The recombinant mt-co2 protein from L. oculatus spans 72 amino acids (aa), with the following N-terminal sequence:
MAHPSQLGFQDAASPVMEELLHFHDHALMIVFLISTLVLYIIVAMVSTKLTNKHILDSQE VEIVWTILPAVI
. While the full-length human mt-co2 comprises 227 aa, the spotted gar variant is truncated, likely reflecting evolutionary divergence.
The protein is classified as a transmembrane protein with a molecular weight of ~19 kDa (estimated from sequence length) .
Tag: N-terminal 10xHis-tag for nickel affinity chromatography .
Purity: Likely >95% based on standard recombinant protein protocols, though specific purity data is not provided.
Parameter | Specification | Source |
---|---|---|
Product Code | CSB-CF015073LEE | |
Storage Buffer | Tris-based buffer with 50% glycerol | |
Storage | -20°C (liquid), -80°C (long-term) |
The protein is used as an antigen in ELISA kits for detecting anti-mt-co2 antibodies, enabling studies on mitochondrial dysfunction in diseases like cardiovascular disorders or cerebellar ataxia .
Condition | Recommendation | Source |
---|---|---|
Freeze-Thaw Cycles | Avoid repeated cycles to prevent denaturation | |
Working Aliquots | Store at 4°C for ≤1 week |
The protein’s shelf life is 6 months (liquid) or 12 months (lyophilized) at -20°C/-80°C .
Feature | L. oculatus mt-co2 | Human mt-co2 |
---|---|---|
Gene Origin | Mitochondrial DNA (mt-co2) | Mitochondrial DNA (MT-CO2) |
Sequence Length | 72 aa (truncated) | 227 aa (full-length) |
Copper Sites | CuA center conserved | CuA and CuB (in subunit 1) |
Function | Electron transfer to subunit 1 | Oxygen reduction to water |
While the spotted gar variant lacks the full-length complexity of human mt-co2, its conserved CuA center retains functional relevance for electron transport studies .
Structural Elucidation: High-resolution crystallography data for L. oculatus mt-co2 remain absent, limiting insights into its 3D conformation.
Functional Assays: No reported studies on recombinant mt-co2 activity in reconstituted lipid bilayers or mitochondrial models.
Cross-Species Relevance: Potential utility in studying ancestral mt-co2 evolution in ancient fish lineages.
Cytochrome c Oxidase Subunit 2 (mt-CO2) in Lepisosteus oculatus functions as a critical component of the mitochondrial respiratory chain, specifically in Complex IV. This subunit contains copper centers that facilitate electron transfer from cytochrome c to molecular oxygen in the final step of the electron transport chain . In gar fish, this protein plays an essential role in cellular respiration and energy production through oxidative phosphorylation. The specific properties of gar mt-CO2 make it valuable for comparative studies of respiratory chain evolution across fish species, particularly when examining the divergence of ancient fish lineages such as the Lepisosteiformes from teleosts .
Lepisosteus oculatus mt-CO2 exhibits structural features that reflect its evolutionary position as a non-teleost ray-finned fish. The protein contains characteristic copper-binding domains similar to those found in other vertebrates, but with distinct sequence variations reflective of its evolutionary history. When comparing gar mt-CO2 to teleost fishes like zebrafish (Danio rerio) or more distantly related vertebrates, researchers can observe conserved functional domains alongside lineage-specific adaptations . These structural differences can be visualized through protein threading analysis similar to methods used for other membrane proteins, where surface-exposed residues often display greater variability than those involved in core catalytic functions or interactions with other subunits .
For the expression of recombinant Lepisosteus oculatus mt-CO2, researchers typically employ prokaryotic systems like E. coli or eukaryotic systems depending on research requirements. E. coli systems (similar to those used for other recombinant proteins) utilize vectors containing appropriate promoters and purification tags . The expressed protein is typically formulated in a buffer system containing stabilizing agents such as glycerol and reducing agents like DTT to maintain protein integrity . Eukaryotic expression systems may be necessary when post-translational modifications are required for functionality studies. A typical expression protocol would include gene optimization for the chosen expression system, vector construction with appropriate tags (His-tag being common), transformation, culture optimization, and purification through affinity chromatography .
Recombinant L. oculatus mt-CO2 requires specific storage conditions to maintain stability and functionality. Based on protocols for similar recombinant proteins, the purified protein is typically stored at -20°C in a buffer solution containing stabilizing agents . A recommended formulation would include 20mM Tris-HCl buffer (pH 8.0), 0.1M NaCl, 10% glycerol, and 2mM DTT to maintain protein integrity and prevent oxidation of sensitive residues . For longer-term storage, aliquoting the protein to minimize freeze-thaw cycles is advisable. When handling the protein, it should be kept on ice and transported using blue ice to maintain temperature stability . Stability testing under various conditions is an important preliminary step for any research protocol involving this recombinant protein.
Measuring the functional interaction between recombinant L. oculatus mt-CO2 and cytochrome c requires specialized biochemical assays that assess electron transfer kinetics. A robust methodological approach involves spectrophotometric assays monitoring the oxidation rate of reduced cytochrome c at 550 nm, which directly correlates with mt-CO2 activity. Researchers should establish a baseline using purified components in a controlled buffer system (typically 50 mM phosphate buffer, pH 7.4) with precise protein concentrations determined through Bradford or BCA assays .
For kinetic analysis, the following protocol is recommended:
Prepare reduced cytochrome c by adding sodium dithionite and removing excess reductant via gel filtration
Add recombinant mt-CO2 at varying concentrations (0.1-10 μg/ml)
Monitor absorbance changes at 550 nm over time at physiologically relevant temperatures (20-25°C for gar-based studies)
Calculate initial reaction rates and derive kinetic parameters (Km, Vmax) through Lineweaver-Burk or Eadie-Hofstee plots
This methodology allows researchers to compare the functional characteristics of gar mt-CO2 with those of other species, providing insights into evolutionary adaptations of the respiratory chain in different vertebrate lineages .
Expression of recombinant L. oculatus mt-CO2 presents challenges due to its hydrophobic regions and potential toxicity to host cells. To overcome these challenges, researchers should employ a multi-faceted approach:
Strategy | Implementation | Expected Outcome |
---|---|---|
Codon optimization | Adjust codons to match host system preferences | Improved translation efficiency |
Fusion tags | N-terminal solubility enhancers (SUMO, MBP, or GST) | Increased solubility and reduced toxicity |
Expression temperature | Reduce to 16-18°C during induction | Slower expression promoting proper folding |
Specialized host strains | C41(DE3) or C43(DE3) designed for membrane proteins | Better tolerance of potentially toxic proteins |
Detergent screening | Test multiple detergents (DDM, LDAO, Triton X-100) | Optimal membrane protein solubilization |
For particularly difficult constructs, a cell-free expression system may be employed, which allows for the direct incorporation of the protein into nanodiscs or liposomes . When conventional approaches fail, researchers might consider expressing individual domains separately or creating chimeric constructs with better-expressing homologs from related species while maintaining the key functional regions of interest.
Molecular dynamics (MD) simulations offer powerful insights into the evolutionary adaptations of L. oculatus mt-CO2 by revealing atomic-level behavior under different conditions. To implement this approach effectively, researchers should:
Begin by creating a high-quality structural model through homology modeling based on crystallographic structures of cytochrome c oxidase from other species, using threading methods to identify key structural features .
Embed the protein model in a simulated lipid bilayer that mimics the mitochondrial inner membrane, with appropriate phospholipid composition.
Perform equilibrium MD simulations (typically 100-500 ns) under physiologically relevant conditions, comparing simulations at different temperatures (10-30°C) to examine temperature adaptations specific to the gar's environment.
Analyze specific parameters including:
Proton transfer pathway dynamics
Conformational flexibility of key residues
Water molecule organization within channels
Interaction energies with other subunits
Compare simulation results with those from other species to identify unique features that may represent evolutionary adaptations .
This computational approach complements experimental data by providing mechanistic hypotheses that can be subsequently tested through site-directed mutagenesis or biochemical assays, creating a powerful iterative research approach.
Studying the assembly of L. oculatus mt-CO2 into functional respiratory complexes requires specialized techniques that can track protein-protein interactions and complex formation. A comprehensive methodological approach includes:
Co-immunoprecipitation studies: Using antibodies against mt-CO2 or epitope tags to pull down interaction partners, followed by mass spectrometry identification of the components. This approach can identify both stable and transient interactions during complex assembly .
Blue Native PAGE analysis: This technique separates intact respiratory complexes while preserving their native interactions. By incorporating a first-dimension BN-PAGE with a second-dimension SDS-PAGE, researchers can visualize both the intact complexes and their subunit composition .
Pulse-chase experiments: These can track the incorporation of newly synthesized mt-CO2 into assembling complexes over time, providing insights into the assembly kinetics and sequencing.
Fluorescence microscopy with split-GFP constructs: By tagging mt-CO2 and other subunits with complementary GFP fragments, researchers can visualize complex assembly in living cells through the reconstitution of fluorescent GFP when the proteins interact.
Crosslinking mass spectrometry (XL-MS): This advanced technique uses chemical crosslinkers to capture protein-protein interactions, followed by mass spectrometry to identify interaction interfaces at amino acid resolution .
These approaches should be conducted in systems that closely mimic the native environment, such as isolated mitochondria or suitable cell culture models expressing the gar proteins.
Lepisosteus oculatus (spotted gar) occupies a unique evolutionary position as a non-teleost ray-finned fish that diverged from the teleost lineage before the teleost-specific genome duplication. This makes recombinant L. oculatus mt-CO2 an invaluable model for understanding respiratory chain evolution through comparative analysis . Researchers can leverage this model by:
Conducting phylogenetic analysis of mt-CO2 sequences across vertebrate lineages to identify conserved regions versus lineage-specific adaptations.
Performing enzymatic activity comparisons under varying conditions (temperature, pH, salt concentration) to correlate sequence differences with functional adaptations.
Using site-directed mutagenesis to introduce teleost-specific or tetrapod-specific residues into the gar protein and assess their impact on function.
Examining the co-evolution of nuclear-encoded and mitochondrial-encoded subunits of the cytochrome c oxidase complex, which can reveal constraints on the evolution of the respiratory chain .
This evolutionary perspective is particularly valuable for understanding how environmental adaptations have shaped mitochondrial function across vertebrate lineages, with the gar representing an important reference point between teleosts and tetrapods.
Purifying active recombinant L. oculatus mt-CO2 presents several challenges due to its hydrophobic nature and requirement for proper cofactor incorporation. These challenges and their solutions include:
Challenge | Solution | Implementation Details |
---|---|---|
Membrane protein solubility | Optimized detergent screening | Test panel of detergents (DDM, LDAO, Fos-choline) at varying concentrations (0.5-2% for extraction, 0.05-0.5% for purification) |
Copper incorporation | Supplementation strategy | Add CuSO₄ (10-50 μM) during expression or reconstitute during purification |
Protein aggregation | Buffer optimization | Include glycerol (10-20%), reducing agents (2-5 mM DTT or β-mercaptoethanol), and appropriate salt concentration (100-300 mM NaCl) |
Maintaining native conformation | Lipid supplementation | Add specific phospholipids (0.1-0.5 mg/mL) found in mitochondrial membranes during purification |
Low expression yields | Expression system selection | Compare prokaryotic (E. coli) vs. eukaryotic (insect cells) systems for optimal expression |
Additionally, researchers should consider implementing a multi-step purification strategy including initial IMAC (immobilized metal affinity chromatography) using the His-tag, followed by ion-exchange and size-exclusion chromatography to separate properly folded, active protein from aggregates or misfolded species . Activity assays should be performed at each purification step to track specific activity and recovery of functional protein.
Differential scanning fluorimetry (DSF) is a powerful technique for optimizing stability conditions for recombinant L. oculatus mt-CO2. This approach involves using environment-sensitive fluorescent dyes that bind to hydrophobic regions exposed during protein unfolding, allowing researchers to determine melting temperatures (Tm) under various conditions. A methodological approach includes:
Prepare purified recombinant mt-CO2 at concentrations of 0.1-0.5 mg/mL in various test buffers.
Add SYPRO Orange or similar fluorescent dye at appropriate dilutions (typically 5X final concentration).
Perform thermal ramping (25-95°C) while monitoring fluorescence emission in a real-time PCR instrument or specialized thermal shift analyzer.
Analyze melting curves to determine Tm values under each condition.
Systematically test multiple parameters:
Buffer composition (HEPES, Tris, phosphate, MES) at pH range 6.0-8.5
Salt type and concentration (NaCl, KCl at 0-500 mM)
Additives (glycerol 5-20%, reducing agents 1-10 mM)
Detergents (type and concentration)
Metal ions (particularly copper at 1-100 μM)
This approach can generate a stability landscape for the protein, identifying optimal conditions that significantly extend shelf-life and maintain functional activity . The resulting optimized buffer system can then be validated through long-term activity assays to confirm that thermal stability correlates with functional longevity.
Measuring interactions between L. oculatus mt-CO2 and potential inhibitors requires sensitive biophysical techniques that can detect binding events and their functional consequences. An effective methodological approach includes:
Surface Plasmon Resonance (SPR):
Immobilize purified mt-CO2 on a sensor chip via His-tag or biotinylation
Flow potential inhibitors at varying concentrations (1 nM to 100 μM)
Measure association and dissociation kinetics
Calculate binding constants (KD, kon, koff)
Isothermal Titration Calorimetry (ITC):
Directly measure thermodynamic parameters of binding
Determine binding stoichiometry, enthalpy (ΔH), and entropy (ΔS)
Distinguish between enthalpy-driven and entropy-driven interactions
Microscale Thermophoresis (MST):
Label protein with fluorescent dye or use intrinsic tryptophan fluorescence
Measure changes in thermophoretic mobility upon inhibitor binding
Requires minimal protein amounts (typically <100 nM)
Functional inhibition assays:
Measure cytochrome c oxidation rates in presence of varying inhibitor concentrations
Determine IC50 values and inhibition mechanisms (competitive, non-competitive)
Correlate binding data with functional impact
Thermal shift assays:
Measure changes in protein thermal stability upon inhibitor binding
Quick screening method for multiple compounds
Can indicate binding mode through stabilization or destabilization effects
These techniques provide complementary information about inhibitor interactions, from pure binding parameters to functional consequences, allowing researchers to develop comprehensive models of inhibitor action .
Ensuring the quality of recombinant L. oculatus mt-CO2 preparations requires multiple complementary assessment techniques. An effective quality control protocol should include:
Purity assessment:
Identity confirmation:
Mass spectrometry (LC-MS/MS) peptide mapping against the theoretical sequence
N-terminal sequencing to confirm proper processing
Immunological detection with subunit-specific antibodies
Structural integrity:
Circular dichroism spectroscopy to confirm secondary structure elements
Fluorescence spectroscopy to assess tertiary folding
Limited proteolysis to verify proper folding (correctly folded proteins show distinct, reproducible digestion patterns)
Functional activity:
Cytochrome c oxidation assays with standardized conditions
Oxygen consumption measurements
Spectroscopic analysis of metal centers (copper absorption features)
Homogeneity assessment:
Dynamic light scattering to evaluate size distribution
Analytical ultracentrifugation to detect multiple oligomeric states
Blue native PAGE to assess complex assembly
Each batch of purified protein should be evaluated against established specifications, with documented acceptance criteria for each parameter. This comprehensive approach ensures that experimental results can be reliably attributed to the protein's intrinsic properties rather than preparation artifacts .
Low expression yields are a common challenge when producing recombinant L. oculatus mt-CO2. Researchers can implement several strategies to overcome this limitation:
Vector optimization:
Test multiple promoter systems (T7, tac, araBAD) to find optimal expression levels
Incorporate enhancer elements or optimize the ribosome binding site
Consider using vectors with tight regulation to minimize toxicity during culture growth
Fusion partner screening:
Evaluate different fusion tags beyond standard His-tags (MBP, GST, SUMO, Trx)
Position tags at either N- or C-terminus to determine optimal configuration
Include precision protease sites for tag removal that don't leave residual amino acids
Culture optimization:
Test expression in specialized media formulations (e.g., Terrific Broth, Auto-induction media)
Optimize induction parameters (inducer concentration, temperature, duration)
Implement fed-batch cultivation to achieve higher cell densities before induction
Co-expression strategies:
Co-express with chaperones (GroEL/ES, DnaK/J) to assist proper folding
Include key interacting partners that might stabilize the protein
Co-express with enzymes required for cofactor synthesis or incorporation
Alternative expression systems:
Evaluate eukaryotic systems (yeast, insect cells) if prokaryotic expression fails
Consider cell-free expression systems for toxic proteins
Test expression in mitochondria-like bacterial systems (Paracoccus denitrificans)
By systematically exploring these options and documenting the impact of each modification, researchers can develop an optimized production protocol that significantly improves yields of functional protein .
Studying post-translational modifications (PTMs) of L. oculatus mt-CO2 requires specialized techniques to identify, locate, and functionally characterize these modifications. An effective methodological approach includes:
PTM identification and mapping:
High-resolution mass spectrometry (MS) with multiple fragmentation techniques (CID, ETD, HCD)
Enrichment strategies for specific modifications (phosphopeptide enrichment, glycopeptide capture)
Site-specific antibodies for common PTMs (phosphorylation, acetylation)
Targeted multiple reaction monitoring (MRM) MS for quantitative analysis of specific modified sites
Temporal dynamics of modifications:
Pulse-chase experiments with metabolic labeling
Time-course analysis following cellular stimulation
In vitro modification using purified modifying enzymes
Functional significance assessment:
Site-directed mutagenesis of modified residues (mimicking modifications or preventing them)
Activity assays comparing modified and unmodified forms
Structural analysis to determine how modifications affect protein conformation
Modification enzymes identification:
Co-immunoprecipitation coupled with proteomic analysis
Activity-based protein profiling for enzyme identification
Inhibitor studies to link specific enzymes to observed modifications
Comparative analysis across species:
This comprehensive approach provides insights into the regulatory mechanisms controlling mt-CO2 function through post-translational modifications, potentially revealing unique aspects of respiratory chain regulation in non-teleost fishes like Lepisosteus oculatus.
Developing specific antibodies against L. oculatus mt-CO2 requires careful antigen design and validation strategies. A comprehensive approach includes:
Antigen design strategy:
Identify unique, surface-exposed peptide regions through sequence analysis and structural prediction
Focus on regions with low sequence conservation across species for species-specific antibodies
Target conserved regions for pan-specific antibodies that work across multiple species
Consider both full-length protein and synthetic peptide approaches
Production methods:
For polyclonal antibodies: Immunize rabbits or other host animals with purified recombinant protein or KLH-conjugated peptides
For monoclonal antibodies: Screen hybridoma libraries against the target antigen
For recombinant antibodies: Perform phage display selection against the target protein
Purification approach:
Implement affinity purification using antigen-conjugated columns
Perform negative selection against homologous proteins from other species to enhance specificity
Characterize antibody isotypes and subclasses for optimal application performance
Validation protocol:
Western blotting against recombinant protein and native tissue extracts
Immunoprecipitation followed by mass spectrometry confirmation
Immunohistochemistry with appropriate controls (pre-immune serum, blocking peptides)
Cross-reactivity testing against homologous proteins from related species
Application optimization:
Determine optimal working dilutions for each application (Western blot, immunoprecipitation, immunohistochemistry)
Establish appropriate buffer conditions and blocking agents
Document lot-to-lot variability and establish validation criteria for each batch
This methodical approach ensures the development of reliable antibody tools that can be used for multiple research applications, from protein localization to interaction studies and functional analyses of L. oculatus mt-CO2 .
CRISPR/Cas9 genome editing offers powerful approaches for studying L. oculatus mt-CO2 function in vivo, despite the challenges of editing mitochondrial genes. Researchers can implement several strategies:
Nuclear-encoded reporter systems:
Create reporter constructs that respond to mt-CO2 activity or assembly
Edit nuclear genes encoding mt-CO2 interacting partners to study their effects on function
Develop inducible systems to modulate expression of assembly factors
Allotopic expression models:
Study design approaches:
Implement tissue-specific or temporally controlled gene editing using conditional Cas9 systems
Create knock-in models with fluorescent or affinity tags to track protein localization and interactions
Generate heterozygous models to study gene dosage effects on respiratory function
Phenotypic analysis methods:
Measure oxygen consumption in isolated mitochondria from edited cells
Assess metabolic shifts using stable isotope labeling
Evaluate reactive oxygen species production and mitochondrial membrane potential
Examine cellular and organismal phenotypes under normal and stressed conditions
Comparative editing strategies:
These approaches circumvent the technical challenges of directly editing mitochondrial DNA while still providing valuable insights into mt-CO2 function, assembly, and regulation in the context of living cells and organisms.
Lepisosteus oculatus mt-CO2 offers unique advantages as a model for studying mitochondrial disorders due to its evolutionary position. Potential research applications include:
Comparative pathogenicity assessment:
Drug screening platforms:
Develop assay systems using recombinant gar mt-CO2 to screen compounds that rescue mutant phenotypes
Compare drug efficacy across species-specific versions of mt-CO2
Identify compounds that specifically modulate cytochrome c oxidase activity or assembly
Evolution-guided therapy development:
Identify naturally occurring variations in gar mt-CO2 that confer resistance to stressors
Study the molecular basis of these protective mechanisms
Apply these insights to develop therapeutic approaches for human disorders
Mitochondrial assembly model:
Use the gar system to study conserved and divergent aspects of Complex IV assembly
Identify species-specific assembly factors and their potential therapeutic relevance
Develop interventions that promote proper assembly of defective oxidase complexes
Environmental susceptibility research:
These applications leverage the unique evolutionary features of gar mt-CO2 to provide insights into fundamental mechanisms of mitochondrial disease and potential therapeutic approaches that might not be apparent from studying mammalian systems alone.
Determining the high-resolution structure of L. oculatus mt-CO2 requires optimized structural biology approaches tailored to this challenging membrane protein. A comprehensive methodological strategy includes:
Crystallography optimization:
Screen detergent/lipid combinations systematically (including detergent:protein:lipid ratios)
Implement lipidic cubic phase (LCP) crystallization for membrane proteins
Use antibody fragments or nanobodies to stabilize flexible regions and promote crystal contacts
Explore fusion protein approaches (T4 lysozyme or BRIL insertions) to enhance crystallizability
Cryo-EM approach refinement:
Optimize sample vitrification conditions (blotting times, grid types, humidity)
Implement GraFix or amphipol reconstitution to improve particle orientation distribution
Consider focused refinement approaches for flexible regions
Use multi-body refinement to capture conformational heterogeneity
Hybrid method integration:
Combine lower-resolution cryo-EM maps with high-resolution X-ray structures of domains
Implement hydrogen-deuterium exchange mass spectrometry to identify flexible regions
Use solid-state NMR to determine key distance constraints in the membrane-embedded regions
Integrate computational models with experimental restraints
Sample preparation strategies:
Express the protein with stabilizing mutations identified through directed evolution
Co-purify with natural binding partners to stabilize native conformations
Implement nanodiscs or amphipols to maintain a native-like lipid environment
Screen metal ions and small molecule binders that might stabilize specific conformations
Data collection and processing optimization:
Implement advanced direct electron detector technologies for cryo-EM
Use microfocus beamlines for small crystals
Apply machine learning approaches for particle picking and classification
Implement Bayesian approaches for dealing with conformational heterogeneity