COPS7A Human, encoded by the COPS7A gene (OMIM: 616009), is a subunit of the COP9 signalosome complex (CSN), a conserved multiprotein complex critical for regulating ubiquitin-dependent processes . The CSN complex mediates deneddylation of cullin subunits in SCF-type E3 ligases, modulating their activity in protein ubiquitination and degradation . COPS7A is essential for maintaining the structural integrity and catalytic function of the CSN, influencing cellular processes such as DNA repair, cell cycle regulation, and immune responses .
COPS7A is ubiquitously expressed across human tissues, with notable presence in:
Neurological tissues: Hippocampus, amygdala, basal ganglia, cerebellum
Endocrine organs: Pituitary gland, adrenal gland, thyroid gland
The CSN complex, including COPS7A, regulates protein stability through interactions with cullin-RING ligases (CRLs) and kinases like CK2/PKD . It modulates substrates such as p53, JUN, and IκBα, influencing tumor suppression, apoptosis, and inflammation .
COPS7A is implicated in hepatocellular carcinoma (HCC) progression, as evidenced by:
Parameter | COPS7A | COPS5 | COPS7B | COPS9 |
---|---|---|---|---|
AUC (HCC diagnosis) | 0.900 | 0.971 | 0.952 | 0.929 |
HR (OS) | N/A | 2.118 | 2.453 | 2.311 |
COPS7A’s interaction network includes oncogenic factors such as cullins (CUL1/2/3/4) and transcription factors (p53, JUN) . Functional studies in HCC models highlight the CSN’s role in promoting proliferation and metastasis .
COPS7A interacts with core CSN subunits and regulatory partners, as mapped in STRING:
Partner | Interaction Score | Role in CSN |
---|---|---|
COPS3 | 0.999 | Structural scaffold |
COPS2 | 0.999 | Core component |
COPS5 | 0.999 | Protease subunit |
CUL4A | 0.945 | SCF-type E3 ligase target |
These interactions underscore COPS7A’s role in maintaining CSN stability and facilitating cullin deneddylation .
Cancer Metabolism: COPS7A upregulation in HCC correlates with immune cell infiltration (e.g., macrophages, T cells) and microsatellite instability .
Therapeutic Targeting: Knockdown of COPS7A in HCC cells reduces proliferation, suggesting CSN inhibition as a potential therapeutic strategy .
Biomarker Potential: Multivariate Cox analysis identifies COPS7A as part of a prognostic signature for HCC, though COPS5, COPS7B, and COPS9 show stronger independent predictive value .
COPS7A (also known as CSN7A) is a protein subunit of the COP9 signalosome complex encoded by the COPS7A gene in humans. The COP9 signalosome is a highly conserved protein complex involved in various cellular processes including protein degradation via the ubiquitin-proteasome pathway . COPS7A functions as part of this multi-protein complex that regulates cullin-RING ligase (CRL) activity through deneddylation. To verify COPS7A's incorporation into the COP9 signalosome, researchers typically employ co-immunoprecipitation assays using antibodies against other CSN subunits or flag-tagged COPS7A protein followed by western blotting analysis .
COPS7A undergoes multiple post-translational modifications (PTMs) at specific sites, including:
To investigate these modifications, researchers typically employ mass spectrometry-based approaches coupled with enrichment strategies specific to each PTM type. For phosphorylation studies, titanium dioxide (TiO2) or immobilized metal affinity chromatography (IMAC) enrichment followed by LC-MS/MS analysis is commonly used. For ubiquitination studies, affinity purification with ubiquitin-binding domains followed by MS analysis is the standard approach .
COPS7A appears to be expressed across all human tissues and cells investigated according to the Genevestigator expression data library . Expression analysis can be performed using RNA-seq data or protein quantification through western blotting. The Human Protein Atlas provides tissue-specific expression data through antibody-based profiling using immunohistochemistry across 44 normal tissue types . For laboratory verification of expression patterns, researchers typically employ qRT-PCR for mRNA quantification and western blotting with specific anti-COPS7A antibodies for protein detection. When studying expression in specific contexts, such as during cellular differentiation, time-course experiments combined with western blotting are effective methods to track expression changes .
COPS7A and COPS7B are paralogous proteins that function as alternative subunits in the COP9 signalosome complex. These variants coexist in human cells, including LiSa-2 preadipocytes, and appear to have both overlapping and distinct functions . While both are necessary for processes such as adipogenesis (as demonstrated by the retardation of adipogenesis when either is downregulated), they exhibit differential expression patterns during adipocyte differentiation. Specifically, endogenous COPS7B expression increases during adipogenesis while COPS7A levels remain constant . To study their distinct roles, researchers can employ variant-specific antibodies for detection and use siRNA-mediated knockdown approaches targeting each variant specifically .
The COPS7A and COPS7B variants appear to have distinct roles in adipogenic differentiation. Research shows that overexpression of COPS7B accelerates adipogenesis, while overexpression of COPS7A does not produce the same effect. Interestingly, downregulation of either COPS7A or COPS7B leads to retardation of adipogenesis, indicating both are necessary for the process .
To investigate these differential effects, researchers can:
Establish stable cell lines expressing Flag-tagged COPS7A or COPS7B (using vectors such as pCMV-3Tag-1a)
Perform transient knockdown using siRNA specific to either COPS7A or COPS7B
Assess adipogenesis through:
Western blot analysis of adipogenic markers (PPAR-γ, CHOP)
Oil Red O (ORO) staining to quantify lipid production
Microscopy to visualize lipid droplet formation
Experimental models for such studies include human preadipocyte cell lines (like LiSa-2) and mouse embryonic fibroblasts (MEFs) .
COPS7A and COPS7B appear to have different affinities for certain proteins, which may explain their distinct functional roles. Flag-pulldown experiments suggest that CSN7A and CSN7B variants bind differently to proteins such as β-TrCP and USP15 . To comprehensively investigate COPS7A-specific protein interactions, researchers should:
Perform immunoprecipitation using Flag-tagged COPS7A followed by mass spectrometry to identify binding partners
Compare interactomes between COPS7A and COPS7B using parallel IP-MS experiments
Validate key interactions using reciprocal co-immunoprecipitation and western blotting
Assess functional relevance of specific interactions through domain mapping and mutagenesis studies
Employ proximity labeling methods (BioID or APEX) to capture transient interactions in living cells
These approaches would help elucidate how COPS7A-specific protein interactions contribute to its unique functions within the COP9 signalosome complex .
Several disease-associated variants of COPS7A have been identified that affect post-translational modification sites:
Site | Variant | Disease Association |
---|---|---|
K206 | R206 | Pancreatic cancer |
T219 | I219, N219 | Colorectal cancer |
R243 | K243 | Bile duct cancer |
R265 | * | Uterine cancer, Cervical cancer |
To investigate the functional impact of these variants, researchers should consider:
Generating site-specific mutations in COPS7A expression constructs using site-directed mutagenesis
Expressing wild-type and mutant COPS7A in relevant cell models
Assessing effects on:
COP9 signalosome complex formation and stability using gel filtration or blue native PAGE
Deneddylating activity using cullins as substrates
Protein-protein interactions compared to wild-type COPS7A
Post-translational modification patterns at other sites
Cellular phenotypes relevant to the associated cancer type
This systematic approach would provide insights into how these variants contribute to pathogenesis in different cancer types .
COPS7A undergoes numerous post-translational modifications, but their precise roles in regulating CSN complex activity remain largely unexplored. To investigate these relationships, researchers should:
Generate phospho-mimetic and phospho-deficient mutants of key COPS7A phosphorylation sites (Y137, T219, S232, T240, T249, S255)
Create lysine-to-arginine mutants to prevent ubiquitination/acetylation at sites K20, K109, K199, K206, K217, K221, K256, K260, K269
Assess the impact of these mutations on:
CSN complex assembly using co-immunoprecipitation followed by western blotting
Deneddylating activity of the CSN complex using in vitro and cellular assays
Interaction with CRLs and other binding partners
Cellular localization using immunofluorescence microscopy
Stability and turnover rate of COPS7A using cycloheximide chase assays
Additionally, researchers should investigate the crosstalk between different modifications, as phosphorylation often regulates subsequent ubiquitination or acetylation events. Mass spectrometry approaches could reveal how modifications at one site affect the modification status at other sites .
Studying COPS7A within its native COP9 signalosome complex presents technical challenges. The most effective methodologies include:
Structural Analysis:
Cryo-electron microscopy to visualize COPS7A within the intact CSN complex
Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces
Functional Analysis:
CRISPR-Cas9 genome editing to create COPS7A knockout or endogenously tagged cell lines
Reconstitution assays with purified components to assess direct contributions to CSN activity
Interaction Analysis:
Proximity labeling techniques (BioID, APEX) to identify neighbors of COPS7A in living cells
Crosslinking mass spectrometry to capture transient interactions within the complex
Expression Analysis:
Single-cell RNA sequencing to identify cell types with unique COPS7A expression patterns
Fluorescence correlation spectroscopy to measure the stoichiometry of COPS7A in living cells
Purification Approaches:
The COP9 Signalosome Subunit 7A (COPS7A) is a crucial component of the COP9 signalosome (CSN) complex, which is an evolutionarily conserved multi-subunit protease. This complex plays a significant role in regulating the activity of the ubiquitin conjugation pathway, which is essential for various cellular processes, including protein degradation, cell cycle control, and signal transduction .
The COP9 signalosome complex is composed of several subunits, including CSN1 (COPS1), CSN2 (COPS2), CSN3 (COPS3), CSN4 (COPS4), CSN5 (COPS5), CSN6 (COPS6), CSN7A (COPS7A), and CSN8 (COPS8). The CSN7A subunit contains a PCI (Proteasome CSN9 initiation factor 3) region and a coiled-coil region, which are crucial for its interaction with other subunits of the complex .
The primary function of the COP9 signalosome is to regulate the ubiquitin conjugation pathway by mediating the deneddylation of SCF-type E3 ligase complexes. This deneddylation process leads to a decrease in the ubiquitin ligase activity of SCF-complexes, thereby controlling the degradation of various proteins . Additionally, the COP9 signalosome is involved in the phosphorylation of several key proteins, including p53, c-Jun, IκB-α, and IRF-8. The phosphorylation of these proteins by the CSN complex protects them from degradation by the ubiquitin-proteasome system .
Mutations or dysregulation of the COP9 signalosome subunits, including COPS7A, have been associated with several diseases and developmental disorders. For instance, alterations in the CSN complex can lead to defects in cell cycle control and signal transduction, contributing to the development of cancer and other proliferative diseases .