Recombinant Bovine Cytochrome c oxidase subunit 6C (COX6C)

Shipped with Ice Packs
In Stock

Description

Functional Roles in Cellular Metabolism

COX6C regulates oxidative phosphorylation (OXPHOS) and mitochondrial energy production. Key findings include:

Oxidative Phosphorylation

  • COX6C forms part of the COX complex, which drives proton pumping across the inner mitochondrial membrane .

  • Knockdown of COX6C reduces COX enzyme activity by 35% and ATP production in pancreatic cancer cells .

Apoptosis Regulation

  • COX6C release from mitochondria is linked to intrinsic apoptosis pathways .

Clinical and Pathological Relevance

COX6C overexpression is observed in cancers and metabolic disorders:

DiseaseCOX6C RoleSource
Pancreatic CancerUpregulated by Kras G12D mutations; enhances COX activity and ATP production
Prostate CancerGene amplification observed in tumor tissues
Chronic Kidney DiseaseAltered expression linked to mitochondrial dysfunction

Kras-Driven Pancreatic Cancer

  • BxPC-3/KrasG12D cells exhibit:

    • 18.2-fold mRNA upregulation and 4.6-fold protein upregulation of COX6C .

    • Increased mitochondrial mass, membrane potential, and ATP production .

  • COX6C siRNA knockdown reduces cell viability, COX activity, and ATP synthesis .

Tissue-Specific Expression

  • Immunohistochemistry: Strong cytoplasmic staining in pancreatic tumors vs. adjacent tissues .

Research Gaps and Future Directions

  1. Bovine-Specific Studies: No data exist on recombinant bovine COX6C. Potential applications could include:

    • Veterinary Medicine: Investigating mitochondrial disorders in livestock.

    • Biotechnology: Engineering COX6C variants for enhanced OXPHOS efficiency.

  2. Therapeutic Targets: COX6C inhibition may mitigate Kras-driven cancers by disrupting ATP production .

Methodological Approaches in COX6C Research

TechniqueApplicationOutcome
CRISPR/Cas9 EditingGenerating Kras G12D knock-in cell lines in pancreatic cancer models Identified COX6C as a Kras effector
siRNA KnockdownEvaluating COX6C’s role in ATP production and cell viability Reduced COX activity and ATP synthesis
ImmunohistochemistryAssessing COX6C overexpression in tumor vs. normal tissues Confirmed COX6C as a biomarker candidate

Product Specs

Form
Lyophilized powder
Please note: We will prioritize shipping the format that is currently in stock. However, if you have specific requirements for the format, please indicate them in your order notes. We will do our best to accommodate your request.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Please note: All our proteins are shipped with standard blue ice packs by default. If you require dry ice shipping, please notify us in advance as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. For optimal results, store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial prior to opening to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We suggest adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer ingredients, temperature, and the stability of the protein itself.
Generally, the shelf life for liquid form is 6 months at -20°C/-80°C. The shelf life for lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type will be determined during the production process. If you have a specific tag type preference, please inform us, and we will prioritize developing it accordingly.
Synonyms
COX6C; Cytochrome c oxidase subunit 6C; Cytochrome c oxidase polypeptide VIc; Cytochrome c oxidase subunit STA
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-74
Protein Length
Full Length of Mature Protein
Species
Bos taurus (Bovine)
Target Names
Target Protein Sequence
STALAKPQMRGLLARRLRFHIVGAFMVSLGFATFYKFAVAEKRKKAYADFYRNYDSMKDF EEMRKAGIFQSAK
Uniprot No.

Target Background

Function
Recombinant Bovine Cytochrome c oxidase subunit 6C (COX6C) is a component of cytochrome c oxidase, the final enzyme in the mitochondrial electron transport chain responsible for driving oxidative phosphorylation. The respiratory chain comprises three multisubunit complexes: succinate dehydrogenase (complex II, CII), ubiquinol-cytochrome c oxidoreductase (cytochrome b-c1 complex, complex III, CIII), and cytochrome c oxidase (complex IV, CIV). These complexes work in concert to transfer electrons derived from NADH and succinate to molecular oxygen, generating an electrochemical gradient across the inner mitochondrial membrane. This gradient drives transmembrane transport and the ATP synthase. Cytochrome c oxidase specifically catalyzes the reduction of oxygen to water. Electrons originating from reduced cytochrome c in the intermembrane space (IMS) are transferred via the dinuclear copper A center (CU(A)) of subunit 2 and heme A of subunit 1 to the active site located in subunit 1. This active site is a binuclear center (BNC) formed by heme A3 and copper B (CU(B)). The BNC reduces molecular oxygen to 2 water molecules using 4 electrons from cytochrome c in the IMS and 4 protons from the mitochondrial matrix.
Database Links
Protein Families
Cytochrome c oxidase subunit 6c family
Subcellular Location
Mitochondrion inner membrane; Single-pass membrane protein.

Q&A

What is Cytochrome c Oxidase Subunit 6C (COX6C) and what is its function in mitochondria?

COX6C is one of the nuclear-coded polypeptide chains of cytochrome c oxidase, which functions as the terminal oxidase in mitochondrial electron transport . As a component of Complex IV of the respiratory chain, COX6C plays a crucial role in cellular energy production through oxidative phosphorylation. The protein participates in the assembly of the cytochrome c oxidase complex, being incorporated during the S3 intermediate stage of assembly along with other subunits including COX2, COX3, COX5B, COX7B, COX7C, and COX8 . This strategic positioning within the complex suggests COX6C contributes to structural stability and optimal enzymatic function of the holoenzyme.

How is recombinant Bovine COX6C typically produced for research applications?

Recombinant Bovine COX6C can be produced through multiple expression systems, with the most common being in vitro E. coli expression systems . Alternative production methods include expression in yeast, baculovirus, mammalian cell culture systems, and in vivo biotinylation in E. coli . Each expression system offers distinct advantages depending on research requirements:

Expression SystemAdvantagesCommon Applications
E. coliHigh yield, cost-effective, rapid productionStructural studies, antibody production
YeastPost-translational modifications, higher eukaryotic processingFunctional studies requiring some modifications
BaculovirusComplex folding, most post-translational modificationsEnzymatic assays, interaction studies
MammalianNative-like folding and modificationsFunctional studies requiring authentic processing
In Vivo BiotinylationSite-specific labeling for detection/purificationProtein-protein interaction studies, pull-downs

The choice of expression system should be guided by the specific downstream applications and the requirement for post-translational modifications that may affect protein function .

What detection methods are most effective for COX6C in experimental settings?

Several validated detection methods exist for COX6C with varying sensitivities and applications:

  • Western Blotting: Effective for quantitative assessment of COX6C expression levels in cell and tissue lysates. Multiple validated antibodies are available (e.g., CSB-PA005856GA01HU, CSB-PA906089) with confirmed reactivity across human, mouse, and rat samples .

  • Immunohistochemistry (IHC): Valuable for assessing tissue localization and expression patterns in fixed tissue sections. This method has been successfully employed to demonstrate COX6C overexpression in cancer tissues compared to adjacent normal tissues .

  • Immunofluorescence (IF): Provides high-resolution cellular localization data, allowing co-localization studies with other mitochondrial markers .

  • ELISA: Enables quantitative measurement of COX6C in solution, with commercially available antibodies validated for this application .

For optimal results, antibody selection should consider species cross-reactivity requirements and the specific experimental application. Validation of antibody specificity through appropriate controls is essential, particularly when studying closely related COX subunits.

How does COX6C expression correlate with mitochondrial function in disease states?

COX6C expression shows significant correlation with mitochondrial function in various pathological conditions. In lung adenocarcinoma (LUAD), COX6C is frequently overexpressed due to copy number amplification of the 8q22.1-22.2 region . This overexpression appears to have functional consequences, as knockdown studies demonstrate that COX6C depletion causes:

  • Deficiency in mitochondrial fusion

  • Impairment of oxidative phosphorylation

  • Reactive oxygen species (ROS) accumulation

  • AMPK pathway activation

  • Mitotic arrest and cellular apoptosis

Similar patterns have been observed in pancreatic cancer, where COX6C overexpression correlates with increased mitochondrial mass, elevated ATP production, and enhanced mitochondrial membrane potential . Experimental evidence shows that silencing COX6C in pancreatic cancer cells with KRAS G12D mutation significantly decreases:

  • Cell viability

  • Cytochrome c oxidase enzyme activity

  • ATP production

These findings suggest that COX6C may serve as a metabolic regulator in cancer cells, potentially linking mitochondrial function to cell proliferation and survival mechanisms.

What role does COX6C play in the assembly and stability of the cytochrome c oxidase complex?

COX6C incorporation occurs during a specific stage of cytochrome c oxidase assembly, suggesting a regulated role in holoenzyme formation. Current models indicate that COX6C is added during the S3 intermediate stage, along with COX2, COX3, COX5B, COX7B, COX7C, and COX8 . This precise timing implies COX6C may serve as a structural component necessary for the subsequent incorporation of later subunits (COX7A, COX6B, and finally COX6A) to complete the holoenzyme.

Alternative models propose a more dynamic assembly process involving modular rather than sequential subunit addition . This model suggests the existence of a late-stage intermediate (LSI) complex and the possibility of nuclear-encoded subunit replacement within the holoenzyme. This mechanism might facilitate:

  • Exchange of isoform pairs during changing physiological conditions

  • Quality control processes to replace damaged components

  • Adaptation to tissue-specific or developmental requirements

Understanding the precise role of COX6C in these assembly processes could provide insights into mitochondrial adaptation mechanisms and potential therapeutic approaches for mitochondrial disorders.

What experimental approaches are most effective for studying COX6C-dependent mitochondrial bioenergetics?

Several methodological approaches have proven effective for investigating COX6C's role in mitochondrial bioenergetics:

  • Genetic Manipulation Techniques:

    • RNA interference (siRNA/shRNA) has been successfully employed to knockdown COX6C expression, demonstrating its effects on cellular bioenergetics

    • CRISPR/Cas9 systems can be utilized for gene editing to create stable cell lines with COX6C modifications

  • Mitochondrial Function Assessment:

    • Cytochrome c oxidase enzymatic activity assays directly measure COX complex function (35% increase observed in KRAS G12D mutant cells compared to wild-type)

    • ATP production assays using metabolite substrates reveal the functional consequences of COX6C manipulation

    • Mitochondrial membrane potential measurements using fluorescent probes indicate the integrity of the electron transport chain

  • Metabolic Phenotyping:

    • Oxygen consumption rate (OCR) measurements using Seahorse technology to assess oxidative phosphorylation capacity

    • Extracellular acidification rate (ECAR) analysis to determine glycolytic shifts following COX6C manipulation

    • Mitochondrial fusion/fission dynamics assessment through confocal microscopy and appropriate fluorescent markers

  • Molecular Interaction Studies:

    • Co-immunoprecipitation assays to identify protein-protein interactions involving COX6C

    • Blue native PAGE to analyze intact respiratory complexes and supercomplexes

    • Crosslinking mass spectrometry to map precise interaction sites within the COX complex

When implementing these approaches, careful experimental design should include appropriate controls and validation steps to account for potential compensatory mechanisms that might mask COX6C-specific effects.

What are the critical factors to consider when designing experiments to study COX6C function?

When investigating COX6C function, several experimental design considerations are critical for generating reliable and interpretable data:

  • Expression System Selection:

    • For structural studies: E. coli-expressed recombinant COX6C may be sufficient

    • For functional studies: Expression systems that maintain native conformation and post-translational modifications (mammalian or baculovirus) are preferred

    • Consider species-specific differences when selecting recombinant proteins for cross-species studies

  • Cell/Tissue Model Selection:

    • Account for tissue-specific expression patterns of COX6C

    • Consider potential differences in mitochondrial function across cell types

    • Cancer vs. normal cell models may exhibit fundamentally different mitochondrial phenotypes and COX6C dependence

  • Knockdown/Knockout Approaches:

    • Transient vs. stable knockdown will reveal different aspects of function (acute vs. adaptive responses)

    • Include rescue experiments with wild-type and mutant constructs to confirm specificity

    • Consider compensatory mechanisms through other subunits, particularly isoform pairs

  • Appropriate Controls:

    • Include mitochondrial mass normalization for bioenergetic measurements

    • Account for cell cycle effects, as COX6C knockdown induces S-G2/M arrest

    • Measure multiple parameters of mitochondrial function to distinguish primary from secondary effects

  • Temporal Considerations:

    • Mitochondrial turnover rates affect the timing of observable phenotypes

    • Assembly dynamics of respiratory complexes may create time-dependent effects

    • ROS accumulation and subsequent signaling represent both acute and chronic responses

How can researchers effectively measure COX6C-dependent changes in mitochondrial electron transport?

To quantify COX6C-dependent alterations in mitochondrial electron transport, researchers should employ complementary methodological approaches:

  • Direct Activity Measurement:

    • Isolated cytochrome c oxidase activity assays using spectrophotometric methods to monitor the oxidation of reduced cytochrome c

    • Polarographic oxygen consumption measurements using Clark-type electrodes

    • High-resolution respirometry with substrate-uncoupler-inhibitor titration protocols

  • Integrated Bioenergetic Assessment:

    • Respirometric analysis of intact cells using the Seahorse XF Analyzer to measure:

      • Basal respiration

      • ATP-linked respiration

      • Maximal respiratory capacity

      • Spare respiratory capacity

      • Proton leak

  • Electron Flow Analysis:

    • Sequential electron transport complex inhibition to isolate Complex IV-specific function

    • Analysis of respiratory control ratios to assess coupling efficiency

    • Membrane potential measurements using potentiometric dyes (TMRM, JC-1)

  • Metabolic Flux Analysis:

    • Isotope labeling studies to track carbon flux through the TCA cycle

    • Metabolomic profiling to identify adaptive metabolic shifts following COX6C manipulation

    • Integration of glycolytic and oxidative metabolism measurements

  • ROS Production Assessment:

    • Site-specific ROS measurements to determine electron leak points

    • Antioxidant system response evaluation

    • Oxidative damage marker quantification

Studies have demonstrated that COX6C knockdown in cancer cells results in significant decreases in COX enzyme activity and subsequent ATP production, highlighting the protein's functional importance in maintaining electron transport chain efficiency .

How does COX6C expression correlate with cancer progression, and what are the implications for targeted therapies?

COX6C expression shows distinct patterns in cancer progression with significant therapeutic implications:

  • Expression Patterns:

    • Lung adenocarcinoma (LUAD): COX6C is frequently overexpressed due to copy number amplification of chromosome 8q22.1-22.2

    • Pancreatic cancer: Significant COX6C overexpression is observed in tumor tissues compared to matched adjacent tissues

    • KRAS-mutant contexts: COX6C expression is particularly elevated in KRAS G12D mutant pancreatic cancer cells (18.2-fold mRNA elevation, 4.6-fold protein elevation)

  • Functional Consequences:

    • Mitochondrial function enhancement in cancer cells

    • Increased ATP production supporting rapid proliferation

    • Metabolic adaptation to oncogenic signaling (particularly KRAS-driven oncogenesis)

  • Therapeutic Vulnerability:

    • COX6C knockdown induces:

      • Mitochondrial dysfunction

      • ROS accumulation

      • AMPK activation

      • Mitotic arrest

      • Cellular apoptosis

    These outcomes suggest COX6C may represent a targetable vulnerability in cancers with its overexpression.

  • Biomarker Potential:

    • Copy amplification-mediated COX6C upregulation might serve as a prognostic biomarker for lung adenocarcinoma

    • Immunohistochemical detection shows promise for clinical application, with significantly more intense and diffuse COX6C immunoreactivity in tumor tissue

What research methods are most effective for evaluating COX6C as a therapeutic target?

Evaluating COX6C as a therapeutic target requires multifaceted research approaches:

  • Target Validation Studies:

    • Genetic silencing through RNA interference or CRISPR/Cas9 in diverse cancer models

    • Correlation of COX6C expression with patient outcomes across cancer types

    • Synthetic lethality screens to identify contexts where COX6C inhibition is selectively toxic

  • Pharmacological Approaches:

    • Development of small molecule inhibitors targeting COX6C specifically

    • Evaluation of compounds that disrupt COX6C incorporation into the holoenzyme

    • Testing of mitochondrial-targeted drug delivery systems to enhance specificity

  • Precision Medicine Applications:

    • Generation of patient-derived models with varying COX6C expression levels

    • Identification of predictive biomarkers for response to COX6C-targeted therapies

    • Integration with genomic profiling to identify synthetic lethal combinations

  • Combination Strategy Assessment:

    • Evaluation with existing chemotherapeutics

    • Testing with mitochondrial-targeted agents

    • Combination with AMPK modulators, given the activation of AMPK following COX6C inhibition

  • Translational Models:

    • Patient-derived xenografts to assess efficacy in heterogeneous tumors

    • Genetically engineered mouse models, particularly those with KRAS mutations

    • Ex vivo tumor slice cultures for rapid personalized therapeutic testing

What are common technical challenges in COX6C research and how can they be addressed?

Researchers working with COX6C encounter several technical challenges that require specific troubleshooting approaches:

  • Protein Solubility and Stability Issues:

    • Challenge: Recombinant COX6C, as a mitochondrial membrane protein component, often presents solubility problems.

    • Solution: Consider using different expression systems (E. coli, yeast, baculovirus, or mammalian) based on experimental needs . For structural studies, incorporate appropriate detergents during purification and stabilize with lipid nanodisc technology when necessary.

  • Antibody Specificity Concerns:

    • Challenge: Cross-reactivity with other cytochrome c oxidase subunits due to structural similarity.

    • Solution: Validate antibodies using knockdown/knockout controls and perform comparative analysis with multiple antibodies targeting different epitopes . Consider using tagged recombinant proteins when possible.

  • Assembly Pathway Complexity:

    • Challenge: Distinguishing direct COX6C effects from secondary consequences on complex assembly.

    • Solution: Employ blue native PAGE to visualize assembly intermediates, use pulse-chase experiments to track assembly kinetics, and implement time-course studies after COX6C manipulation .

  • Functional Redundancy:

    • Challenge: Compensatory mechanisms through other subunits may mask COX6C-specific phenotypes.

    • Solution: Consider acute manipulation strategies (inducible systems), analyze expression changes in other subunits after COX6C targeting, and employ combination approaches targeting multiple components.

  • Mitochondrial Heterogeneity:

    • Challenge: Variations in mitochondrial function across cell types and conditions.

    • Solution: Include appropriate cellular controls, normalize results to mitochondrial content, and consider single-cell approaches to account for population heterogeneity.

How can researchers optimize experimental protocols for studying COX6C-protein interactions and complex assembly?

Optimizing protocols for studying COX6C interactions and assembly requires attention to several methodological considerations:

  • Crosslinking Approaches:

    • Implement chemical crosslinking followed by mass spectrometry (XL-MS) to capture transient interactions

    • Use membrane-permeable crosslinkers for in vivo studies of intact mitochondria

    • Optimize crosslinker concentration and reaction time to avoid artifacts while maximizing capture efficiency

  • Co-immunoprecipitation Optimization:

    • Select detergents that preserve native interactions while solubilizing membrane proteins

    • Consider proximity-dependent biotinylation (BioID, TurboID) to capture weak or transient interactions

    • Include appropriate controls: isotype antibodies, reverse IP verification, and validation in knockdown systems

  • Assembly Kinetics Assessment:

    • Employ pulse-chase labeling with inducible expression systems

    • Analyze assembly intermediates using blue native PAGE at multiple time points

    • Combine with selective inhibitors of mitochondrial protein import or translation to distinguish assembly steps

  • Structural Studies Enhancement:

    • Consider cryo-electron microscopy for complex structures

    • Implement hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • Use molecular dynamics simulations to predict conformational changes upon binding

  • System Perturbation Approaches:

    • Apply mild stress conditions to reveal conditional dependencies

    • Test assembly under varying energy states to identify regulatory mechanisms

    • Evaluate the impact of post-translational modifications on interaction dynamics

Current research indicates that COX6C incorporation occurs during specific assembly stages, with integration into the S3 intermediate alongside other subunits . Understanding these assembly dynamics can provide insights into both normal mitochondrial function and disease mechanisms.

What emerging technologies could advance our understanding of COX6C function?

Several cutting-edge technologies show promise for deepening our understanding of COX6C biology:

  • Cryo-Electron Tomography:

    • Enables visualization of COX6C within the native mitochondrial membrane environment

    • Provides structural insights at near-atomic resolution without crystallization

    • Can reveal conformational changes under different functional states

  • Single-Cell Omics:

    • Single-cell proteomics to assess COX6C expression heterogeneity

    • Spatial transcriptomics to map expression patterns within complex tissues

    • Integration of genomic, transcriptomic, and proteomic data for comprehensive understanding

  • Optogenetic and Optochemical Tools:

    • Light-inducible protein targeting to achieve temporal control of COX6C function

    • Photoswitchable inhibitors for precise manipulation of activity

    • Genetically encoded sensors for real-time monitoring of complex assembly

  • CRISPR Base Editing and Prime Editing:

    • Precise modification of COX6C without double-strand breaks

    • Introduction of specific mutations to study structure-function relationships

    • Modeling of disease-associated variants with minimal off-target effects

  • Microfluidic Organ-on-Chip Technology:

    • Recapitulation of tissue-specific microenvironments

    • Real-time assessment of mitochondrial function in physiologically relevant models

    • High-throughput screening of compounds targeting COX6C or its interactions

These technologies could help address unresolved questions about COX6C's precise role in complex assembly, tissue-specific functions, and its potential as a therapeutic target in conditions like cancer where its expression is dysregulated .

What are the most promising areas for future COX6C research in disease contexts?

Several research directions show particular promise for advancing our understanding of COX6C in disease:

  • Cancer Metabolism and Therapy:

    • Further investigation of COX6C as a biomarker for prognosis and treatment response

    • Development of COX6C-targeted therapies for cancers with overexpression

    • Exploration of synthetic lethal interactions with other metabolic pathways

    • Understanding the role of COX6C in metastasis and therapy resistance

  • Neurodegenerative Diseases:

    • Examination of COX6C's role in maintaining neuronal bioenergetics

    • Investigation of potential alterations in assembly or function in conditions like Alzheimer's and Parkinson's diseases

    • Exploration of neuroprotective strategies targeting COX6C-dependent pathways

  • Metabolic Disorders:

    • Analysis of COX6C's contribution to mitochondrial dysfunction in diabetes and obesity

    • Evaluation of tissue-specific roles in metabolic adaptation

    • Investigation of post-translational modifications affecting COX6C function in metabolic disease states

  • Aging and Mitochondrial Dynamics:

    • Characterization of age-associated changes in COX6C expression and incorporation

    • Investigation of COX6C's role in mitochondrial quality control mechanisms

    • Exploration of interventions targeting COX6C to improve mitochondrial function in aging

  • Developmental Biology:

    • Analysis of COX6C's role during embryonic development and cellular differentiation

    • Investigation of tissue-specific isoform expression patterns

    • Understanding the regulatory mechanisms controlling COX6C expression during development

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.