Recombinant Pongo pygmaeus Cytochrome c oxidase subunit 6C (COX6C)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them when placing your order, and we will accommodate your request.
Lead Time
Delivery time may vary depending on the purchasing method and location. Kindly consult your local distributors for specific delivery timelines.
Note: All proteins are shipped with standard blue ice packs by default. Should you require dry ice shipping, please inform us in advance, as additional charges may apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging this vial prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard final glycerol concentration is 50%. Customers may use this as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer components, storage temperature, and the protein's inherent stability.
Typically, the shelf life for liquid form is 6 months at -20°C/-80°C. For lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
Tag type is determined during production. If you have a specific tag type in mind, please inform us, and we will prioritize developing the specified tag.
Synonyms
COX6C; Cytochrome c oxidase subunit 6C; Cytochrome c oxidase polypeptide VIc
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-75
Protein Length
Full Length of Mature Protein
Species
Pongo pygmaeus (Bornean orangutan)
Target Names
Target Protein Sequence
APEVLPKPRMRGLLARRLRNHMAVAFVLSLGVAALYKFRVADRRKKAYADFYRNYDVMKD FEEMRKAGIFQSVK
Uniprot No.

Target Background

Function
Cytochrome c oxidase subunit 6C (COX6C) is a component of cytochrome c oxidase, the terminal 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 collaborate to transfer electrons derived from NADH and succinate to molecular oxygen, generating an electrochemical gradient across the inner mitochondrial membrane. This gradient powers transmembrane transport and the activity of ATP synthase. Cytochrome c oxidase is the component that catalyzes the reduction of oxygen to water. Electrons originating from reduced cytochrome c in the intermembrane space (IMS) are relayed through the dinuclear copper A center (CU(A)) of subunit 2 and heme A of subunit 1 to the active site within 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.
Protein Families
Cytochrome c oxidase subunit 6c family
Subcellular Location
Mitochondrion inner membrane; Single-pass membrane protein.

Q&A

What is the genomic location and structure of COX6C?

COX6C is located on chromosome 8 (8q22.1-22.2) in humans. The gene encodes a protein that functions as a component of the mitochondrial electron transport chain, specifically Complex IV (cytochrome c oxidase). This complex serves as the final electron acceptor in the respiratory chain, facilitating the transfer of electrons to oxygen. While this information pertains to human COX6C, the Pongo pygmaeus (orangutan) COX6C shares significant homology with its human counterpart due to evolutionary conservation of mitochondrial proteins among primates .

How does COX6C contribute to mitochondrial function?

COX6C functions as a critical component of the cytochrome c oxidase complex, which is the terminal enzyme of the mitochondrial respiratory chain. Experimental evidence demonstrates that COX6C depletion causes deficiency in mitochondrial fusion and impairment of oxidative phosphorylation. This disruption leads to reactive oxygen species (ROS) accumulation and activation of the AMPK pathway, which can subsequently affect various cellular processes including spindle formation and chromosome segregation .

What are the key differences between recombinant and endogenous COX6C?

Recombinant Pongo pygmaeus COX6C is produced through molecular cloning and expression in heterologous systems (commonly E. coli, yeast, or mammalian cells), allowing for protein modification, purification, and functional characterization. Unlike endogenous COX6C, recombinant versions typically include affinity tags (e.g., His-tag, GST-tag) to facilitate purification and may exhibit differences in post-translational modifications depending on the expression system used. These differences must be considered when interpreting experimental results, as they can affect protein folding, activity, and interaction with other cellular components.

What are the optimal methods for expressing and purifying recombinant Pongo pygmaeus COX6C?

For recombinant Pongo pygmaeus COX6C expression, researchers should consider the following protocol:

  • Expression System Selection: Bacterial systems (E. coli BL21) are suitable for high yield but may lack proper post-translational modifications. Yeast (Pichia pastoris) or insect cell (Sf9) systems better approximate eukaryotic modifications.

  • Vector Construction: Insert the COX6C cDNA into an expression vector containing an appropriate promoter (T7 for bacteria, AOX1 for yeast) and a purification tag (His6, GST, or FLAG).

  • Expression Optimization: For bacterial expression, optimize induction conditions (IPTG concentration: 0.1-1.0 mM; temperature: 16-37°C; duration: 3-18 hours). Lower temperatures (16-20°C) often improve folding of mitochondrial proteins.

  • Purification Strategy:

    • Affinity chromatography using Ni-NTA (for His-tagged proteins)

    • Size exclusion chromatography for higher purity

    • Ion exchange chromatography as a final polishing step

  • Protein Refolding: Since COX6C is a mitochondrial membrane-associated protein, consider detergent-assisted refolding using mild detergents (0.1% DDM or CHAPS).

What assays are most effective for measuring COX6C activity and function?

Several complementary approaches can be employed to assess COX6C activity and function:

  • Mitochondrial Membrane Potential (MMP) Assay: Using JC-1 dye, which accumulates in mitochondria and exhibits potential-dependent fluorescence. COX6C functionality directly impacts this potential, with functional COX6C maintaining higher red/green fluorescence ratios .

  • Oxygen Consumption Rate (OCR): Measure using a Seahorse XF Analyzer to quantify the rate of oxidative phosphorylation, which is directly influenced by COX6C function.

  • Complex IV Activity Assay: Spectrophotometric monitoring of cytochrome c oxidation at 550 nm, providing direct measurement of the enzymatic activity of the complex containing COX6C.

  • Intracellular ROS Measurement: Utilizing H2DCFDA (10 μM, 20 min incubation at 37°C) followed by flow cytometry to detect ROS accumulation, which increases with COX6C dysfunction .

  • Blue Native PAGE: For assessing the integrity and assembly of respiratory complexes containing COX6C.

How can researchers effectively knockdown or overexpress COX6C in cellular models?

For manipulating COX6C expression levels in research models:

  • Knockdown Approaches:

    • siRNA transfection: Most effective with lipid-based transfection reagents (Lipofectamine) using 20-50 nM siRNA for 24-72 hours

    • shRNA lentiviral vectors: For stable knockdown in difficult-to-transfect cells

    • CRISPR-Cas9: For complete knockout, targeting exon 2 or 3 of COX6C

  • Overexpression Systems:

    • Transient transfection with expression vectors containing strong promoters (CMV, EF1α)

    • Lentiviral or adenoviral vectors for stable expression or in vivo studies

    • Inducible expression systems (Tet-On/Off) for temporal control

  • Validation Methods:

    • Western blotting to confirm protein levels

    • qRT-PCR to measure mRNA expression (72-96% knockdown efficiency is typically achievable)

    • Functional assays to assess mitochondrial function after expression modification

How does COX6C expression change in muscle tissue damage models?

In rat muscle injury models, COX6C mRNA expression follows a distinct temporal pattern. Within 6 hours post-contusion, COX6C expression increases significantly compared to control groups. This elevated expression is followed by a dramatic decline during the 6-36 hour post-injury period. This temporal expression pattern correlates with wound staging and pathological features, making COX6C a potential biomarker for estimating wound stages. Similar evidence has demonstrated that COX6C expression positively correlates with skeletal muscle injury timing .

Conversely, in chronic obstructive pulmonary disease (COPD) patients experiencing acute exacerbations, COX6C expression decreases in muscle tissue. This reduction suggests that mitochondrial dysfunction may contribute to the pathophysiological changes observed in this condition, potentially offering a therapeutic target for treating muscle loss during acute disease exacerbations .

What is the relationship between COX6C and kidney diseases?

COX6C expression is significantly altered in various kidney disease states:

  • Chronic Kidney Disease (CKD): COX6C mRNA expression is notably downregulated in peripheral blood of patients undergoing peritoneal dialysis. Similarly, patients with CKD show lower COX6C expression levels, particularly when receiving renal replacement therapy. This decrease in expression may reflect the imbalance between oxidation and antioxidant defense mechanisms in CKD, with renal tissue damage in stage 5 CKD being associated with oxidative stress .

  • End-Stage Renal Disease (ESRD): COX6C and its upstream regulator PGC-1α (peroxisome proliferator-activated receptor-gamma coactivator 1 alpha) have emerged as potential prognostic indicators for ESRD/hemodialysis patients, particularly in relation to cardiovascular disease outcomes .

  • Diabetic Nephropathy (DN): Contrary to the pattern in CKD, COX6C mRNA is upregulated in glomerular cells of DN patients. Experimental models using telmisartan treatment in rats with DN show increased COX6C mRNA levels compared to controls, suggesting that the mitochondrial oxidative phosphorylation pathway may be implicated in DN pathogenesis .

How does COX6C function in response to viral infections?

During early-stage influenza virus infection, host cells upregulate COX6C expression by silencing miR-4276. This response occurs immediately after initial contact with the virus. The influenza virus infection induces changes in the host miRNA profile, potentially leading to cell death and tissue damage. COX6C appears to function as a key player in the apoptosis pathway during viral infection, potentially serving as an essential regulator during the early stages of influenza infection .

This relationship between COX6C and viral response mechanisms suggests that modulating COX6C function might offer a novel approach for interventions in viral infections, particularly during the critical early stages when the virus first interacts with host cells.

What role does COX6C amplification play in lung adenocarcinoma progression?

COX6C amplification plays a significant role in lung adenocarcinoma (LUAD) progression through several mechanisms:

  • Genomic Amplification: High-throughput sequencing and nucleic acid flight mass spectrometry analyses reveal that the 8q22.1-22.2 region, where COX6C is located, is frequently amplified in early-stage LUAD tissues. This amplification drives enhanced COX6C expression .

  • Expression Correlation: TCGA data analysis shows a positive correlation between COX6C copy number and its expression levels in LUAD. Both protein and mRNA levels of COX6C are significantly upregulated in LUAD compared to normal lung tissues .

  • Functional Impact: Knockdown experiments demonstrate that COX6C suppression significantly inhibits cell proliferation, induces S-G2/M cell cycle arrest, causes mitosis deficiency, and promotes apoptosis in LUAD cells .

  • Molecular Mechanism: COX6C depletion leads to:

    • Deficiencies in mitochondrial fusion

    • Impairment of oxidative phosphorylation

    • ROS accumulation

    • AMPK pathway activation

    • Abnormalities in spindle formation and chromosome segregation

    • Activation of spindle assembly checkpoints

    • Mitotic arrest and cellular apoptosis

These findings suggest that copy amplification-mediated COX6C upregulation may serve as a potential biomarker for prognosis and a target for therapy in LUAD patients.

How do COX6C expression patterns differ across various cancer types?

COX6C expression exhibits distinct patterns across different cancer types:

  • Prostate Cancer: COX6C mRNA is upregulated in clinical prostate tumor tissues. As a component of the mitochondrial inner respiratory chain, elevated COX6C levels support the high oxidative respiration rates required for malignant tumor growth. This upregulation has meaningful clinical significance for prostate cancer diagnosis .

  • Lung Adenocarcinoma: COX6C is frequently amplified and upregulated in LUAD tissues, with both protein and mRNA levels significantly increased compared to normal lung tissues. This upregulation correlates with copy number alterations and contributes to cancer cell proliferation .

  • Retroperitoneal Lipoma: COX6C translocations occur at the ETO8q chromosome breakpoint (8q22) in retroperitoneal lipoma tissue cells. Karyotype analysis of these translocations can aid in the classification of adipose tumor tissues .

  • Follicular Thyroid Cancer: DERL/COX6C translocations have been identified in thyroid cancer cell tissues, potentially serving as a diagnostic marker for follicular thyroid cancer .

  • Esophageal Cancer: COX6C is significantly upregulated during the early stages of exposure to different doses of ionizing radiation, providing insights into the molecular basis of radiotherapy responses .

  • Chronic Lymphocytic Leukemia (CLL): Recombinant IL-24 stimulates COX6C expression after 36 hours, while COX6C transcription involved in DNA replication and metabolism is inhibited within 6 hours .

What are the implications of targeting COX6C in cancer therapy development?

Targeting COX6C in cancer therapy development offers several promising strategies:

  • Biomarker Potential: Copy amplification-mediated COX6C upregulation serves as a prospective biomarker for cancer prognosis, particularly in lung adenocarcinoma patients .

  • Therapeutic Target: The critical role of COX6C in cancer cell proliferation and survival makes it an attractive target for therapeutic intervention. Specifically:

    • In lung adenocarcinoma, COX6C inhibition leads to cell cycle arrest and apoptosis

    • In prostate cancer, COX6C supports the high oxidative respiration rates necessary for tumor growth

  • Combination Therapy Approaches: COX6C inhibition could sensitize cancer cells to:

    • Conventional chemotherapeutics by disrupting energy metabolism

    • Radiation therapy, as suggested by COX6C's involvement in radiation response in esophageal cancer

    • Targeted therapies affecting mitochondrial function or cell cycle regulation

  • Stemness Regulation: COX6C knockdown effectively inhibits sphere formation and reduces expression of stemness-related markers including SOX2, OCT4, and Nanog. This suggests that targeting COX6C could potentially inhibit cancer stem cell populations, which are often responsible for therapy resistance and recurrence .

  • Delivery Challenges: Developing effective COX6C-targeting therapies requires addressing:

    • Selective delivery to cancer cells to minimize effects on normal tissues

    • Penetration of the mitochondrial membrane to reach the site of action

    • Potential compensatory mechanisms that may arise in response to COX6C inhibition

What cell-based assays best demonstrate COX6C's impact on cellular function?

Multiple cell-based assays effectively demonstrate COX6C's impact on cellular function:

  • Proliferation Assays:

    • Cell counting chamber method: Seed cells (7000 cells/well in 24-well plates or 3000 cells/well in 48-well plates) and count at defined intervals

    • Colony formation assay: Demonstrates long-term effects of COX6C manipulation on cell growth and survival capacity

  • Cell Cycle Analysis:

    • Flow cytometry with propidium iodide staining: Reveals COX6C knockdown-induced S-G2/M phase accumulation and polyploidy

    • Western blotting for cell cycle regulators: COX6C knockdown increases expression of cyclins B1, A2, D1, and CDK2

  • Apoptosis Detection:

    • Annexin V/7-AAD staining: Quantifies early and late apoptotic cells following COX6C manipulation

    • Flow cytometric analysis: Determines percentage of annexin V-positive cells

  • Mitochondrial Function Assays:

    • JC-1 staining: Measures mitochondrial membrane potential (MMP); cells are incubated with JC-1 solution for 20 min at 37°C

    • Confocal microscopy: Captures red/green fluorescence ratio representing MMP

  • ROS Detection:

    • H2DCFDA staining: Cells are incubated with 10 μM H2DCFDA for 20 min at 37°C

    • Flow cytometry: Quantifies relative fluorescence intensity as ratio of experimental to control groups

How can researchers distinguish between direct and indirect effects of COX6C manipulation?

Distinguishing between direct and indirect effects of COX6C manipulation requires a systematic approach:

  • Temporal Analysis:

    • Time-course experiments to establish the sequence of events following COX6C manipulation

    • Short-term effects (0-6 hours) more likely represent direct consequences

    • Long-term effects (24+ hours) may include secondary adaptations or compensatory mechanisms

  • Rescue Experiments:

    • Re-expression of wild-type COX6C in knockdown models should reverse direct effects

    • Domain-specific mutants can help identify which protein regions mediate specific functions

    • Use of non-degradable COX6C variants resistant to siRNA targeting

  • Pathway Inhibitors:

    • Specific inhibitors of downstream pathways (e.g., AMPK inhibitors) to block indirect effects

    • If inhibition prevents certain phenotypes, those effects are likely indirect and pathway-dependent

  • Protein-Protein Interaction Studies:

    • Co-immunoprecipitation to identify direct binding partners

    • Proximity ligation assays to confirm interactions in intact cells

    • FRET/BRET approaches to measure real-time interactions

  • Subcellular Fractionation:

    • Isolate mitochondria to determine direct effects on respiratory chain complexes

    • Compare effects in mitochondrial versus other cellular compartments

What are the critical controls for COX6C overexpression and knockdown experiments?

For robust COX6C manipulation experiments, the following controls are critical:

  • For Knockdown Experiments:

    • Negative Controls:

      • Non-targeting siRNA/shRNA with similar GC content to experimental siRNA

      • Empty vector controls for CRISPR-Cas9 systems

      • Multiple independent siRNA sequences targeting different regions of COX6C to confirm specificity

    • Validation Controls:

      • Western blot and qRT-PCR to confirm knockdown efficiency (>70% reduction recommended)

      • Rescue experiments with overexpression of siRNA-resistant COX6C

      • Dose-response experiments with varying levels of knockdown

  • For Overexpression Experiments:

    • Vector Controls:

      • Empty vector expressing the same tag (if applicable)

      • Irrelevant protein expression at similar levels

      • Untagged COX6C to control for tag-specific effects

    • Expression Level Controls:

      • Titration of expression plasmid to achieve physiologically relevant levels

      • Comparison with endogenous COX6C expression in relevant tissues

      • Inducible expression systems to control timing and magnitude

  • Cellular Context Controls:

    • Multiple cell lines to test consistency across different genetic backgrounds

    • Primary cells where possible to validate findings from cell lines

    • Comparison of effects in normal versus cancer cells

  • Functional Controls:

    • Mitochondrial membrane potential (MMP) assay as a surrogate for COX6C function

    • Oxygen consumption rate measurements

    • ATP production assays

Data Interpretation and Research Challenges

Translating in vitro COX6C findings to in vivo models involves addressing several critical considerations:

  • Model Selection and Relevance:

    • Choose animal models that recapitulate human disease features

    • Consider species differences in COX6C function and regulation

    • Evaluate tissue-specific expression patterns in target organisms

  • Delivery Systems for Manipulation:

    • Viral vectors (AAV, lentivirus) for tissue-specific expression or knockdown

    • CRISPR-Cas9 systems for genome editing in specific tissues

    • For recombinant Pongo pygmaeus COX6C, consider humanized models or evaluate cross-species functionality

  • Dosage and Duration Considerations:

    • Titrate interventions to achieve physiologically relevant levels

    • Account for differences in protein half-life and turnover rates between in vitro and in vivo settings

    • Design time-course studies to capture both acute and chronic effects

  • Validation Strategies:

    • Confirm target engagement using tissue samples

    • Validate functional consequences through relevant physiological measurements

    • Use multiple independent methodologies to corroborate findings

  • Translational Challenges:

    • The challenge of creating xenograft models with COX6C knockdown cells, as multiple attempts were unsuccessful in nude mice

    • Stem cell properties may be affected by COX6C manipulation, as evidenced by reduced sphere formation and decreased expression of stemness markers (SOX2, OCT4, Nanog) in COX6C knockdown cells

How can researchers address potential compensatory mechanisms when studying COX6C function?

Addressing compensatory mechanisms in COX6C research requires sophisticated experimental approaches:

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.