COX5A (Cytochrome c oxidase subunit 5A) is a critical subunit of the cytochrome c oxidase (COX) complex, which functions as the terminal enzyme in the mitochondrial electron transport chain. It contains heme A and plays an essential role in cellular respiration by catalyzing the final step of electron transfer to molecular oxygen. This process is fundamental for ATP production as it drives the creation of a proton gradient across the inner mitochondrial membrane, ultimately leading to ATP synthesis .
COX5A expression is dynamically regulated in response to oxygen availability - increasing during oxygen abundance to enhance aerobic respiration and decreasing under hypoxic conditions to conserve oxygen, highlighting its critical role in maintaining cellular energy homeostasis . Recent research has demonstrated that COX5A is involved in protecting against doxorubicin-induced cardiotoxicity and may play roles in memory function and cognitive processes .
COX5A serves as one of the two subunits of COX5 within the cytochrome c oxidase complex (Complex IV), which catalyzes the final step of the electron transport chain. Specifically, COX5A contains heme A and facilitates efficient electron transfer from cytochrome c to molecular oxygen, resulting in the formation of water. During this process, protons are pumped across the inner mitochondrial membrane, contributing to the electrochemical gradient necessary for ATP synthesis .
The protein has a calculated molecular weight of 17 kDa based on its 150 amino acid sequence, though it is typically observed at 13 kDa in experimental conditions . This discrepancy is likely due to post-translational modifications and processing that occur after the protein is synthesized. COX5A's activity directly influences the efficiency of oxidative phosphorylation and consequently affects cellular energy availability and mitochondrial function.
Research has established several important connections between COX5A dysfunction and pathological conditions:
Cardiovascular disease: COX5A expression is significantly decreased in patients with end-stage dilated cardiomyopathy (DCM). Furthermore, COX5A has been shown to protect against doxorubicin-induced cardiotoxicity by alleviating oxidative stress, mitochondrial dysfunction, and cardiomyocyte apoptosis .
Neurodegenerative conditions: COX5A plays a vital role in memory function, with evidence indicating that overexpression of COX5A can improve spatial recognition memory and hippocampal synaptic plasticity in aging mouse models. This suggests potential implications for age-related cognitive decline and neurodegenerative disorders .
Mitochondrial disorders: As a key component of the electron transport chain, dysfunction in COX5A can contribute to primary mitochondrial disorders characterized by impaired energy production and increased oxidative stress .
The clinical significance of COX5A extends to its potential as a therapeutic target, particularly in conditions involving mitochondrial dysfunction, oxidative damage, and energy metabolism impairment .
When selecting a COX5A antibody, researchers should consider several critical factors to ensure optimal experimental outcomes:
Intended application: Different COX5A antibodies perform optimally in specific applications. For instance, the COX5A antibody (11448-1-AP) has been validated for Western Blot (1:500-1:1000 dilution), Immunohistochemistry (1:20-1:200 dilution), and Immunofluorescence (1:20-1:200 dilution) . Similarly, the COX5A Antibody (A-5) is suitable for western blotting, immunoprecipitation, immunofluorescence, immunohistochemistry with paraffin-embedded sections, and ELISA .
Species reactivity: Verify the antibody's species reactivity to ensure compatibility with your experimental model. The COX5A antibody (11448-1-AP) has confirmed reactivity with human, mouse, and rat samples, with cited reactivity extending to pig, monkey, zebrafish, and chimpanzee models .
Clonality: Consider whether a monoclonal (like A-5, which is a mouse monoclonal IgG1 kappa light chain antibody ) or polyclonal antibody (like 11448-1-AP ) is more suitable for your application.
Validation data: Review published studies that have successfully used the antibody in similar applications. For example, the COX5A antibody (11448-1-AP) has been cited in 18 publications for Western Blot and 3 publications for IHC applications .
Conjugation requirements: Determine if you need a conjugated antibody for specific detection methods. Some COX5A antibodies are available in various conjugated forms, including agarose, HRP, PE, FITC, and multiple Alexa Fluor® conjugates .
It is highly recommended to titrate the antibody in your specific experimental system to determine the optimal working concentration, as antibody performance can vary based on sample type, preparation method, and detection system .
For optimal Western Blot results with COX5A antibodies, follow these detailed protocol recommendations:
Sample Preparation:
Extract proteins from tissue or cells using RIPA buffer supplemented with protease inhibitors.
For mitochondrial enrichment (recommended for enhanced detection), perform subcellular fractionation using a mitochondrial isolation kit.
Quantify protein concentration using BCA or Bradford assay.
Western Blot Protocol:
Load 20-30 μg of protein per lane on a 12-15% SDS-PAGE gel (COX5A is a relatively small protein).
Transfer proteins to a nitrocellulose membrane (as used in Xiyang et al. study) .
Block the membrane with 5% non-fat milk in TBST for 1 hour at room temperature.
Incubate with primary COX5A antibody:
Incubate at 4°C for 24 hours (as described in the Xiyang et al. protocol) or overnight.
Incubate with appropriate HRP-conjugated secondary antibody for 1 hour at room temperature.
Develop using enhanced chemiluminescence.
Expected Results:
COX5A should be detected at approximately 13 kDa (observed molecular weight), although its calculated molecular weight is 17 kDa .
Positive controls should include samples known to express COX5A, such as mouse brain tissue, human liver tissue, rat brain tissue, L02 cells, or HeLa cells .
Troubleshooting Tips:
If signal is weak, consider mitochondrial enrichment during sample preparation or increasing antibody concentration.
For high background, increase washing steps or decrease antibody concentration.
For multiple bands, confirm specificity using positive controls and consider using a knockout/knockdown validation approach.
This protocol has been successfully employed in research studying COX5A's role in doxorubicin-induced cardiotoxicity and memory function .
For successful immunohistochemistry with COX5A antibodies, implement these research-validated practices:
Tissue Preparation and Antigen Retrieval:
Fix tissues in 4% paraformaldehyde and embed in paraffin following standard procedures.
Section tissues at 4-6 μm thickness.
For antigen retrieval with COX5A antibody (11448-1-AP):
Staining Protocol:
Deparaffinize and rehydrate sections.
Perform antigen retrieval as specified above.
Block endogenous peroxidase activity with 3% H₂O₂ for 10 minutes.
Block non-specific binding with 10% normal serum in PBS for 30 minutes.
Incubate with primary COX5A antibody:
Wash with PBS (3 × 5 minutes).
Apply appropriate secondary antibody and develop according to your detection system (DAB recommended).
Counterstain, dehydrate, and mount.
Validation Controls:
Positive tissue controls: Human gliomas tissue, human colon tissue, and human lung cancer tissue have been validated for positive IHC detection with COX5A antibody (11448-1-AP) .
Negative controls: Perform staining without primary antibody.
Specificity control: Consider using siRNA knockdown tissues if available.
Advanced Considerations:
For co-localization studies, double immunofluorescence staining can be performed with other mitochondrial markers.
For quantitative analysis, consider using digital image analysis software to measure staining intensity.
When studying tissues with high autofluorescence (like brain or heart), use Sudan Black B treatment to reduce background.
These protocols have been effectively applied in research investigating COX5A's role in cardiac and neurological tissues, producing reliable and reproducible results .
Researchers have successfully employed several approaches to modulate COX5A expression in experimental models:
Overexpression Systems:
AAV9-mediated overexpression: For in vivo studies, particularly in cardiac tissue, adeno-associated virus serum type 9 (AAV9) has been effectively used to upregulate COX5A expression. This approach was successfully employed in C57BL/6J mice to investigate COX5A's role in DOX-induced cardiomyopathy .
Lentiviral vectors for in vitro studies: For cell culture experiments, COX5A overexpression has been achieved using lentiviral systems. Specifically, the pReceiver-Lv130 plasmid vector fused to Enhanced Green Fluorescent Protein (EGFP) has been used, with COX5A cDNA inserted between NspV (5′-GCTTGGAAGGAGTTCGAACCATG-3′) and XhoI (5′-ACGCCGGCGTGAGCTCCAT-3′) restriction sites .
Knockdown/Silencing Systems:
shRNA approaches: For COX5A silencing, shRNA sequences targeting COX5A mRNA (NCBI Accession Number: GeneID: 12858) have been designed. These typically include:
A sense strand (e.g., 5′-CCGACAACCACTACCTGA-3′)
A hairpin loop (5′-TCAAGAG-3′)
An anti-sense strand (e.g., 5′-CGTGAAGA-ATGTGCGAGAC-3′)
These sequences have been inserted between BamHI and EcoRI restriction sites in psiHIV-U6 (HIV-based) plasmid vectors .
Transgenic Animal Models:
Transgenic mice with systemic COX5A overexpression (achieving approximately 51% increase in expression) have been established to study the role of COX5A in memory function and hippocampal synaptic plasticity .
Experimental Validation:
When implementing these systems, it's crucial to confirm successful modulation of COX5A expression through:
Western blot analysis using validated COX5A antibodies
qRT-PCR for mRNA expression levels
Functional assays to confirm biological effects (e.g., mitochondrial respiration)
These genetic modulation approaches provide powerful tools for investigating COX5A's functional roles in various biological processes and potential therapeutic applications.
To comprehensively evaluate mitochondrial function in relation to COX5A activity, researchers should employ multiple complementary techniques:
1. Cytochrome c Oxidase (COX) Activity Assays:
Direct measurement of COX enzymatic activity provides the most relevant assessment of COX5A function. This can be performed using:
Spectrophotometric assays measuring the oxidation rate of reduced cytochrome c
Polarographic methods using oxygen electrodes
Histochemical staining for COX activity in tissue sections
2. ATP Production Measurement:
Since COX5A is integral to oxidative phosphorylation and energy production, ATP levels serve as an important functional readout:
Luminescence-based ATP assays
HPLC-based methods for more precise quantification
As demonstrated in COX5A overexpression studies, ATP content was significantly improved in DOX-treated mice with COX5A upregulation compared to controls .
3. Mitochondrial Respiration Analysis:
Oxygen consumption rate (OCR) measurements using platforms like Seahorse XF Analyzer provide detailed insights into:
Basal respiration
Maximal respiration
Spare respiratory capacity
ATP-linked respiration
Research has shown that COX5A overexpression restored DOX-impaired maximal respiration and spare capacity in H9c2 cells .
4. Reactive Oxygen Species (ROS) Quantification:
As COX5A modulates oxidative stress, ROS assessment is critical:
Dihydroethidium (DHE) staining for superoxide detection
DCFH-DA staining for general ROS detection
4-HNE immunohistochemistry for lipid peroxidation assessment
Studies have demonstrated that COX5A overexpression markedly inhibited DOX-induced oxidative stress as measured by these techniques .
5. Mitochondrial Morphology Analysis:
Transmission electron microscopy to assess ultrastructural changes
Confocal microscopy with mitochondrial-specific dyes
Immunofluorescence using antibodies against mitochondrial markers
6. Mitochondrial Membrane Potential:
JC-1 or TMRM dyes to assess mitochondrial membrane potential
Flow cytometry or fluorescence microscopy for quantification
These methodologies should be applied in combination to provide a comprehensive assessment of how COX5A modulation affects mitochondrial function across multiple parameters.
Researchers investigating COX5A across different tissue types and experimental models should consider these critical tissue-specific and model-dependent factors:
Tissue-Specific Considerations:
Cardiac Tissue:
Neural Tissue:
Liver and Highly Metabolic Tissues:
Experimental Model Considerations:
Cell Culture Models:
Mouse Models:
Disease Models:
Cross-Species Considerations:
The COX5A antibody (11448-1-AP) shows confirmed reactivity with human, mouse, and rat samples, with further cited reactivity in pig, monkey, zebrafish, and chimpanzee , allowing for comparative studies across species.
By accounting for these tissue-specific and model-dependent considerations, researchers can design more robust studies and facilitate more accurate interpretation of results across different experimental systems.
When confronted with conflicting results regarding COX5A function across experimental systems, researchers should implement a systematic approach to interpretation:
1. Evaluate Methodological Differences:
Antibody selection: Different antibodies may detect distinct epitopes or isoforms of COX5A. The molecular weight discrepancy between calculated (17 kDa) and observed (13 kDa) COX5A suggests post-translational modifications that may affect detection .
Expression systems: Overexpression levels achieved through different vectors (AAV9 vs. lentiviral) may yield variable biological responses. The transgenic model showing 51% increase in COX5A expression demonstrated improvement in cognitive function , while the percent increase necessary for cardioprotection may differ.
Assay sensitivity: Different techniques for assessing mitochondrial function have varying sensitivities and measure distinct aspects of COX5A activity.
2. Consider Context-Dependent Functions:
Tissue-specific roles: COX5A function in cardiac tissue (protection against DOX-induced apoptosis) may differ from its role in neural tissue (improvement of synaptic plasticity) .
Pathological context: COX5A's protective effects may be evident only under specific stress conditions (e.g., doxorubicin treatment, aging) rather than in normal physiological states.
Developmental stage: COX5A function may vary between developing, adult, and aging systems, as suggested by the differential expression between 2-month and 12-month-aged SAMP8 mice .
3. Analyze Pathway Interactions:
Signaling mechanisms: COX5A operates through different signaling pathways depending on the tissue context:
Compensatory mechanisms: Contradictory results may reflect activation of different compensatory mechanisms across systems.
4. Resolution Strategies:
Direct comparison experiments: Design studies that directly compare different systems under identical conditions.
Dose-response relationships: Establish whether differences reflect threshold effects rather than fundamental functional differences.
Genetic background considerations: Account for strain-specific or genetic background effects that may influence COX5A function.
Time-course analyses: Determine whether apparent contradictions reflect temporal differences in COX5A action.
By systematically addressing these factors, researchers can better contextualize seemingly conflicting data and develop more nuanced models of COX5A function across biological systems.
COX5A exerts its protective effects through distinct but potentially interconnected signaling pathways in different tissue types:
COX5A's cardioprotective effects against doxorubicin-induced injury are primarily mediated through the PI3K/Akt signaling cascade:
Akt Phosphorylation: COX5A overexpression significantly increases the phosphorylation of Akt at both Thr308 and Ser473 residues in DOX-treated cardiac tissue and H9c2 cells .
Mechanistic Verification: The causative role of this pathway was confirmed through pharmacological inhibition:
Functional Outcomes: LY294002 at 10μM concentration decreased ATP production, maximal respiration, and spare capacity in COX5A-overexpressing cells to levels comparable to control cells, confirming the PI3K/Akt pathway's essential role in mediating COX5A's effects on mitochondrial energetics .
In contrast, COX5A's neuroprotective effects are primarily mediated through the BDNF/ERK1/2 signaling axis:
BDNF Upregulation: COX5A overexpression results in increased BDNF levels in hippocampal tissue .
ERK1/2 Activation: Following BDNF upregulation, enhanced phosphorylation of ERK1/2 occurs, activating this critical signaling pathway involved in synaptic plasticity and memory function .
Functional Verification: The PD98059 ERK1/2 inhibitor was used to establish the necessity of this pathway for COX5A's effects on neuronal development and function .
Morphological Outcomes: COX5A overexpression led to recovery of hippocampal CA1 dendrites, which was dependent on BDNF/ERK1/2 signaling .
While these pathways appear distinct, they may exhibit cross-talk and convergence:
Common Downstream Effects: Both pathways ultimately influence:
Mitochondrial function
Oxidative stress response
Cell survival mechanisms
Potential Integrative Model: COX5A may serve as a mitochondrial sensor that triggers tissue-specific protective signaling based on energy demands and stress conditions.
This tissue-specific signaling diversity highlights the importance of contextual factors when designing therapeutic strategies targeting COX5A for different pathological conditions.
Post-translational modifications (PTMs) significantly impact both COX5A antibody detection and functional activity, presenting important considerations for research interpretation:
Effects on Antibody Detection:
Molecular Weight Discrepancies: The COX5A protein has a calculated molecular weight of 17 kDa based on its 150 amino acid sequence, yet is typically observed at 13 kDa in experimental conditions . This 4 kDa difference likely reflects post-translational processing, including:
Removal of mitochondrial targeting sequences
Proteolytic cleavage
Other modifications affecting electrophoretic mobility
Epitope Masking: PTMs can mask antibody epitopes, potentially leading to:
False negative results
Reduced signal intensity
Differential detection across tissues with varying PTM profiles
Antibody Selection Implications: Researchers should consider using antibodies raised against different epitopes or modified forms when discrepancies are observed. The polyclonal antibody (11448-1-AP) may detect multiple epitopes, while the monoclonal antibody (A-5) recognizes a specific epitope that could be affected by certain PTMs.
Impact on Functional Activity:
Regulation of Enzyme Activity: PTMs directly modulate COX5A's functional activity within the cytochrome c oxidase complex:
Phosphorylation can alter the efficiency of electron transfer
Acetylation may affect protein-protein interactions within the COX complex
Oxidative modifications can impair function under conditions of oxidative stress
Subcellular Localization: PTMs influence COX5A's mitochondrial import and retention, directly affecting its availability for incorporation into functional COX complexes.
Protein Stability and Turnover: Certain modifications (e.g., ubiquitination) regulate COX5A's half-life and degradation, impacting steady-state protein levels.
Methodological Considerations:
Detection of Modified Forms:
Two-dimensional gel electrophoresis can separate differently modified COX5A isoforms
Mass spectrometry approaches can identify specific modification sites
Phospho-specific antibodies may be required for detecting specific modified states
Functional Assessment:
Activity assays may need to be interpreted in light of the PTM status
In vitro modification systems can help establish causative relationships
Physiological Relevance:
The oxidative environment of mitochondria makes COX5A particularly susceptible to oxidative modifications
These modifications may represent both regulatory mechanisms and pathological changes
Understanding the impact of PTMs on COX5A is essential for accurate interpretation of experimental results and may reveal novel regulatory mechanisms that could be targeted therapeutically.
COX5A modulation shows promising therapeutic potential across both cardiovascular and neurodegenerative conditions, with distinct applications in each field:
Cardiovascular Therapeutic Applications:
Protection Against Chemotherapy-Induced Cardiotoxicity:
COX5A overexpression effectively protected against doxorubicin-induced oxidative stress, mitochondrial dysfunction, and cardiomyocyte apoptosis in both in vivo and in vitro models .
This suggests potential for COX5A-targeted therapies as cardioprotective agents during cancer treatment with anthracyclines.
Dilated Cardiomyopathy Treatment:
The dramatic decrease in COX5A expression observed in patients with end-stage dilated cardiomyopathy indicates that restoring COX5A levels could potentially slow disease progression .
Gene therapy approaches using cardiac-specific delivery systems (such as AAV9) could provide targeted COX5A upregulation in affected tissues.
Heart Failure Management:
By improving mitochondrial function and energy metabolism, COX5A enhancement could address the bioenergetic deficit commonly observed in heart failure.
Combined approaches targeting COX5A and the PI3K/Akt pathway may provide synergistic benefits.
Neurodegenerative Therapeutic Applications:
Age-Related Cognitive Decline:
Alzheimer's Disease Intervention:
Neuroprotective Strategies:
COX5A-targeted therapies could potentially address the mitochondrial dysfunction observed in various neurodegenerative conditions.
Combination approaches targeting both COX5A and downstream signaling pathways may offer enhanced neuroprotection.
Delivery Systems and Therapeutic Strategies:
Gene Therapy Approaches:
Small Molecule Modulators:
Compounds that enhance COX5A stability or function
Molecules targeting the PI3K/Akt or BDNF/ERK1/2 pathways downstream of COX5A
Combined Therapeutic Approaches:
Antioxidant therapy with COX5A modulation may provide synergistic benefits
Metabolic support agents combined with COX5A enhancement
These emerging therapeutic directions highlight COX5A as a promising target for addressing both cardiovascular and neurodegenerative conditions through tissue-specific approaches tailored to the relevant signaling pathways.
Researchers face several significant technical challenges when developing and utilizing experimental models to study COX5A function:
1. Achieving Physiologically Relevant Expression Levels:
Challenge: Both insufficient and excessive COX5A expression can yield misleading results. Studies have shown that a 51% increase in COX5A expression produced beneficial effects in cognitive models , but the optimal expression level may vary by tissue and context.
Solution Approaches:
Inducible expression systems with titratable control
Knock-in models preserving endogenous regulation
Careful quantification of expression relative to physiological levels
2. Tissue-Specific Targeting:
Challenge: COX5A functions differently across tissues, operating through the PI3K/Akt pathway in cardiac tissue and the BDNF/ERK1/2 pathway in neural tissue .
Solution Approaches:
3. Isolating COX5A-Specific Effects:
Challenge: As part of the cytochrome c oxidase complex, isolating COX5A-specific effects from general effects on mitochondrial function can be difficult.
Solution Approaches:
Structure-function studies with site-directed mutagenesis
Comparison with models targeting other COX subunits
Rescue experiments in knockdown/knockout systems
4. Temporal Regulation:
Challenge: COX5A's role may vary during development, adulthood, and aging, as suggested by expression differences between 2-month and 12-month-aged SAMP8 mice .
Solution Approaches:
Temporally controlled expression systems
Age-stratified experimental designs
Longitudinal studies with multiple timepoints
5. Mitochondrial-Nuclear Coordination:
Challenge: As a nuclear-encoded mitochondrial protein, COX5A function depends on proper coordination between nuclear transcription and mitochondrial integration.
Solution Approaches:
Dual assessment of nuclear expression and mitochondrial localization
Monitoring of import machinery components
Evaluation of mitochondrial-nuclear communication pathways
6. Experimental Variability in Functional Assessments:
Challenge: Mitochondrial function assessments (e.g., oxygen consumption, ATP production) show substantial technical variability.
Solution Approaches:
Standardized protocols with appropriate controls
Multiple complementary functional assays
Sufficient biological and technical replicates
Normalization to mitochondrial content
7. Translating Between In Vitro and In Vivo Systems:
Challenge: Findings from cell models (e.g., H9c2 cells, cultured neurons) may not fully translate to the complexity of intact organisms.
Solution Approaches:
Validation across multiple model systems
Organoid or ex vivo tissue slice cultures as intermediate models
Careful selection of cell lines with appropriate metabolic profiles
Addressing these technical challenges is essential for developing robust experimental systems that can accurately elucidate COX5A's complex functions and potential therapeutic applications.
The integration of multi-omics approaches provides a comprehensive framework for understanding COX5A's multifaceted roles in cellular homeostasis:
1. Genomics and Transcriptomics Integration:
Approach: Combining genomic profiling with transcriptomic analysis has revealed differential expression patterns of COX5A across tissues and disease states.
Key Insights: Genomic profiling in the hippocampus of Senescence-Accelerated Mouse-prone 8 (SAMP8) mice demonstrated differential COX5A expression between 12-month-aged and 2-month-aged mice, highlighting age-dependent regulation .
Future Directions: Single-cell transcriptomics could further reveal cell-type-specific expression patterns within heterogeneous tissues like brain and heart.
2. Proteomics and Post-Translational Modifications:
Approach: Mass spectrometry-based proteomics coupled with enrichment for specific modifications.
Key Insights: The discrepancy between COX5A's calculated molecular weight (17 kDa) and observed weight (13 kDa) suggests important post-translational processing that could be mapped through proteomic approaches.
Application: Phosphoproteomics could characterize how the PI3K/Akt pathway directly modifies COX5A or associated proteins in cardiac tissue , while global proteomics can identify co-regulated protein networks.
3. Metabolomics and Bioenergetics:
Approach: Combining metabolomic profiling with functional bioenergetic assays.
Key Insights: COX5A overexpression improves ATP production and mitochondrial respiration in DOX-treated mice , but the broader metabolic consequences remain to be fully characterized.
Integration Potential: Correlation of COX5A expression levels with metabolite profiles could reveal unexpected roles in metabolic pathway regulation beyond oxidative phosphorylation.
4. Interactomics:
Approach: Protein-protein interaction mapping using proximity labeling or co-immunoprecipitation coupled with mass spectrometry.
Key Insights: Beyond its structural role in the COX complex, COX5A may interact with signaling proteins, potentially explaining its influence on the BDNF/ERK1/2 pathway in neural tissue and PI3K/Akt pathway in cardiac tissue .
Future Applications: Systematic mapping of the COX5A interactome across different tissues could reveal context-specific interaction partners driving tissue-specific functions.
5. Integrative Analysis Frameworks:
Approach: Machine learning and network analysis to integrate multi-omics datasets.
Potential Applications:
Identification of novel regulatory mechanisms controlling COX5A expression
Discovery of biomarkers that predict response to COX5A-targeting therapies
Construction of predictive models for cellular responses to COX5A modulation
6. Clinical Translation:
Approach: Integration of multi-omics data from patient samples with experimental models.
Current Evidence: Decreased COX5A expression has been observed in patients with end-stage dilated cardiomyopathy , but comprehensive multi-omics profiling could reveal additional disease associations.
Future Impact: Patient stratification based on integrated molecular profiles could identify individuals most likely to benefit from COX5A-targeting therapeutic approaches.
The integration of these multi-omics approaches provides a systems-level understanding of how COX5A contributes to cellular homeostasis across different tissues and pathological contexts, potentially revealing novel therapeutic targets and biomarkers for mitochondrial dysfunction-related diseases.
Cytochrome c oxidase subunit Va (COX Va) is a critical component of the mitochondrial respiratory chain, specifically involved in the electron transport chain. This subunit is part of the larger cytochrome c oxidase complex (Complex IV), which plays a pivotal role in cellular respiration by facilitating the transfer of electrons from cytochrome c to molecular oxygen, ultimately contributing to the generation of ATP through oxidative phosphorylation.
COX Va is essential for the proper functioning of the cytochrome c oxidase complex. It is involved in maintaining the structural integrity and catalytic activity of the enzyme. The subunit is encoded by nuclear DNA and is imported into the mitochondria, where it integrates into the inner mitochondrial membrane. The proper assembly and function of COX Va are crucial for efficient energy production in cells.
Research has shown that COX Va is implicated in various diseases, particularly those related to mitochondrial dysfunction. For instance, alterations in COX Va expression have been associated with neurodegenerative diseases such as Alzheimer’s disease. Studies have demonstrated that COX Va plays a role in memory impairment associated with brain aging via the BDNF/ERK1/2 signaling pathway . Additionally, COX Va has been identified as a potential biomarker for the aggressiveness of non-small cell lung cancer (NSCLC), with its expression correlating with migration, invasion, and metastasis of cancer cells .
The mouse anti-human COX Va antibody is a monoclonal antibody specifically designed to target the human COX Va protein. This antibody is widely used in research to study the expression and function of COX Va in various biological contexts. It is particularly useful in techniques such as western blotting, immunohistochemistry, and immunofluorescence, allowing researchers to visualize and quantify COX Va in tissue samples and cell lines.