COPZ1 is encoded by the COPZ1 gene located on human chromosome 12 and functions as a subunit of the COPI (Coatomer Protein Complex I). Key features include:
Cellular Role: Facilitates retrograde protein transport between the Golgi apparatus and endoplasmic reticulum (ER), essential for maintaining organelle integrity and protein sorting .
Interactions: Binds to COPG1 and ARF1, regulating vesicle budding and cargo selection .
Iron Metabolism: Modulates ferritin degradation via NCOA4, influencing intracellular iron levels and ferroptosis .
COPZ1 is overexpressed in gliomas and correlates with aggressive tumor behavior:
Knockdown Effects:
In Vivo Impact:
Autosomal recessive COPZ1 mutations cause severe congenital neutropenia (CN):
| Mutation Type | Clinical Presentation | Cellular Defect |
|---|---|---|
| Truncated | Pan-hematopoietic defects, multi-organ | Impaired Golgi-ER transport |
| Missense | Isolated neutropenia | Partial JAK/STAT signaling loss |
HIF1α Activation: IOX2 rescues granulopoiesis in zebrafish and human CD34+ cells .
COPZ2 Compensation: Ectopic COPZ2 expression restores differentiation in COPZ1-mutant cells .
Key functional partners identified via STRING analysis :
| Protein | Role | Interaction Score |
|---|---|---|
| COPG1 | COPI complex stability | 0.998 |
| ARF1 | GTPase for vesicle coat assembly | 0.927 |
| SCYL1 | Golgi-ER trafficking regulation | 0.925 |
MGSSHHHHHH SSGLVPRGSH MEALILEPSL YTVKAILILD NDGDRLFAKY YDDTYPSVKE QKAFEKNIFN KTHRTDSEIA LLEGLTVVYK SSIDLYFYVI GSSYENELML MAVLNCLFDS LSQMLRKNVE KRALLENMEG LFLAVDEIVD GGVILESDPQ QVVHRVALRG EDVPLTEQTV SQVLQSAKEQ IKWSLLR.
COPZ1 is a member of the adaptor complexes small subunit family and serves as a critical component of the coatomer protein complex I (COPI). This complex contains alpha, beta, beta', gamma, delta, epsilon, and zeta subunits . COPZ1 plays essential roles in:
Regulating coat assembly in the COPI complex
Controlling the rate of biosynthetic protein transport
Participating in the maturation of endosomes
Contributing to autophagy pathways
COPZ1 functions primarily through its association-dissociation properties with the coatomer complex, which mediates retrograde protein transport from the Golgi to the endoplasmic reticulum .
COPZ1 Human is a single, non-glycosylated polypeptide chain containing 177 amino acids with a molecular mass of 22.3kDa. The protein contains highly conserved regions that are critical for interactions with its COPI complex partners, particularly COPG1 .
For experimental studies, recombinant COPZ1 Human protein is available as an N-terminal His-tagged protein expressed in E. coli . This recombinant protein can be used for:
Protein-protein interaction studies
Structural analyses
Development of antibodies for detection
Biochemical characterization of COPZ1 function
The protein's secondary structure contains regions involved in COPI complex assembly, providing potential targets for structure-function studies .
COPZ1 exhibits significant overexpression in multiple cancer types compared to corresponding normal tissues. RNA expression analysis through TIMER2 database has demonstrated COPZ1 upregulation in:
Bladder urothelial carcinoma (BLCA)
Breast carcinoma (BRCA)
Cholangiocarcinoma (CHOL)
Colon adenocarcinoma (COAD)
Esophageal carcinoma (ESCA)
Head and neck squamous cell cancer (HNSC)
Kidney renal papillary cell carcinoma (KIRP)
Liver hepatocellular cancer (LIHC)
Lung adenocarcinoma (LUAD)
Lung squamous cell cancer (LUSC)
Prostate adenocarcinoma (PRAD)
Rectum adenocarcinoma (READ)
Stomach adenocarcinoma (STAD)
In gliomas specifically, COPZ1 mRNA levels increase progressively from low-grade (WHO II) to high-grade tumors (WHO III and IV) compared to non-neoplastic brain tissue. Protein levels show approximately 4-fold higher expression in grade IV gliomas relative to normal brain tissue .
The negative prognostic impact of COPZ1 overexpression appears consistent across different cancer types, suggesting its potential utility as a universal prognostic biomarker in oncology .
COPZ1 expression in tumors is regulated through multiple mechanisms:
Genomic alterations: Copy number variations (CNVs) can lead to abnormal COPZ1 expression
Epigenetic regulation: DNA methylation patterns influence COPZ1 transcription
Transcriptional control: Specific transcription factors modulate COPZ1 expression
Post-transcriptional regulation: MicroRNAs can regulate COPZ1 mRNA stability and translation
These multi-level regulatory mechanisms contribute to the aberrant expression of COPZ1 observed in various cancer types, presenting potential targets for therapeutic intervention .
In normal cells, both COPZ1 and COPZ2 are expressed and can functionally compensate for each other. This redundancy explains why normal cells require the simultaneous knockdown of both COPZ1 and COPZ2 to inhibit growth .
In contrast, most cancer cells display:
Maintained or elevated COPZ1 expression
Significant downregulation of COPZ2
Experimental evidence shows that reexpression of COPZ2 in tumor cells protects them from death induced by COPZ1 knockdown, confirming that tumor cell dependence on COPZ1 results directly from COPZ2 silencing .
COPZ2 harbors microRNA 152, which functions as a tumor suppressor both in vitro and in vivo. In cancer cells, both microRNA 152 and its host gene COPZ2 are frequently silenced concurrently . This relationship creates an intriguing cancer biology paradigm:
COPZ2 itself displays no direct tumor-suppressive activities
microRNA 152 embedded within COPZ2 acts as a tumor suppressor
Silencing of this microRNA in cancer leads to concurrent COPZ2 downregulation
This silencing makes cancer cells exclusively dependent on COPZ1 for survival
This dependency creates a potential therapeutic vulnerability that could be exploited by COPZ1-targeting agents .
Several complementary approaches can be employed to study COPZ1 function in cancer:
Gene silencing:
siRNA or shRNA for transient or stable knockdown
CRISPR-Cas9 for complete gene knockout
Comparison with COPZ2 knockdown as control
Cellular phenotype analysis:
Functional rescue experiments:
Animal models:
To comprehensively evaluate COPZ1-associated pathways:
Transcriptomic analysis:
Tumor microenvironment assessment:
Regulatory mechanism investigation:
COPZ1 mutations can cause a newly identified inherited bone marrow failure syndrome characterized by severe congenital neutropenia. Both stop-codon (truncating) and missense mutations in evolutionarily conserved regions of COPZ1 have been identified in affected patients .
The pathophysiology involves:
Impaired granulocytic differentiation in human CD34+ cells
Defective myelopoiesis in zebrafish embryos
Diminished interaction with COPI complex partner COPG1
The severity of the clinical phenotype varies with mutation type. While truncating mutations may affect multiple hematological lineages and non-hematological tissues, missense mutations appear to cause isolated neutropenia .
COPZ1 mutations disrupt multiple critical signaling pathways in hematopoietic cells:
Downregulation of key signaling pathways:
Abnormal activation of stress responses:
The dysregulation extent correlates with mutation type, with truncating mutations causing more severe pathway disruption than missense variants .
Two promising therapeutic strategies have demonstrated efficacy in preclinical models:
Small molecule therapy: Treatment with the HIF1α activator IOX2 restored defective granulopoiesis in COPZ1-mutated CD34+ cells. This suggests that targeting the hypoxia pathway could compensate for COPZ1 deficiency .
Gene therapy approach: Transduction of cells with COPZ2 cDNA successfully rescued the granulopoiesis defect in COPZ1-mutated cells, indicating that COPZ2 can functionally compensate for COPZ1 loss in hematopoietic cells .
These findings provide rational therapeutic strategies for patients with congenital neutropenia caused by COPZ1 mutations .
COPZ1 inhibition induces multiple cellular stress and death pathways:
Disruption of cellular architecture:
Blockade of cytoprotective mechanisms:
Induction of programmed cell death:
Importantly, these death mechanisms affect both dividing and non-dividing tumor cells, potentially overcoming a major limitation of conventional cytotoxic therapies that primarily target proliferating cells .
COPZ1 inhibition induces ferroptosis, a form of regulated cell death characterized by iron-dependent lipid peroxidation. This process is mediated by the NCOA4 protein .
The molecular mechanism involves:
NCOA4-mediated degradation of ferritin (ferritinophagy)
Increase in free iron pool
Enhanced lipid peroxidation
This represents a novel mechanism through which COPZ1 targeting could overcome resistance to apoptosis in cancer cells, as ferroptosis operates independently of apoptotic machinery .
COPZ1 expression shows significant associations with the tumor immune microenvironment:
Negative correlation with immune infiltration:
Impact on cytokine signaling:
Macrophage polarization:
These associations indicate that COPZ1 may modulate tumor immunogenicity and potentially influence response to immunotherapy .
Several strategic approaches could be developed for COPZ1-targeted cancer therapy:
Small molecule inhibitors:
Direct inhibitors of COPZ1 protein function
Destabilizers of COPZ1-containing protein complexes
Compounds inducing COPZ1 protein degradation
Gene therapy approaches:
Combination strategies:
The therapeutic window exists because cancer cells with silenced COPZ2 are uniquely vulnerable to COPZ1 inhibition, while normal cells with intact COPZ2 expression would be protected from toxicity .
Integrated multi-omics approaches can provide comprehensive insights into COPZ1 biology:
Genomic and transcriptomic integration:
Epigenomic analysis:
Proteomic and interactomic studies:
COPZ1 protein interaction network mapping
Post-translational modification profiling
Subcellular localization under various conditions
Single-cell analysis:
These multi-dimensional approaches can reveal new aspects of COPZ1 biology and inform more precise therapeutic strategies.
The coatomer complex binds to dilysine motifs and reversibly associates with Golgi non-clathrin-coated vesicles. This association mediates biosynthetic protein transport from the endoplasmic reticulum (ER), through the Golgi apparatus, and up to the trans-Golgi network . The zeta subunit, COPZ1, is particularly involved in regulating the coat assembly and the rate of biosynthetic protein transport due to its association-dissociation properties with the coatomer complex .
The COPZ1 gene is located on chromosome 12 and is a protein-coding gene. It has several aliases, including CGI-120, COPZ, and HSPC181 . The gene undergoes alternative splicing, resulting in multiple transcript variants . The protein encoded by COPZ1 is essential for the retrograde Golgi-to-ER transport of dilysine-tagged proteins .
Mutations or dysregulation of COPZ1 can have significant implications for cellular function and may be associated with various diseases. The coatomer complex, including COPZ1, is essential for the proper functioning of cellular transport mechanisms, and any disruption can lead to cellular dysfunction .