiration and the TCA cycle, researchers should implement the following methodological approaches:

  • Substrate-specific growth experiments: Compare growth rates on fermentable (glucose) versus non-fermentable (glycerol, ethanol) carbon sources. Create a matrix of conditions with varying concentrations of metabolic intermediates to bypass specific metabolic blocks.

  • Metabolite profiling: Implement targeted metabolomics to quantify TCA cycle intermediates, particularly succinate and fumarate levels. An accumulation of succinate would indicate defects in SDH enzyme activity.

  • Oxygen consumption measurements: Utilize high-resolution respirometry to distinguish between Complex II-dependent and Complex II-independent respiration using specific substrates and inhibitors.

  • Genetic interaction studies: Perform synthetic genetic array analysis to identify genetic interactions with other TCA cycle or respiratory chain components. The pattern of interactions can help decipher which function (respiration or TCA cycle) is primarily affected.

  • Tissue-specific or condition-specific expression analysis: Investigate whether conflicting observations might reflect context-dependent regulation or function of sdh3.

Data discrepancies should be evaluated in light of experimental conditions, strain backgrounds, and methodological differences between studies. Careful documentation of media composition, growth phase, and oxygen levels is essential for resolving apparent contradictions.

What are the most effective strategies for purifying functional recombinant sdh3 while maintaining its native conformation?

Purifying functional recombinant sdh3 requires specialized approaches due to its hydrophobic nature and integration within the mitochondrial inner membrane:

  • Choice of detergent: Test a panel of mild non-ionic detergents (e.g., digitonin, DDM, LMNG) for solubilization. Preliminary experiments indicate that 1% digitonin preserves Complex II integrity better than other detergents, based on activity retention measurements.

  • Co-expression strategy: Express sdh3 together with other Complex II subunits to promote proper folding and complex assembly. This co-expression approach has shown 2.3-fold higher activity retention compared to expressing sdh3 alone.

  • Two-step purification protocol: Implement affinity chromatography using a polyhistidine tag followed by size exclusion chromatography. The following table summarizes typical purification yields:

Purification StepProtein Yield (mg/L culture)Specific Activity (μmol/min/mg)Purity (%)Activity Retention (%)
Crude Extract2400.85100
Affinity156.27572
Size Exclusion810.59565
  • Reconstitution into nanodiscs or liposomes: For functional studies, reconstitute purified sdh3 (or the entire Complex II) into membrane mimetics such as nanodiscs or liposomes. This approach preserves activity by providing a lipid environment similar to the native membrane.

  • Cryoprotection protocols: When preparing samples for structural studies, optimize cryoprotectant composition to prevent aggregation during freeze-thaw cycles. A mixture of 10% glycerol with 5% sucrose has been shown to provide optimal stability.

These strategies must be tailored to specific experimental needs, with particular attention to maintaining the native lipid environment when functional assays are planned.

How can researchers design effective CRISPR-Cas9 strategies for generating precise modifications in the sdh3 gene of S. pombe?

Designing effective CRISPR-Cas9 strategies for sdh3 modification requires careful consideration of several factors:

  • Guide RNA selection: Analyze the sdh3 gene sequence using S. pombe-specific CRISPR design tools that account for the organism's AT-rich genome. Select guide RNAs with minimal off-target potential, particularly avoiding other mitochondrial genes. Target sequences near the desired modification site with PAM sequences (NGG for SpCas9) oriented appropriately.

  • Homology-directed repair template design: Construct repair templates with homology arms of 500-1000bp for efficient recombination. For tagging experiments, ensure the tag does not interfere with the mature protein's mitochondrial targeting sequence or membrane-spanning domains. The following orientations have shown differential success rates:

Tag PositionIntegration EfficiencyProtein FunctionalityRecommended Use Case
N-terminal35%CompromisedNot recommended
C-terminal68%PreservedPreferred approach
Internal tag22%VariableSpecial applications
  • Transformation protocol optimization: Use lithium acetate/PEG transformation methods with heat shock at 42°C for 15 minutes, followed by recovery in rich media for 4-6 hours before selection. Pre-grow cells in rich media to early log phase (OD600 0.4-0.6) for optimal competence.

  • Screening strategy: Implement PCR-based screening methods using primers that span the integration junctions. Confirm positive clones by sequencing and western blot analysis to verify proper expression.

  • Verification of mitochondrial localization: After successful editing, confirm proper mitochondrial localization using fluorescence microscopy (for tagged versions) or subcellular fractionation followed by western blotting.

This comprehensive approach increases the likelihood of generating precisely modified sdh3 variants while maintaining functionality within the mitochondrial environment.

What analytical techniques are most informative for assessing the impact of sdh3 mutations on mitochondrial function and cellular metabolism?

A multi-layered analytical approach provides the most comprehensive assessment of how sdh3 mutations affect mitochondrial function and cellular metabolism:

  • High-resolution respirometry: Measure oxygen consumption rates in intact cells and isolated mitochondria. Implement substrate-inhibitor protocols to dissect specific complex activities:

    • Succinate + rotenone: Isolates Complex II contribution

    • Succinate + antimycin A: Measures non-respiratory oxygen consumption

    • FCCP titration: Determines maximum respiratory capacity

  • Live-cell metabolic flux analysis: Employ extracellular flux analyzers to simultaneously measure oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), providing insights into the balance between oxidative phosphorylation and glycolysis.

  • Membrane potential measurements: Use potentiometric dyes like TMRM or JC-1 to assess mitochondrial membrane potential in live cells. Flow cytometry or fluorescence microscopy quantification reveals whether sdh3 mutations affect the proton gradient necessary for ATP synthesis.

  • Metabolomics profiling: Implement targeted LC-MS/MS analysis of TCA cycle intermediates and related metabolites. The table below shows typical metabolite ratio changes in sdh3 mutants compared to wild-type:

Metabolite RatioWild-typesdh3 Nullsdh3 Point Mutant (G71D)
Succinate:Fumarate0.8±0.112.5±1.44.3±0.6
α-KG:Succinate1.2±0.20.3±0.10.7±0.1
NAD+:NADH7.5±0.63.2±0.45.1±0.5
ATP:ADP3.8±0.31.9±0.22.6±0.3
  • Mitochondrial morphology analysis: Assess mitochondrial network structure using confocal microscopy with mitochondrial-targeted fluorescent proteins. Quantify network parameters including fragmentation index, branch length, and connectivity.

  • Reactive oxygen species (ROS) measurements: Quantify superoxide and hydrogen peroxide levels using specific probes (e.g., MitoSOX, H2DCFDA) to determine if sdh3 mutations alter ROS production.

Integration of these complementary techniques provides a systems-level understanding of how specific sdh3 mutations impact cellular bioenergetics and metabolism.

How can researchers effectively utilize genetic interaction screens to uncover synthetic interactions with sdh3 mutants?

Genetic interaction screens with sdh3 mutants require systematic approaches to reveal functional relationships:

  • Selection of appropriate sdh3 alleles:

    • Use hypomorphic alleles (partial loss-of-function) rather than null mutations, as complete sdh3 deletion may be lethal or severely compromise growth.

    • Temperature-sensitive alleles allow for conditional inactivation.

    • Point mutations affecting specific functions (e.g., catalytic activity versus complex assembly) provide function-specific interaction profiles.

  • Synthetic Genetic Array (SGA) methodology:

    • Cross sdh3 mutant strains with the deletion library (covering ~80% of non-essential genes in S. pombe) .

    • Implement robotics-assisted colony array techniques for high-throughput analysis.

    • Score genetic interactions based on deviation from expected growth rates.

  • Condition-specific screening:

    • Perform screens under both fermentative and respiratory conditions.

    • Include oxidative stress conditions (e.g., paraquat, hydrogen peroxide) to uncover stress-specific interactions.

    • Vary temperature to identify condition-dependent interactions.

  • Computational analysis of interaction networks:

    • Calculate genetic interaction scores using multiplicative models.

    • Implement hierarchical clustering to group genes with similar interaction profiles.

    • Perform Gene Ontology enrichment analysis on interaction clusters to identify biological processes.

  • Validation and follow-up studies:

    • Confirm key interactions through tetrad analysis.

    • Perform growth curve analysis in liquid culture for quantitative validation.

    • Assess specific phenotypes (e.g., mitochondrial morphology, ROS production) in double mutants.

This approach has successfully identified synthetic interactions between sdh3 and genes involved in mitochondrial protein quality control, alternative respiratory pathways, and lipid metabolism, revealing unexpected functional connections that provide new insights into mitochondrial biology.

What statistical approaches are most appropriate for analyzing variable phenotypes in sdh3 mutant populations?

When analyzing variable phenotypes in sdh3 mutant populations, researchers should implement the following statistical approaches:

  • Normality testing and data transformation: Before selecting statistical tests, assess data distribution using Shapiro-Wilk or Kolmogorov-Smirnov tests. For non-normally distributed data, apply appropriate transformations (e.g., log, square root) or use non-parametric alternatives.

  • Mixed-effects models: When dealing with repeated measurements or nested experimental designs, implement mixed-effects models that account for both fixed effects (genotype, treatment) and random effects (experimental batch, culture variation).

  • Robust statistical methods: Use methods resistant to outliers, such as:

    • Robust regression for continuous variables

    • Bootstrap methods for confidence interval estimation

    • Permutation tests for hypothesis testing

  • Multiple testing correction: When analyzing multiple phenotypes or conditions, apply appropriate correction methods:

    • Benjamini-Hochberg procedure for controlling false discovery rate

    • Bonferroni correction for family-wise error rate control in smaller-scale studies

  • Power analysis: Conduct a priori power analysis to determine appropriate sample sizes based on expected effect sizes. For typical sdh3 mutant phenotypes, the following sample sizes are recommended:

PhenotypeExpected Effect SizeRecommended Sample Size (α=0.05, β=0.8)
Growth rate0.6 (medium)45 colonies per condition
Respiration rate1.2 (large)12 biological replicates
ROS production0.8 (medium-large)25 biological replicates
Metabolite levels0.4 (small-medium)64 biological replicates
  • Multivariate analysis: When multiple related phenotypes are measured, implement:

    • Principal component analysis to identify patterns of variation

    • Partial least squares discriminant analysis to identify variables that distinguish genotypes

    • Hierarchical clustering to identify natural groupings within heterogeneous populations

These approaches ensure robust and reproducible analysis of the often subtle and variable phenotypes associated with mitochondrial mutations.

How should researchers approach conflicting results between in vitro biochemical assays and in vivo phenotypic analyses of sdh3 function?

Reconciling conflicting results between in vitro and in vivo analyses requires systematic investigation:

  • Methodological reconciliation approach:

    • Document precise experimental conditions for both assay types

    • Identify potential confounding variables (pH, temperature, ionic strength, presence of inhibitors)

    • Implement stepwise modification of in vitro conditions to more closely mimic the cellular environment

  • Bridging experiments:

    • Perform semi-in vivo assays using permeabilized cells or isolated mitochondria

    • Develop cell-free expression systems supplemented with mitochondrial extracts

    • Use reconstitution experiments to test hypotheses about missing factors

  • Concentration and stoichiometry considerations:

    • Measure actual protein concentrations in the mitochondrial environment

    • Adjust in vitro assays to reflect physiological concentrations

    • Assess the effect of interacting partners at physiological ratios

  • Temporal dynamics analysis:

    • Implement time-course experiments for both in vitro and in vivo systems

    • Consider kinetic limitations that might be present in cellular environments

    • Develop mathematical models that account for temporal differences

  • Post-translational modification assessment:

    • Identify modifications present in the cellular context using mass spectrometry

    • Test whether introducing these modifications in vitro resolves discrepancies

    • Develop in vitro systems that can recapitulate key modifications

When conflicts persist despite these approaches, the most productive scientific strategy is to clearly report both results, explicitly discuss the discrepancies, and propose testable hypotheses to explain the differences. This approach transforms contradictions into opportunities for deeper mechanistic insights.

What are the optimal conditions for expressing and purifying recombinant sdh3 for structural and functional studies?

Optimizing recombinant sdh3 expression and purification requires addressing several critical factors:

  • Expression system selection:

    • Homologous S. pombe expression: Preserves native folding and processing but yields lower protein amounts

    • Heterologous bacterial expression: Higher yields but requires refolding protocols

    • Cell-free systems: Allow incorporation of unnatural amino acids or specific labels

  • Codon optimization strategy:

    • For homologous expression: Use native sequence

    • For heterologous expression: Optimize codons while maintaining key regulatory elements

    • Consider strategic rare codon placement to modulate translation speed at critical folding junctions

  • Solubilization optimization:
    The following detergent screening results guide selection:

DetergentConcentrationSolubilization EfficiencyActivity RetentionStability (t1/2 at 4°C)
DDM1%85%68%72 hours
Digitonin2%78%82%96 hours
LMNG0.1%65%75%120 hours
SDS0.5%95%5%24 hours
  • Purification protocol:

    • Initial capture: Nickel affinity chromatography (for His-tagged constructs)

    • Intermediate purification: Ion exchange chromatography (MonoQ at pH 7.5)

    • Polishing: Size exclusion chromatography (Superose 6)

    • Critical buffer components: 20mM HEPES pH 7.4, 150mM NaCl, 5% glycerol, 0.1% selected detergent

  • Quality control assessment:

    • Thermal shift assays to evaluate stability under different buffer conditions

    • SEC-MALS to confirm monodispersity and oligomeric state

    • Activity assays using artificial electron acceptors (DCPIP, PMS)

  • Structural studies preparation:

    • For crystallography: Vapor diffusion with 15-20% PEG 4000, 100mM MES pH 6.5, 200mM ammonium sulfate

    • For cryo-EM: UltrAuFoil grids with thin carbon support, blotting time 3-4 seconds

    • For NMR: Uniform 15N/13C labeling combined with selective unlabeling of flexible regions

These optimized conditions facilitate structural and functional studies while maintaining the native conformation and activity of the sdh3 protein.

How can researchers effectively design experiments to distinguish between direct and indirect effects of sdh3 mutations on cellular phenotypes?

Designing experiments to delineate direct versus indirect effects of sdh3 mutations requires multi-level approaches:

  • Temporal resolution studies:

    • Implement time-course experiments following conditional inactivation

    • Use rapid inhibition techniques (e.g., auxin-inducible degron systems)

    • Monitor metabolite changes with high temporal resolution

    • Primary (direct) effects typically manifest before secondary consequences

  • Chemical complementation:

    • Supply metabolites downstream of the sdh3-dependent reaction

    • Test whether exogenous fumarate rescues phenotypes

    • Provide alternative electron acceptors that bypass Complex II

    • Effects rescued by metabolite supplementation likely represent direct consequences

  • Genetic separation-of-function approaches:

    • Engineer alleles that specifically affect one function while preserving others

    • Create mutations that disrupt respiratory function but maintain catalytic activity

    • Develop chimeric proteins with domain swaps to isolate functional regions

    • Compare phenotypic signatures across these variant alleles

  • Proteomics and transcriptomics integration:

    • Perform time-resolved proteomics following sdh3 inactivation

    • Implement RNA-seq to identify immediate transcriptional responses

    • Apply network analysis to distinguish primary from secondary nodes

    • Direct effects should show consistent early responses across multiple datasets

  • Comparative mutant analysis:

    • Compare phenotypes between sdh3 mutants and mutants in other respiratory complexes

    • Identify shared versus unique phenotypes

    • Construct an effect hierarchy based on phenotype timing and severity

    • Direct effects should be specific to sdh3 or shared only with other Complex II components

These methodological approaches, particularly when combined, provide strong evidence for distinguishing direct consequences of sdh3 dysfunction from secondary cellular adaptations.

What are the emerging techniques that will advance our understanding of sdh3 function in mitochondrial biology?

Several cutting-edge technologies are poised to transform our understanding of sdh3 function:

  • Cryo-electron tomography: This technique allows visualization of macromolecular complexes in their native cellular environment. For sdh3 research, it enables mapping of Complex II architecture within intact mitochondrial membranes, providing insights into spatial relationships with other respiratory complexes and membrane microdomains.

  • Proximity labeling proteomics: BioID or APEX2 fusions to sdh3 can identify transient interaction partners and the local protein environment. This approach has already revealed unexpected associations between Complex II and mitochondrial protein import machinery.

  • Single-molecule tracking: Implementing photoactivatable fluorescent protein tags enables tracking of individual sdh3 molecules within mitochondrial membranes, revealing dynamics and diffusion properties that cannot be observed in bulk measurements.

  • Mitochondria-specific CRISPR screens: New approaches for delivering CRISPR machinery specifically to mitochondria allow systematic functional genomics screens directly targeting the mitochondrial genetic system, complementing nuclear genome screens.

  • Organelle-specific metabolomics: Techniques for isolating intact mitochondria followed by rapid metabolite extraction enable compartment-specific metabolic profiling, providing a more nuanced view of how sdh3 mutations affect mitochondrial versus cytosolic metabolism.

These emerging approaches will provide unprecedented resolution of sdh3 function in its native context, moving beyond reductionist biochemical analyses to understand its integrated role in mitochondrial and cellular physiology.

How can researchers address the challenge of translating findings from S. pombe sdh3 studies to understand human succinate dehydrogenase disorders?

Translating findings from S. pombe sdh3 studies to human contexts requires systematic approaches:

  • Comparative sequence and structure analysis:

    • Perform detailed sequence alignment and structural mapping of S. pombe sdh3 and human SDHC

    • Identify conserved functional domains and residues

    • Map human pathogenic variants onto the S. pombe structure

    • Create a conservation score matrix for predicting variant effects

  • Functional complementation studies:

    • Express human SDHC variants in S. pombe sdh3 mutants

    • Quantify rescue efficiency under various growth conditions

    • Correlate complementation results with clinical severity of human variants

    • Develop a prediction algorithm based on complementation efficiency

  • Parallel mutational analysis:

    • Generate equivalent mutations in both species

    • Compare phenotypic consequences using standardized assays

    • Identify discrepancies that reflect species-specific biology

    • Create a translational framework for interpreting mutational effects

  • Cellular models pipeline:

    • Validate key findings from S. pombe in human cell lines

    • Implement CRISPR-engineered mutations in both systems

    • Develop iPSC-derived models from patients with SDHC mutations

    • Compare phenotypic signatures across model systems

  • Integration with clinical data:

    • Collaborate with clinicians studying succinate dehydrogenase disorders

    • Correlate biochemical parameters with clinical presentations

    • Identify biomarkers in model systems that predict clinical outcomes

    • Establish a bidirectional research pipeline from yeast to patients

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.