Recombinant Trachypithecus cristatus COX6C is produced using E. coli expression systems, ensuring high purity and scalability. Key details include:
This recombinant protein is utilized in:
ELISA: Detecting COX6C antibodies or quantifying protein levels in primate models .
Comparative studies: Investigating evolutionary divergence of mitochondrial complexes between primates and humans .
Antibody development: Polyclonal antibodies against human COX6C (e.g., ab150422) show cross-reactivity, aiding immunohistochemistry in primate tissues .
Mitochondrial dysfunction: COX6C mutations in humans are linked to metabolic disorders and cancers (e.g., prostate cancer) . Studying the primate homolog clarifies conserved mechanistic roles.
Assembly mechanisms: COX6C interacts with metallochaperones like COX16 and SCO1 during Complex IV maturation, as shown in human cell studies .
Trachypithecus cristatus (silvered leaf-monkey) COX6C is a nuclear-encoded structural subunit of cytochrome c oxidase (Complex IV), the terminal enzyme in the mitochondrial electron transport chain. This subunit plays a crucial role in the regulation of oxidative phosphorylation (OXPHOS). The protein consists of approximately 75 amino acids and is involved in electron transfer from reduced cytochrome c to molecular oxygen, ultimately contributing to ATP production .
Unlike several other COX subunits, COX6C has no paralogs in any vertebrate lineage, including mammals, suggesting its evolutionarily conserved and non-redundant function . The mature protein shows significant homology between species, with approximately 69-73% sequence similarity when comparing rat, bovine, and human variants, indicating its functional importance across mammalian species .
When studying T. cristatus COX6C, researchers should note that it functions within the respiratory chain that contains three multisubunit complexes: succinate dehydrogenase (Complex II), ubiquinol-cytochrome c oxidoreductase (Complex III), and cytochrome c oxidase (Complex IV) . These complexes cooperatively transfer electrons to create an electrochemical gradient that drives ATP synthesis.
Designing expression vectors for recombinant T. cristatus COX6C requires careful consideration of multiple factors:
Gene sequence optimization: The COX6C gene sequence should be codon-optimized for the selected expression system. This typically involves:
Adapting codon usage bias to match the expression host
Removing cryptic splice sites and unwanted regulatory elements
Optimizing GC content and removing repetitive sequences
Vector selection: Consider vectors with:
Affinity tag placement: Based on structural considerations and the nature of COX6C:
N-terminal tags are often preferred due to the membrane-associating properties of COX6C
Inclusion of a protease cleavage site between the tag and COX6C
Consider TEV or PreScission protease sites for tag removal
When designing your constructs, it's crucial to verify the full 933-bp linear DNA sequence of COX6C to ensure all functional domains are preserved . Given that COX6C is typically located at chromosome 8q22.2 in humans (with potential variations in T. cristatus), proper gene annotation verification is essential before proceeding with cloning .
Several expression systems have been employed for recombinant COX6C production, each with distinct advantages:
For T. cristatus COX6C specifically, wheat germ cell-free systems have shown success with human COX6C and would likely be appropriate for the closely related T. cristatus variant . This system preserves the protein in the 1 to 75 amino acid range with high fidelity to the native structure.
When selecting an expression system, consider the downstream applications. For interaction studies with other mitochondrial proteins, mammalian expression may be preferable to maintain physiologically relevant modifications. For structural studies requiring large protein quantities, E. coli or insect cell systems might be more appropriate.
Purification of recombinant T. cristatus COX6C requires a multi-step approach to achieve high purity while maintaining protein functionality:
Initial capture: Affinity chromatography based on the fusion tag:
Intermediate purification:
Ion exchange chromatography (IEX): Based on COX6C's theoretical isoelectric point
Size exclusion chromatography (SEC): To separate monomeric COX6C from aggregates
Tag cleavage: Using specific proteases like TEV or PreScission
Polishing step:
Reverse-phase HPLC for highest purity
Second affinity step to remove cleaved tags
A typical purification protocol might include:
Lysis in a buffer containing 20 mM Tris-HCl (pH 8.0), 150 mM NaCl, 1% detergent (if membrane-associated), and protease inhibitors
Affinity purification using the appropriate resin
Buffer exchange to remove imidazole or glutathione
Overnight tag cleavage at 4°C
Second affinity step to remove cleaved tag
Size exclusion chromatography for final polishing
Throughout purification, it's crucial to monitor COX6C by SDS-PAGE and Western blotting using antibodies against COX6C or the fusion tag . Activity assays measuring cytochrome-c oxidase activity should be performed to confirm functionality .
Verification of recombinant T. cristatus COX6C requires comprehensive characterization:
Structural integrity assessment:
Mass spectrometry (MS): To confirm the exact molecular weight
Circular dichroism (CD): To evaluate secondary structure content
Fluorescence spectroscopy: To assess tertiary structure
Limited proteolysis: To determine domain stability
Dynamic light scattering (DLS): To check homogeneity and aggregation state
Functional verification:
Immunological confirmation:
A functional COX6C should demonstrate:
Proper integration into the cytochrome c oxidase complex
Contribution to electron transfer from cytochrome c to oxygen
Expected protein-protein interactions with other subunits
Correct subcellular localization to mitochondria when expressed in cells
Given COX6C's role in respiratory function, oxidative phosphorylation activity measurements in reconstituted systems provide the most direct assessment of functionality .
Studying the COX6C interactome requires sophisticated approaches to capture both stable and transient interactions:
Affinity purification-mass spectrometry (AP-MS):
Express tagged T. cristatus COX6C in relevant cell lines
Perform gentle cell lysis preserving native interactions
Capture COX6C and associated proteins via affinity purification
Identify interacting partners through LC-MS/MS
Compare to control pulldowns to eliminate false positives
Proximity-based labeling techniques:
BioID: Fusion of COX6C with a promiscuous biotin ligase
APEX2: Fusion with an engineered ascorbate peroxidase
TurboID: Using an improved biotin ligase for faster labeling
These approaches allow identification of proteins in close proximity to COX6C in living cells
Crosslinking mass spectrometry (XL-MS):
Use homo- or hetero-bifunctional crosslinkers
Identify crosslinked peptides by MS to map interaction interfaces
Particularly valuable for membrane protein complexes like cytochrome c oxidase
Known interacting partners of COX6C include MT-CO1, MT-CO2, COX5A, and other components of the cytochrome c oxidase complex . Additionally, interactions with regulatory proteins such as SURF1 and LRRK2 have been reported . These established interactions serve as positive controls for new interactome studies with T. cristatus COX6C.
The interactome data can be analyzed using:
Protein interaction network visualization tools
Functional enrichment analysis
Evolutionary conservation mapping across primate species
Integration with pathway databases focused on mitochondrial function
Investigating tissue-specific expression patterns of T. cristatus COX6C requires a multi-faceted approach:
Transcriptional profiling:
RNA-Seq analysis across different T. cristatus tissues
qRT-PCR validation of expression levels using tissue-specific samples
Single-cell RNA-Seq to identify cell-type-specific expression
Comparison with expression patterns in other primates for evolutionary insights
Protein-level detection:
Immunohistochemistry (IHC) on tissue sections using COX6C-specific antibodies
Western blotting of tissue lysates with quantitative analysis
Targeted proteomics (SRM/MRM) for absolute quantification
Analysis of post-translational modifications across tissues
Regulatory mechanism investigation:
Based on studies in other species, COX6C expression may vary significantly across tissues, with particularly high expression in metabolically active tissues like heart, brain, and skeletal muscle . Mitochondrial biogenesis in response to environmental factors such as temperature can also influence COX6C expression levels .
A comparative analysis approach should include:
Correlation of expression with tissue-specific metabolic demands
Examination of co-regulated genes in the OXPHOS pathway
Analysis of expression changes during development and aging
Comparison with human expression patterns to identify primate-specific regulation
Post-translational modifications (PTMs) of T. cristatus COX6C can be comprehensively studied using:
Mass spectrometry-based approaches:
Bottom-up proteomics: Enzymatic digestion followed by LC-MS/MS
Top-down proteomics: Analysis of intact protein to preserve PTM combinations
Middle-down approach: Limited proteolysis for analysis of larger fragments
Targeted MS methods: SRM/MRM for quantification of specific modifications
Enrichment strategies for specific PTMs:
Phosphorylation: TiO2, IMAC, or phospho-specific antibodies
Acetylation: Anti-acetyl lysine antibodies
Ubiquitination: Tandem ubiquitin binding entities (TUBEs)
Oxidative modifications: Derivatization approaches (e.g., dimedone for sulfenic acids)
Functional impact assessment:
Site-directed mutagenesis of modified residues
In vitro enzymatic assays with modified and unmodified protein
Structural analysis using NMR or X-ray crystallography
Molecular dynamics simulations to predict PTM effects on protein dynamics
While specific PTMs of T. cristatus COX6C are not well-characterized, studies in human COX6C suggest potential regulatory modifications. The human protein shows evidence of regulation through splicing efficiency by DAZAP1, which may involve downstream PTM regulation .
A comprehensive PTM analysis workflow should include:
Careful sample preparation to preserve labile modifications
Parallel analysis of samples from different physiological conditions
Validation of identified PTMs using orthogonal methods
Functional studies to determine the impact on COX6C activity
Evolutionary comparison of modification sites across primate species
Studying evolutionary conservation of COX6C across primates requires:
Comparative genomic analysis:
Multiple sequence alignment of COX6C coding sequences
Phylogenetic tree construction using maximum likelihood or Bayesian methods
Calculation of selection pressure using dN/dS ratios
Identification of conserved domains and species-specific variations
Synteny analysis of the genomic region containing COX6C
Structural and functional comparison:
Homology modeling of T. cristatus COX6C based on available structures
Identification of functionally important residues across species
Analysis of protein-protein interaction interfaces
Prediction of functional consequences of species-specific variations
Experimental validation:
Recombinant expression of COX6C from multiple primate species
Comparative functional assays for cytochrome c oxidase activity
Cross-species complementation experiments
Chimeric protein studies to identify functionally divergent regions
COX6C is notable for having no paralogs in any vertebrate lineage, suggesting strong evolutionary constraints . The mature protein shows significant sequence conservation with approximately 69-73% homology between rats, bovines, and humans .
Research should focus on:
Correlation between evolutionary changes and primate-specific metabolic adaptations
Comparison of regulatory mechanisms across primate species
Analysis of COX6C in relation to chromosomal rearrangements in primate evolution
Integration of findings with broader studies of mitochondrial evolution in primates
Investigating the role of T. cristatus COX6C in response to environmental stressors requires:
Experimental stress models:
Multi-omics approaches:
Transcriptomics: RNA-Seq to monitor COX6C expression changes
Proteomics: Quantitative analysis of COX6C and interacting proteins
Metabolomics: Assessment of changes in OXPHOS metabolites
Integrated pathway analysis combining multiple data types
Dynamic measurements:
Real-time quantification of COX6C expression using reporter constructs
Live-cell imaging of mitochondrial function
Temporal analysis of stress response pathways
Recovery kinetics after stress removal
Studies have shown that COX6C expression can be modulated by various stressors. For example, mitochondrial biogenesis stimulated by low temperature is reflected in increased expression and activity of COX6C . During viral infections, particularly influenza, host cells initially boost COX6C mRNA expression by silencing miR-4276 as a defense mechanism .
A comprehensive experimental design should include:
Time-course experiments to capture both immediate and adaptive responses
Dose-response studies to determine threshold effects
Combined stressors to model complex environmental conditions
Parallel analysis in multiple cell types or tissues
Comparison with responses in human or other primate cells
Structural characterization of T. cristatus COX6C presents several technical challenges:
Protein production challenges:
Maintaining structural integrity during recombinant expression
Ensuring proper membrane association and folding
Obtaining sufficient quantities for structural studies
Preventing aggregation of this hydrophobic protein
Crystallization barriers:
Difficulty in crystallizing membrane-associated proteins
Need for appropriate detergents or lipid environments
Challenge of obtaining well-diffracting crystals
Requirement for stabilizing protein-protein interactions
Alternative structural approaches:
Cryo-EM for structure determination within the intact cytochrome c oxidase complex
Solution NMR for dynamic studies of isolated domains
Small-angle X-ray scattering (SAXS) for low-resolution envelope determination
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for conformational dynamics
Computational methods:
Molecular dynamics simulations in membrane environments
Ab initio structure prediction using deep learning approaches
Integrative modeling combining multiple experimental data sources
Evolutionary coupling analysis to predict structural contacts
The most effective approach is likely to be integrative, combining:
High-resolution structural techniques for isolated domains
Cryo-EM for the intact cytochrome c oxidase complex
Crosslinking mass spectrometry to map interaction interfaces
Computational modeling to fill gaps in experimental data
Researchers should consider that COX6C functions as part of the larger cytochrome c oxidase complex, which includes multiple subunits that cooperatively catalyze the reduction of oxygen to water . Structural studies should aim to understand both the isolated protein and its role within this larger complex.
Optimizing enzymatic activity assays for recombinant T. cristatus COX6C requires careful consideration of multiple factors:
Direct activity measurement approaches:
Oxygen consumption assays using Clark-type electrodes
Spectrophotometric assays monitoring cytochrome c oxidation
Polarographic methods to measure electron transfer rates
Artificial electron acceptor/donor systems for isolated protein studies
Reconstitution strategies:
Incorporation into liposomes or nanodiscs
Co-expression with other cytochrome c oxidase subunits
Stepwise assembly of the complex in vitro
Integration into mitochondrial membrane fractions
Assay optimization parameters:
Buffer composition (pH, ionic strength, divalent cations)
Detergent type and concentration
Lipid composition of reconstitution systems
Temperature and reaction time course
Substrate concentrations (reduced cytochrome c)
Controls and validation:
Specific inhibitors (e.g., cyanide, azide) for validation
Comparison with native enzyme activity
Mutational analysis of key residues
Competition assays with purified COX6C
A comprehensive enzymatic characterization should include:
Determination of kinetic parameters (Km, Vmax, kcat)
Effects of physiological regulators and inhibitors
pH and temperature profiles
Stability studies under various conditions
Comparison with human COX6C activity
Given the role of COX6C in electron transfer and the reduction of oxygen to water , researchers should ensure that activity assays effectively capture this specific function within the context of the complete cytochrome c oxidase complex.
Resolving contradictions in COX6C functional data requires systematic approaches:
Standardized methodology development:
Establish consensus protocols for COX6C expression and purification
Define standard activity assay conditions
Create reference datasets for benchmarking
Develop validated antibodies and detection methods
Multi-laboratory validation studies:
Collaborative projects testing the same hypotheses
Round-robin testing of identical protein preparations
Meta-analysis of published data with rigorous quality assessment
Pre-registered replication studies of key findings
Addressing specific contradictions:
Species-specific differences: Systematic comparison across species including T. cristatus
Isoform-specific effects: Careful characterization of all potential isoforms
Context-dependent functions: Testing in multiple cellular/tissue environments
Regulatory discrepancies: Comprehensive analysis of regulatory pathways
Advanced technologies to provide new perspectives:
Single-molecule techniques to address population heterogeneity
In situ structural studies using cryo-electron tomography
Systems biology approaches integrating multiple data types
CRISPR screening for genetic interactions
When examining contradictory findings, researchers should consider:
The dual role of COX6C in electron transport and potential regulatory functions
The influence of experimental conditions on membrane protein behavior
Tissue-specific variations in function and regulation
Potential differences between recombinant and native proteins
For example, contradictory findings regarding COX6C in disease models (such as its upregulation in diabetic nephropathy but downregulation in end-stage renal disease) can be addressed by careful time-course studies and consideration of disease progression stages.
T. cristatus COX6C offers unique opportunities for understanding primate-specific mitochondrial biology:
Comparative studies across primates:
Functional comparison with human and other primate COX6C
Analysis of primate-specific adaptations in mitochondrial function
Investigation of lineage-specific selection pressures
Integration with broader studies of primate evolution
Experimental model development:
T. cristatus cell lines for mitochondrial studies
CRISPR-engineered human cells expressing T. cristatus COX6C
Hybrid mitochondrial complexes with components from different species
Computational models of species-specific mitochondrial function
Evolutionary medicine applications:
Insights into primate-specific mitochondrial disorders
Understanding adaptive changes in OXPHOS efficiency
Investigation of metabolic adaptations to different environments
Analysis of mitochondrial contributions to primate longevity
T. cristatus represents an interesting evolutionary position, with its COX6C showing significant homology to human COX6C but with species-specific variations. The silvered leaf monkey has also been the subject of detailed chromosomal homology studies with humans, which can provide context for understanding the genomic environment of COX6C .
Future research directions could include:
The role of COX6C in primate-specific metabolic adaptations
Comparative analysis of regulatory mechanisms across primates
Investigation of mitochondrial-nuclear co-evolution in primates
Development of primate-specific mitochondrial disease models
Several cutting-edge technologies promise to revolutionize COX6C research:
Advanced imaging techniques:
Super-resolution microscopy for subunit localization within complexes
Single-particle cryo-EM for high-resolution structural studies
Correlative light and electron microscopy (CLEM) for structure-function studies
Live-cell imaging with genetically encoded sensors
Next-generation genomic and transcriptomic approaches:
Long-read sequencing for complete locus characterization
Single-cell multi-omics for cell-type-specific analysis
Spatial transcriptomics for tissue-specific expression patterns
CRISPR screens for functional genomics
Protein engineering and synthetic biology:
Directed evolution of COX6C variants with enhanced properties
De novo design of artificial COX6C with novel functions
Optogenetic control of COX6C activity
Engineered allosteric regulators of COX6C function
Computational and AI approaches:
AlphaFold2 and RoseTTAFold for structure prediction
Machine learning for predicting functional effects of mutations
Systems biology models of mitochondrial function
Network analysis of mitochondrial protein interactions
These technologies will enable:
Higher resolution understanding of COX6C's role within Complex IV
More precise manipulation of COX6C function in experimental systems
Better integration of structural, functional, and evolutionary data
Development of new therapeutic approaches targeting COX6C-related pathways
As research progresses, integration of data across multiple technologies will be crucial for a comprehensive understanding of T. cristatus COX6C biology and its relevance to human mitochondrial function and disease.