COPZ1 Human

Coatomer Protein Complex, Subunit Zeta 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function

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 .

Clinical Implications in Glioblastoma

COPZ1 is overexpressed in gliomas and correlates with aggressive tumor behavior:

Prognostic Significance

ParameterObservationSource
mRNA ExpressionElevated in WHO grades II–IV vs. normalTCGA data
Protein Levels4× higher in GBM vs. non-neoplastic tissueIHC/WB
SurvivalHigh COPZ1 linked to shorter OS (HR = 1.8)TCGA

Mechanistic Insights

  • Knockdown Effects:

    • Reduces proliferation in U87MG, U251, and P3#GBM cell lines by 60–80% .

    • Induces ferroptosis via NCOA4-mediated ferritin degradation, increasing labile iron pools and lipid peroxidation .

  • In Vivo Impact:

    • COPZ1 silencing in xenografts decreases tumor volume by 60% and extends survival from 20.8 to 27.8 days (P < 0.05) .

Role in Genetic Disorders

Autosomal recessive COPZ1 mutations cause severe congenital neutropenia (CN):

Mutation Types and Phenotypes

Mutation TypeClinical PresentationCellular Defect
TruncatedPan-hematopoietic defects, multi-organImpaired Golgi-ER transport
MissenseIsolated neutropeniaPartial JAK/STAT signaling loss

Therapeutic Strategies

  • HIF1α Activation: IOX2 rescues granulopoiesis in zebrafish and human CD34+ cells .

  • COPZ2 Compensation: Ectopic COPZ2 expression restores differentiation in COPZ1-mutant cells .

COPZ1 Interaction Network

Key functional partners identified via STRING analysis :

ProteinRoleInteraction Score
COPG1COPI complex stability0.998
ARF1GTPase for vesicle coat assembly0.927
SCYL1Golgi-ER trafficking regulation0.925

Research Gaps and Future Directions

  • Therapeutic Targeting: Small-molecule inhibitors of COPZ1 for GBM require validation in clinical trials.

  • Tissue-Specific Roles: COPZ1’s function in non-cancerous tissues remains underexplored .

  • Mutation Spectrum: Broader sequencing studies needed to assess COPZ1 variants in CN and other disorders .

Product Specs

Introduction
COPZ1, a member of the adaptor complexes small subunit family, constitutes a subunit of the coatomer complex. This complex plays a crucial role in biosynthetic protein transport. COPZ1, specifically, contributes to the regulation of coat assembly and influences the rate of protein transport through its dynamic association and dissociation with the coatomer complex.
Description
Recombinant COPZ1, of human origin, is expressed in E. coli and fused with a 20 amino acid His tag at its N-terminus. This protein is a non-glycosylated polypeptide chain composed of 197 amino acids (residues 1-177), resulting in a molecular weight of 22.3 kDa. Purification of COPZ1 is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a sterile, colorless solution that has been filtered for clarity.
Formulation
The COPZ1 solution is supplied at a concentration of 1 mg/ml and is formulated in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 10% glycerol, 1 mM DTT, and 0.1 M NaCl.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To further enhance long-term stability, consider adding a carrier protein such as HSA or BSA (0.1%). Repeated freezing and thawing of the product should be avoided.
Purity
The purity of COPZ1 is determined by SDS-PAGE analysis to be greater than 95.0%.
Synonyms
Coatomer protein complex subunit zeta 1, Zeta-1 COP, CGI-120, COPZ.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MEALILEPSL YTVKAILILD NDGDRLFAKY YDDTYPSVKE QKAFEKNIFN KTHRTDSEIA LLEGLTVVYK SSIDLYFYVI GSSYENELML MAVLNCLFDS LSQMLRKNVE KRALLENMEG LFLAVDEIVD GGVILESDPQ QVVHRVALRG EDVPLTEQTV SQVLQSAKEQ IKWSLLR.

Q&A

What is COPZ1 and what are its primary cellular functions?

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

  • Constructing coated vesicles on the Golgi membrane

COPZ1 functions primarily through its association-dissociation properties with the coatomer complex, which mediates retrograde protein transport from the Golgi to the endoplasmic reticulum .

How is COPZ1 structurally characterized and what experimental tools are available?

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 .

How does COPZ1 expression pattern differ between tumor and normal tissues?

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)

  • Uterine corpus endometrial carcinoma (UCEC)

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 .

What is the prognostic significance of COPZ1 expression in cancer?

The negative prognostic impact of COPZ1 overexpression appears consistent across different cancer types, suggesting its potential utility as a universal prognostic biomarker in oncology .

What mechanisms regulate COPZ1 expression in cancer cells?

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 .

What is the functional relationship between COPZ1 and COPZ2 in normal versus cancer cells?

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

  • Dependency on COPZ1 for survival (synthetic lethality)

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 .

How does the COPZ2-microRNA 152 relationship impact cancer biology?

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 .

What approaches are most effective for studying COPZ1 function in cancer models?

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:

    • Golgi apparatus morphology examination via microscopy

    • Autophagy assessment using LC3-II markers

    • Cell death characterization (apoptosis, ferroptosis)

    • Cell cycle and proliferation analysis

  • Functional rescue experiments:

    • COPZ1 reexpression in knockout models

    • COPZ2 overexpression to assess functional complementation

    • HIF1α pathway modulation using IOX2

  • Animal models:

    • Zebrafish embryos for myelopoiesis studies

    • Conditional knockout mouse models

    • Patient-derived xenografts for translational research

How can researchers effectively analyze COPZ1-associated pathways in tumors?

To comprehensively evaluate COPZ1-associated pathways:

  • Transcriptomic analysis:

    • RNA-seq to identify differentially expressed genes upon COPZ1 inhibition

    • Single-sample Gene Set Enrichment Analysis (ssGSEA) to calculate hypoxia and stemness scores

    • Analysis of stemness indices, proliferation signatures, and EMT markers

  • Tumor microenvironment assessment:

    • CIBERSORT algorithm to deconvolute immune cell infiltration

    • ESTIMATE algorithm to evaluate immunological and stromal scores

    • Analysis of 29 different tumor microenvironment signatures

    • "Cytosig" tool to evaluate activity of 43 cytokine pathways

  • Regulatory mechanism investigation:

    • Methylation analysis

    • SNP and CNV evaluation

    • Transcription factor binding prediction using databases like hTFtarget

    • MicroRNA regulatory network analysis

How do COPZ1 mutations contribute to hematopoietic disorders?

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

  • Blocked retrograde protein transport from Golgi to ER

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 .

What molecular pathways are disrupted by COPZ1 mutations in hematopoietic cells?

COPZ1 mutations disrupt multiple critical signaling pathways in hematopoietic cells:

  • Downregulation of key signaling pathways:

    • JAK/STAT signaling

    • CEBPE (a crucial granulocytic transcription factor)

    • G-CSF receptor expression

    • Hypoxia-responsive pathways

  • Abnormal activation of stress responses:

    • Induction of STING pathway

    • Upregulation of interferon-stimulated genes

    • Increased oxidative phosphorylation activity

    • Elevated reactive oxygen species (ROS) levels

The dysregulation extent correlates with mutation type, with truncating mutations causing more severe pathway disruption than missense variants .

What therapeutic approaches show promise for COPZ1-related neutropenia?

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 .

How does COPZ1 inhibition trigger cell death in cancer cells?

COPZ1 inhibition induces multiple cellular stress and death pathways:

  • Disruption of cellular architecture:

    • Collapse of the Golgi apparatus

    • Impaired vesicular trafficking

    • Disruption of protein secretion and sorting

  • Blockade of cytoprotective mechanisms:

    • Inhibition of autophagy, a survival mechanism often exploited by cancer cells

    • Dysregulation of cellular homeostasis

  • Induction of programmed cell death:

    • Activation of apoptosis in both proliferating and non-dividing tumor cells

    • Induction of ferroptosis, an iron-dependent form of regulated 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 .

What is the relationship between COPZ1 and ferroptosis?

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

  • Oxidative cell damage leading to ferroptotic death

This represents a novel mechanism through which COPZ1 targeting could overcome resistance to apoptosis in cancer cells, as ferroptosis operates independently of apoptotic machinery .

How does COPZ1 influence tumor immune microenvironment?

COPZ1 expression shows significant associations with the tumor immune microenvironment:

  • Negative correlation with immune infiltration:

    • COPZ1 expression negatively correlates with immune score and stromal score

    • Low COPZ1 expression associates with higher antitumor immune cell infiltration

  • Impact on cytokine signaling:

    • Low COPZ1 expression correlates with increased pro-inflammatory cytokines

    • COPZ1 expression affects the activity of multiple immune response pathways

  • Macrophage polarization:

    • COPZ1 expression shows inverse relationship with anti-inflammatory M2 macrophage infiltration

    • This suggests COPZ1 may influence immunosuppressive tumor microenvironment

These associations indicate that COPZ1 may modulate tumor immunogenicity and potentially influence response to immunotherapy .

What are potential approaches for targeting COPZ1 in cancer therapy?

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:

    • COPZ1-specific siRNA delivery systems

    • CRISPR-based COPZ1 inactivation strategies

    • COPZ2/microRNA 152 reexpression vectors

  • Combination strategies:

    • Synergistic pairing with ferroptosis inducers

    • Combination with autophagy inhibitors

    • Integration with immune checkpoint blockade

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 .

How can multi-omics approaches enhance COPZ1 research?

Integrated multi-omics approaches can provide comprehensive insights into COPZ1 biology:

  • Genomic and transcriptomic integration:

    • Correlation of COPZ1 CNV with expression patterns

    • Analysis of alternative splicing and transcript variants

    • Identification of co-expression networks

  • Epigenomic analysis:

    • Characterization of DNA methylation patterns regulating COPZ1

    • Histone modification profiling at COPZ1 locus

    • Chromatin accessibility assessment

  • Proteomic and interactomic studies:

    • COPZ1 protein interaction network mapping

    • Post-translational modification profiling

    • Subcellular localization under various conditions

  • Single-cell analysis:

    • Cell type-specific expression patterns

    • Identification of tumor subpopulations with differential COPZ1 dependency

    • Characterization of COPZ1's relationship with cellular states (stemness, EMT)

These multi-dimensional approaches can reveal new aspects of COPZ1 biology and inform more precise therapeutic strategies.

Product Science Overview

Structure and Function

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 .

Gene and Protein Information

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 .

Biological Pathways

COPZ1 is involved in several critical biological pathways, including:

  • Transport to the Golgi and subsequent modification
  • wtCFTR and delta508-CFTR traffic / Generic schema (norm and CF)
Expression and Localization

COPZ1 is expressed in various tissues, including lymphoid tissue, bone marrow, testis, and skeletal muscle . It is involved in processes such as cell proliferation, innate immune response, protein ubiquitination, and lymph vessel development .

Clinical Significance

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 .

Research and Applications

Human recombinant COPZ1 is used in research to study its role in intracellular protein trafficking and autophagy. It is also utilized in various assays and experiments to understand its function and regulation within the cell .

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.