COMMD7 (COMM Domain Containing 7) is a cytoplasmic protein encoded by the COMMD7 gene located at chromosome 20q11.22 . It belongs to the COMM domain-containing protein family, which plays critical roles in copper metabolism, protein trafficking, and transcriptional regulation . COMMD7 is characterized by its COMM domain, a conserved structural motif that facilitates interactions with other proteins and membranes .
COMMD7 is overexpressed in various cancers, including acute myeloid leukemia (AML), hepatocellular carcinoma (HCC), and pancreatic ductal adenocarcinoma (PDAC) .
AML: High COMMD7 expression is linked to high white blood cell (WBC) counts, peripheral blood (PB) blasts (>70%), and poor survival .
HCC: COMMD7 overexpression correlates with tumor invasion and metastasis .
Clinical Feature | COMMD7 High Expression | P-Value | Source |
---|---|---|---|
WBC >20 × 10⁹/L | OR = 3.16 | <0.001 | |
PB Blasts >70% | OR = 2.89 | 0.002 | |
FLT3 Mutation | OR = 0.32 | 0.002 |
COMMD7 promotes tumor cell proliferation and inhibits apoptosis via the NF-κB pathway .
COMMD7 activates NF-κB by phosphorylating p65 and reducing IκBα degradation . This leads to:
Zinc finger protein 460 (ZNF460) regulates COMMD7 expression, forming a ZNF460-COMMD7-NF-κB axis :
ZNF460 Overexpression: ↑ COMMD7 transcription.
COMMD7 Knockdown: ↓ NF-κB activity, inhibiting proliferation.
AML: Silencing COMMD7 with shRNA reduces tumor growth in xenograft models .
HCC: COMMD7 knockdown inhibits cell invasion and metastasis by downregulating CXCL10 and MMP9 .
COMMD7 is a candidate biomarker for AML diagnosis (AUC = 0.760) . Targeting the ZNF460-COMMD7-NF-κB axis may offer therapeutic strategies for AML and HCC .
COMMD7 (COpper Metabolism gene MURR1 Domain 7) is a member of the COMMD protein family that functions as a regulator of various cellular processes, most notably the NF-κB signaling pathway. This protein contains a conserved COMM domain that mediates protein-protein interactions and influences transcriptional regulation .
COMMD7 appears to have context-dependent functions. In hepatocellular carcinoma (HCC), COMMD7 has been shown to positively regulate NF-κB signaling, promoting cell proliferation and survival . This stands in contrast to some reports suggesting that COMMD7 might bind to the IκB kinase alpha (IKK) complex through NEMO (NF-κB essential modulator) to induce p65 degradation and terminate NF-κB signaling in certain contexts .
The expression pattern of COMMD7 varies across tissues, with abnormally high expression observed in hepatocellular carcinoma tissues compared to adjacent non-cancerous tissues .
COMMD7 demonstrates interesting relationships with other COMMD family members, particularly COMMD1. Research has revealed an inverse correlation between COMMD7 and COMMD1 expression in hepatocellular carcinoma .
Analysis of 35 pairs of HCC tissues and adjacent para-carcinoma tissues showed that while COMMD7 was significantly elevated in HCC tissues, COMMD1 expression was reduced. Statistical analysis confirmed a negative correlation between COMMD7 and COMMD1 (r = −0.6247, p < 0.001) . This suggests potential regulatory interactions between these family members.
When researchers forced expression of COMMD1 in Nanog+ hepatocellular cancer stem cells (HCSCs), they observed that co-transfection with COMMD7 reduced COMMD1 levels, indicating that COMMD7 may antagonize COMMD1 function or expression . The molecular mechanism of this antagonistic relationship warrants further investigation, as it may reveal important insights into the regulation of NF-κB signaling in cancer.
COMMD7 shows distinct expression patterns between normal and cancerous tissues. Multiple studies have demonstrated that COMMD7 is aberrantly overexpressed in hepatocellular carcinoma compared to normal liver tissue .
In a comparative analysis of HCC cell lines, researchers found that:
Cell Line | Cell Type | Relative COMMD7 mRNA Expression |
---|---|---|
L02 | Normal hepatocytes | 0.529±0.065 |
HepG2 | HCC cells | 1.613±0.238 |
Huh7 | HCC cells | 0.874±0.106 |
Hep3B | HCC cells | 0.793±0.116 |
The data demonstrates significantly higher COMMD7 expression in HCC cell lines, particularly HepG2, compared to normal hepatocytes (p<0.01) . Western blot analysis confirmed these findings at the protein level.
Beyond HCC, research examining COMMD7 expression in other cancer types remains limited. Researchers interested in COMMD7 biology should consider evaluating its expression across a broader spectrum of cancer types to determine whether its overexpression is specific to liver cancer or represents a more general feature of malignancy.
COMMD7 exhibits a complex relationship with NF-κB signaling that appears to be context-dependent. In hepatocellular carcinoma, COMMD7 positively regulates NF-κB activity through several mechanisms:
Direct activation of p65 phosphorylation: Overexpression of COMMD7 in Huh7 cells increases phosphorylation of p65, a key component of the NF-κB complex .
ROS-mediated NF-κB activation: COMMD7 triggers the production of reactive oxygen species (ROS), which subsequently activates the NF-κB pathway. When HCC cells overexpressing COMMD7 were treated with N-acetyl cysteine (NAC), an ROS inhibitor, the phosphorylation of p65 was markedly reduced, suggesting that ROS production is an intermediate step in COMMD7-mediated NF-κB activation .
Positive feedback loop: Interestingly, research has found that NF-κB directly binds to the COMMD7 promoter and activates COMMD7 transcription, creating a positive feedback loop that may amplify signaling in cancer cells .
This mechanism differs from some reports suggesting that COMMD proteins, including COMMD7, can negatively regulate NF-κB by facilitating the degradation of p65 . This discrepancy highlights the context-specific nature of COMMD7 function and the need for further research to fully understand its role in different cellular environments.
COMMD7 plays a significant role in hepatocellular cancer stem cell (HCSC) biology, potentially contributing to the self-renewal and tumorigenic properties of these cells:
Elevated expression in HCSCs: Analysis of Nanog-positive HCSCs revealed significantly higher expression of COMMD7 compared to both HCC cell lines (Huh7) and normal liver cell lines (HL-7702) . This suggests a potential role for COMMD7 in stemness maintenance.
Association with stem cell markers: COMMD7 expression positively correlates with the expression of Nanog, a key stemness marker, in HCC tissues and cell lines .
Functional impact on stemness: Silencing COMMD7 in Nanog+ HCSCs inhibited their proliferation and metastatic potential, suggesting that COMMD7 is necessary for maintaining the aggressive phenotype of cancer stem cells .
The relationship between COMMD7 and HCSCs represents an important area for further investigation, as targeting COMMD7 could potentially disrupt cancer stem cell maintenance and improve treatment outcomes for HCC patients.
COMMD7 activates the expression of CXCL10 (C-X-C motif chemokine 10), a chemokine that plays important roles in inflammatory response and tumor progression:
COMMD7 induces CXCL10 production: Overexpression of COMMD7 in Huh7 cells significantly increases CXCL10 production, which can be detected by ELISA in the culture medium .
NF-κB-dependent mechanism: The COMMD7-induced CXCL10 production is dependent on NF-κB activation. Treatment with NF-κB inhibitors (celastrol or Bay 11-7085) blocks COMMD7-mediated CXCL10 production .
ROS-dependent mechanism: COMMD7-induced CXCL10 production also depends on ROS generation. Treatment with NAC (N-acetyl cysteine) substantially reduces CXCL10 production in COMMD7-overexpressing cells .
Autocrine stimulation of proliferation: CXCL10 produced by COMMD7-overexpressing cells acts in an autocrine manner to stimulate HCC cell proliferation. This was demonstrated in experiments where conditioned medium from COMMD7-overexpressing cells promoted the proliferation of naïve HCC cells, an effect that was blocked by treatment with NBI-74330, an inhibitor of CXCR3 (the receptor for CXCL10) .
This COMMD7-ROS-NF-κB-CXCL10 axis represents a significant pathway through which COMMD7 promotes HCC progression, offering multiple potential points for therapeutic intervention.
COMMD7 significantly impacts cellular redox status, with important implications for cancer cell biology:
COMMD7 induces ROS production: Overexpression of COMMD7 in Huh7 cells leads to increased intracellular ROS levels, as measured by DCF (CM-H2DCFDA) probing .
Redox imbalance: COMMD7 overexpression reduces the GSH/GSSG ratio (glutathione/oxidized glutathione), indicating a shift toward a more oxidized intracellular environment .
Condition | Relative DCF Fluorescence | GSH/GSSG Ratio |
---|---|---|
Control | 1.0 (normalized) | ≈3.0 |
COMMD7 overexpression | ≈2.5 | ≈1.5 |
ROS-dependent signaling: The oxidative stress induced by COMMD7 appears to be functionally important, as treatment with the ROS scavenger NAC blocks COMMD7-mediated NF-κB activation and subsequent CXCL10 production .
This relationship between COMMD7 and oxidative stress represents an important mechanism by which COMMD7 may promote cancer progression. It also suggests that targeting redox balance could be an effective strategy for counteracting COMMD7-mediated effects in cancer cells.
Several effective approaches have been employed to modulate COMMD7 expression in experimental studies:
RNA interference (RNAi) for COMMD7 silencing:
shRNA targeting COMMD7: Recombinant pGenesil-COMMD7-shRNA has been successfully used to silence COMMD7 expression in HepG2 cells . This approach achieved significant knockdown of COMMD7 at both mRNA and protein levels.
siRNA transfection: Small interfering RNA can be used for transient knockdown of COMMD7 expression.
Overexpression systems:
Stable expression systems:
When designing experiments to modulate COMMD7 expression, researchers should carefully consider:
The baseline expression level of COMMD7 in the selected cell line
The desired duration of expression modulation (transient vs. stable)
Potential off-target effects of the selected approach
The impact of expression modulation on cell viability and proliferation
Several complementary techniques have proven effective for evaluating COMMD7's impact on NF-κB signaling:
Western blotting for phosphorylated p65:
Nuclear/cytoplasmic fractionation:
Electrophoretic mobility shift assay (EMSA):
Luciferase reporter assays:
Immunofluorescence microscopy:
When designing experiments to assess COMMD7-mediated NF-κB activation, researchers should consider using multiple complementary approaches to build a comprehensive understanding of the pathway regulation.
Designing robust in vivo experiments to study COMMD7 requires careful consideration of several factors:
Model selection:
Xenograft models: Subcutaneous injection of COMMD7-modulated HCC cell lines (e.g., HepG2, Huh7) into immunodeficient mice has been successfully used to evaluate the impact of COMMD7 on tumor growth .
Orthotopic models: While more technically challenging, orthotopic liver implantation provides a more physiologically relevant microenvironment.
Genetically engineered mouse models: Development of COMMD7 transgenic or knockout mice could provide valuable insights but has not been reported in the literature to date.
Experimental endpoints and measurements:
Tumor volume: Regular measurement of tumor dimensions using calipers is a standard approach .
Immunohistochemistry: Analysis of proliferation markers (e.g., Ki67), apoptosis markers, and pathway components in tumor sections .
Molecular analysis: RNA and protein extraction from tumor tissues for expression analysis.
Intervention strategies:
Statistical considerations:
A well-designed in vivo study examining COMMD7 should include multiple complementary endpoints and carefully consider the translational relevance of the selected model and interventions.
COMMD7 overexpression has been associated with poor prognosis in hepatocellular carcinoma patients. While the search results don't provide detailed clinical correlation data, previous studies have indicated that COMMD7 is linked to tumor invasion and poor prognosis .
Researchers investigating the clinical relevance of COMMD7 should consider:
These approaches could help establish the clinical utility of COMMD7 as a prognostic biomarker and potentially identify patient subgroups that might benefit from COMMD7-targeted therapies.
COMMD7 represents a promising target for cancer therapy, particularly in hepatocellular carcinoma, based on several lines of evidence:
Preclinical efficacy of COMMD7 silencing:
Mechanism-based combination strategies:
Targeting the COMMD7-NF-κB axis: Combining COMMD7 inhibition with NF-κB inhibitors (e.g., celastrol, Bay 11-7085) might enhance therapeutic efficacy .
Targeting the CXCL10/CXCR3 pathway: NBI-74330 (a CXCR3 inhibitor) reduced tumor growth by 41.17% in COMMD7-overexpressing xenografts, suggesting potential for combination therapy .
Targeting ROS: Antioxidants could potentially counteract COMMD7-mediated effects by reducing ROS levels and subsequent NF-κB activation .
Potential therapeutic approaches:
RNA interference: siRNA or shRNA targeting COMMD7
Small molecule inhibitors: Development of compounds that disrupt COMMD7 interactions or function
Proteolysis-targeting chimeras (PROTACs): Targeted degradation of COMMD7 protein
Challenges and considerations:
Specificity: Ensuring selective targeting of COMMD7 without affecting other COMMD family members
Delivery: Developing effective delivery systems, particularly for RNA-based therapeutics
Resistance mechanisms: Identifying potential compensatory pathways that might emerge following COMMD7 inhibition
The development of COMMD7-targeted therapeutics represents an exciting frontier in cancer research, with potential applications beyond HCC if COMMD7 is found to play similar roles in other cancer types.
While the search results don't provide direct evidence about COMMD7's interaction with the immune system, several aspects of COMMD7 biology suggest potential immunomodulatory effects:
CXCL10 regulation:
NF-κB pathway modulation:
Oxidative stress regulation:
Future research directions to explore COMMD7's immunomodulatory effects could include:
Analysis of immune cell infiltration in COMMD7-high versus COMMD7-low tumors
Evaluation of immune checkpoint molecule expression in relation to COMMD7 levels
Assessment of COMMD7 expression in immune cells within the tumor microenvironment
Investigation of how COMMD7 modulation affects response to immunotherapies
Understanding the relationship between COMMD7 and the immune system could have important implications for the development of combination therapies that target both COMMD7 and immune checkpoints.
Accurately quantifying COMMD7 protein in clinical samples presents several technical challenges that researchers should address:
Antibody specificity and validation:
Ensuring antibodies specifically recognize COMMD7 without cross-reactivity to other COMMD family members
Validating antibodies using positive and negative controls (e.g., COMMD7-overexpressing and COMMD7-knockdown samples)
Sample preservation and processing:
COMMD7 stability during sample collection and storage
Optimization of protein extraction protocols for formalin-fixed paraffin-embedded (FFPE) versus fresh-frozen tissues
Standardization of normalization methods (loading controls, housekeeping proteins)
Detection methods optimization:
Western blotting: Determining optimal antibody concentrations and incubation conditions
Immunohistochemistry (IHC): Optimizing antigen retrieval, blocking, and staining protocols
Developing quantitative scoring systems for IHC to enable reliable comparison across samples
Contextual interpretation:
Accounting for heterogeneity within tumors (different regions may have varying COMMD7 expression)
Establishing clinically relevant thresholds for "high" versus "low" expression
Correlating protein expression with mRNA levels and functional readouts (e.g., NF-κB activation)
Researchers should consider employing multiple complementary techniques (e.g., western blotting, IHC, mass spectrometry) and including appropriate controls to ensure reliable COMMD7 quantification in clinical samples.
Distinguishing the specific functions of COMMD7 from other COMMD family members requires careful experimental design:
Expression profiling strategies:
Comprehensive analysis of all COMMD family members in the same experimental system
Correlation analysis to identify potential functional redundancy or antagonism
Single-cell analysis to examine co-expression patterns
Selective modulation approaches:
Specific siRNA or shRNA targeting individual COMMD family members
Rescue experiments (e.g., silencing one member while overexpressing another)
CRISPR/Cas9-mediated knockout of individual COMMD genes
Interaction mapping:
Comparative interactome analysis for different COMMD proteins
Domain-specific interaction studies to identify unique versus shared binding partners
Competition assays to examine whether different COMMD proteins compete for the same binding partners
Functional readouts:
Pathway-specific reporter assays to compare effects on signaling
Phenotypic assays to assess functional consequences of modulating specific COMMD proteins
Simultaneous knockdown of multiple COMMD family members to assess additive or synergistic effects
The observed negative correlation between COMMD7 and COMMD1 in HCC tissues provides an interesting starting point for investigating the potentially antagonistic relationship between these family members . Researchers should leverage this observation to design experiments that can elucidate the specific functions of each protein and their potential interplay in normal and diseased states.
Several notable inconsistencies exist in the current understanding of COMMD7 function that warrant further investigation:
Resolving these inconsistencies will require:
Careful comparative studies in multiple cell types and experimental systems
Detailed mechanistic investigations examining direct protein interactions and post-translational modifications
Integration of in vitro findings with in vivo models and clinical data
These efforts will contribute to a more nuanced understanding of COMMD7 biology and potentially reconcile the apparent contradictions in the current literature.
COMMD7 is a 200 amino acid protein that contains a single COMM domain . This domain facilitates interactions with other proteins, including COMMD1, another member of the COMMD family . The protein is widely expressed in various tissues, with the highest expression observed in the lung .
One of the key functions of COMMD7 is its association with the NF-kappa-B complex. NF-kappa-B is a transcription factor that plays a critical role in regulating immune response, inflammation, and cell survival. COMMD7 interacts with this complex and suppresses its transcriptional activity . This suppression is essential for maintaining cellular homeostasis and preventing excessive inflammatory responses.
Recombinant human COMMD7 protein is produced using recombinant DNA technology. The protein is typically expressed in E. coli and purified using conventional chromatography techniques . The recombinant version of COMMD7 often includes a His-tag at the N-terminus, which facilitates its purification and detection .
The recombinant protein is used in various research applications, including studies on protein-protein interactions, signal transduction pathways, and the regulation of transcriptional activity. It is also valuable for investigating the role of COMMD7 in diseases where NF-kappa-B signaling is dysregulated, such as cancer and inflammatory disorders.
The ability of COMMD7 to modulate NF-kappa-B activity makes it a potential target for therapeutic interventions. By understanding how COMMD7 interacts with the NF-kappa-B complex, researchers can develop strategies to modulate this pathway in diseases characterized by chronic inflammation and immune dysregulation.
Additionally, the study of COMMD7 and its interactions with other proteins can provide insights into the broader functions of the COMMD family. This knowledge can contribute to the development of novel therapeutic approaches for a range of diseases.