COMMD1 Human

Copper Metabolism Domain Containing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Complex Assembly

COMMD1 belongs to the COMMD protein family (COMMD1–10), characterized by a conserved C-terminal COMM domain for protein interactions and variable N-terminal regions . Within the Commander complex, COMMD1 integrates into a hetero-hexadecameric structure comprising:

SubcomplexComponentsFunction
COMMD CoreCOMMD1–10Scaffold for complex assembly
Effector ModuleDENND10, Retriever (VPS26C, VPS29, VPS35L)Vesicle trafficking and cargo sorting
Scaffold ProteinsCCDC22, CCDC93Structural stabilization and functional linkage

Cryo-EM studies reveal that COMMD1 resides in the flexible N-terminal region of the complex, enabling dynamic interactions with transcription factors and signaling molecules .

Copper Homeostasis

COMMD1 regulates copper excretion by promoting the degradation of copper-transporting ATPases ATP7A/ATP7B. Mutations in COMMD1 cause hepatic copper accumulation in Bedlington Terriers and are implicated in Wilson disease-like pathologies .

Endosomal Trafficking

As part of the Commander complex, COMMD1 facilitates recycling of transmembrane proteins (e.g., LDLR, CFTR) from endosomes to the cell surface, preventing lysosomal degradation .

Transcriptional Regulation

  • NF-κB Pathway: COMMD1 inhibits NF-κB by enhancing IκB stability and promoting nuclear export of RelA .

  • HIF-1α Inhibition: Binds HIF-1α’s N-terminus, blocking dimerization with HIF-1β and suppressing hypoxia-responsive genes (e.g., VEGFA, CXCR4) .

Cell Cycle and Apoptosis

COMMD1 modulates p21 Cip1 levels, influencing G1/S transition and proliferation. Its interaction with DRR1 and F-actin further regulates neuroblastoma growth .

Cancer

COMMD1 acts as a tumor suppressor across multiple malignancies:

Cancer TypeMechanismClinical Impact
Lung CancerSilencing promotes invasion via NF-κB/HIF-1α activation Poor prognosis
Hepatocellular CarcinomaBinds HIF-1α, reducing angiogenesis and metastasis Enhanced survival in high-expression cases
Prostate CancerDownregulates cyclin D1, arresting cell cycle Reduced tumor proliferation
Multiple MyelomaOverexpression confers bortezomib resistance via SCF complex inhibition Therapeutic resistance

Low COMMD1 expression correlates with advanced tumor stage and metastasis in colorectal, breast, and head/neck cancers .

Copper Metabolism Disorders

  • Canine Copper Toxicosis: Exon 2 deletions in COMMD1 cause hepatic copper accumulation and cirrhosis .

  • Wilson Disease: COMMD1 variants may modify ATP7B dysfunction, though direct causality remains unproven .

Inflammatory Diseases

COMMD1 represses pro-inflammatory genes (e.g., CXCL10, IL-12B) in macrophages, protecting against colitis in murine models . Inflammation-induced COMMD1 downregulation exacerbates tissue damage .

Therapeutic Implications

  • Targeting HIF-1α: COMMD1 mimetics (e.g., CIGB-552) suppress HIF-driven angiogenesis in preclinical models .

  • Copper Chelation: Restoring COMMD1 function could alleviate copper overload in metabolic disorders .

  • NF-κB Inhibitors: Enhancing COMMD1 activity may mitigate inflammation in autoimmune diseases .

Research Gaps and Future Directions

  • Mechanistic Insights: The COMMD1-HIF-1α interaction site and allosteric regulation require structural elucidation .

  • Clinical Biomarkers: COMMD1’s utility as a prognostic marker in cancers warrants validation in large cohorts .

  • Drug Development: Small molecules modulating COMMD1’s interactions with Retriever or NF-κB remain unexplored .

Product Specs

Introduction
COMMD1 is responsible for inhibiting TNF-induced NF-κB p50 and facilitating biliary copper excretion within hepatocytes. It is highly expressed in liver tissue, with lower expressions in lung, heart, kidney and brain tissue. COMMD1 interacts directly with COMMD6 and ATP7B, and indirectly with Iκ-β and COMMD7.
Description
Recombinant Human COMMD1, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 210 amino acids (amino acids 1-190) with a molecular weight of 23.3 kDa. It is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
COMMD1 Human solution in 20mM Tris buffer, pH 8.0, containing 0.1M NaCl and 20% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms
MURR1, C2orf5, COMMD1, Copper Metabolism Domain Containing 1, MGC27155.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MAAGELEGGK PLSGLLNALA QDTFHGYPGI TEELLRSQLY PEVPPEEFRP FLAKMRGILK SIASADMDFN QLEAFLTAQT KKQGGITSDQ AAVISKFWKS HKTKIRESLM NQSRWNSGLR GLSWRVDGKS QSRHSAQIHT PVAIIELELG KYGQESEFLC LEFDEVKVNQ ILKTLSEVEE SISTLISQPN.

Q&A

What is COMMD1 and what are its primary cellular functions?

COMMD1 is a scaffold protein that mediates levels, stability, and proteolysis of various substrate proteins. It functions in multiple cellular pathways, most notably:

  • Copper homeostasis regulation

  • NF-κB signaling inhibition

  • Hypoxia signaling regulation through HIF-1α interaction

  • Protein quality control and aggregation modulation

The protein contains a distinctive COMM domain at its C-terminus which is necessary and sufficient for interaction with binding partners such as HIF-1α . COMMD1 acts as an inhibitor of transcription factors including NF-κB and HIF, with implications for inflammatory responses and oxygen homeostasis .

How is COMMD1 detected in laboratory settings?

COMMD1 can be detected through several standard laboratory techniques:

  • Western Blot: Using specific antibodies like the Mouse Anti-Human COMMD1 Monoclonal Antibody (MAB7526), COMMD1 appears as a band at approximately 20 kDa under reducing conditions .

  • Immunofluorescence: COMMD1 can be visualized in cells using immunofluorescence techniques, where it shows localization primarily in the cytoplasm and nuclei .

  • Immunoprecipitation: COMMD1 interactions with binding partners can be studied using co-immunoprecipitation assays, as demonstrated with LDLR .

  • Sucrose gradient fractionation: This technique helps identify COMMD1's presence in specific cellular compartments and protein complexes .

For optimal results, researchers should validate antibody specificity, as exemplified by control experiments showing specific staining in U2OS human osteosarcoma cell lines but not in COMMD1-deficient cells .

What cellular compartments contain COMMD1?

COMMD1 demonstrates a complex subcellular distribution pattern:

  • Nuclear localization: COMMD1 can be found in the nucleus where it interacts with transcription factors like HIF-1α .

  • Cytoplasmic presence: Immunofluorescence studies show clear cytoplasmic staining .

  • Endosomal association: COMMD1 has been identified in endosomal compartments where it associates with the WASH complex .

  • Membrane protein interactions: COMMD1 interactions with membrane proteins like LDLR suggest its presence near cellular membranes .

In fluorescence microscopy studies, COMMD1 shows both diffuse cytoplasmic staining and punctate structures, suggesting its presence in specific cellular compartments or protein complexes . The protein's distribution may change under different cellular conditions and in response to stressors.

How does COMMD1 expression correlate with cancer progression?

COMMD1 expression is frequently suppressed in human cancers, with significant implications for disease progression:

  • Multiple studies demonstrate reduced COMMD1 expression across various cancer types .

  • Decreased COMMD1 expression correlates with a more invasive tumor phenotype .

  • Direct repression of COMMD1 in human cell lines leads to increased tumor invasion in experimental models .

  • Conversely, increased COMMD1 expression in mouse melanoma cells results in decreased lung metastasis .

This inverse relationship between COMMD1 levels and cancer invasiveness suggests COMMD1 may function as a tumor suppressor. The mechanism appears to involve multiple pathways, notably through regulation of HIF-mediated gene expression, which controls processes critical for tumor growth including angiogenesis and metabolic adaptation .

What is the mechanism by which COMMD1 regulates HIF activity in cancer cells?

COMMD1 regulates HIF activity through a direct protein-protein interaction mechanism:

  • COMMD1 binds directly to the amino terminus of HIF-1α (amino acids 1-300), specifically interacting with the bHLH and PAS domains .

  • This interaction prevents HIF-1α from dimerizing with HIF-1β, a necessary step for HIF transcriptional activity .

  • By preventing dimerization, COMMD1 inhibits HIF DNA binding and subsequent transcriptional activation of target genes .

  • The COMM domain of COMMD1 is both necessary and sufficient for this HIF-1α binding interaction .

  • The binding appears to be oxygen-regulated, with preferential interaction during normoxia, though this regulation does not depend on prolyl hydroxylation (unlike the HIF-1α/VHL interaction) .

This mechanism explains why COMMD1 deficiency leads to increased HIF target gene expression even without changes in HIF-1α protein levels. In reporter assays, decreased COMMD1 expression results in greater HIF transcriptional activity, particularly during normoxia .

What experimental approaches can assess COMMD1's role in tumor invasion?

Researchers investigating COMMD1's role in tumor invasion employ several experimental approaches:

  • Gene expression manipulation:

    • RNA interference to decrease COMMD1 expression

    • Overexpression systems to increase COMMD1 levels

    • Creation of COMMD1-deficient cell lines using knockout techniques

  • In vivo models:

    • Chick xenograft models for assessing invasive behavior of human cancer cells with modified COMMD1 expression

    • Mouse metastasis models, particularly lung metastasis assays with melanoma cells expressing different levels of COMMD1

  • Gene expression analysis:

    • Correlation of COMMD1 levels with expression of invasion-promoting genes

    • Specific analysis of HIF target genes in response to COMMD1 manipulation

  • Biochemical interaction studies:

    • Co-immunoprecipitation to identify COMMD1 protein partners

    • In vitro binding assays using recombinant proteins

    • ELISA-based binding assays to measure direct interactions

These complementary approaches provide insights into both the molecular mechanisms and biological consequences of COMMD1 expression changes in cancer progression.

How does COMMD1 affect protein aggregation in neurodegenerative diseases?

COMMD1 demonstrates a complex, client-specific effect on protein aggregation relevant to neurodegenerative disorders:

  • Amyotrophic Lateral Sclerosis (ALS): COMMD1 enhances the formation of mutant SOD1 (mSOD1) aggregates upon binding . It co-localizes with mSOD1 inclusions and forms high molecular weight complexes in the presence of mSOD1 .

  • Parkinson's Disease: In contrast to its effect on mSOD1, COMMD1 decreases the abundance of mutant Parkin inclusions associated with Parkinson's disease .

  • Huntington's Disease: COMMD1 appears to have minimal effect on the aggregation of polyglutamine-expanded Huntingtin protein, causative of Huntington's disease .

This differential effect suggests COMMD1 may recognize specific structural features of misfolded proteins rather than acting as a general regulator of protein aggregation. The opposing effects on different disease-associated proteins indicate COMMD1 likely functions through multiple mechanisms depending on the client protein involved .

What experimental methods are used to study COMMD1's impact on protein aggregation?

Several specialized techniques are employed to investigate COMMD1's role in protein aggregation:

  • Fluorescence microscopy: Used to visualize protein inclusions and co-localization of COMMD1 with aggregated proteins in cellular models .

  • Filter trap assays: Quantitative measurement of detergent-insoluble protein aggregates retained on cellulose acetate membranes .

  • Protein fractionation methods: Including differential centrifugation to separate soluble and insoluble protein fractions, followed by immunoblotting analysis.

  • Size exclusion chromatography: Identifies high molecular weight complexes formed between COMMD1 and aggregation-prone proteins .

  • Pulse-chase experiments: Assess the turnover rates of normal and aggregated proteins in the presence or absence of COMMD1.

  • Cell viability assays: Determine whether COMMD1-mediated changes in protein aggregation affect cellular health and survival.

When designing experiments to study COMMD1's role in aggregation, researchers should include multiple client proteins to account for the differential effects observed across disease models .

How does COMMD1 interact with the HIF pathway components?

COMMD1 interacts with multiple components of the HIF pathway in an intricate manner:

  • HIF-1α binding:

    • COMMD1 binds directly to the amino-terminal region of HIF-1α (amino acids 1-300)

    • This region contains the bHLH and PAS domains involved in dimerization and DNA binding

    • The COMM domain of COMMD1 is necessary and sufficient for this interaction

    • The binding is oxygen-regulated but independent of prolyl hydroxylation

  • HIF-2α and HIF-1β interactions:

    • COMMD1 can also bind to HIF-2α and HIF-1β

    • Like with HIF-1α, COMMD1 interacts with the bHLH/PAS regions of these proteins

    • The interaction with HIF-1β appears to be direct and not mediated through HIF-1α or HIF-2α

  • Competition with heterodimerization:

    • COMMD1 and HIF-1β compete for binding to HIF-1α

    • Increased HIF-1β expression diminishes the interaction between COMMD1 and HIF-1α

    • Conversely, COMMD1 overexpression reduces the interaction between HIF-1α and HIF-1β

This competitive binding mechanism explains how COMMD1 inhibits HIF-mediated transcription by preventing the formation of functional HIF-1α/β heterodimers necessary for DNA binding and transcriptional activation .

What techniques are most effective for studying COMMD1 protein-protein interactions?

Several complementary techniques have proven effective for investigating COMMD1's protein interactions:

  • Co-immunoprecipitation (Co-IP): Effectively demonstrates interactions between COMMD1 and partners like HIF-1α, SOD1, and LDLR in cellular contexts . Works well with both endogenous and tagged proteins.

  • GST pull-down assays: Useful for confirming direct interactions, as shown with GST-tagged COMMD1 or domain fragments . This approach helped identify the importance of the COMM domain in binding.

  • Solid-phase ELISA binding assays: Appropriate for quantifying direct binding between purified recombinant proteins, as demonstrated with COMMD1 and HIF-1α fragments .

  • Immunofluorescence co-localization: Valuable for visualizing spatial relationships between COMMD1 and its partners in cells, particularly for aggregation-prone proteins .

  • Sucrose gradient fractionation: Helps identify COMMD1's presence in specific cellular compartments and protein complexes .

  • Competition assays: Critical for understanding the dynamic interplay between COMMD1 and other proteins competing for the same binding partners, as shown with HIF-1α/β .

For optimal results, combinations of these techniques should be employed, starting with co-IP to identify interactions, followed by in vitro binding assays to confirm direct binding, and finally cellular studies to validate physiological relevance.

What structural domains of COMMD1 are critical for its various functions?

COMMD1's functional activities map to specific structural domains:

  • COMM domain (C-terminal region):

    • Necessary and sufficient for binding to HIF-1α

    • Critical for most protein-protein interactions

    • Defining feature of the COMMD protein family

    • Essential for the protein's role in regulating transcription factors

  • N-terminal region:

    • Contains sequences that may regulate cellular localization

    • May contribute to client specificity in protein aggregation contexts

    • Provides additional binding surfaces for certain interaction partners

  • Full protein structure:

    • The complete protein structure is required for some functions

    • Certain activities may depend on proper folding and orientation of multiple domains

    • Post-translational modifications may affect domain functions

Experimental approaches that employ domain mapping, using truncated constructs or point mutations, have been invaluable in identifying these critical regions. For example, studies with the isolated COMM domain demonstrated its sufficiency for HIF-1α binding, while full-length protein was needed for certain other functions .

What are the optimal conditions for detecting COMMD1-HIF interactions?

Detecting COMMD1-HIF interactions requires careful consideration of experimental conditions:

  • Oxygen conditions: The COMMD1-HIF-1α interaction is oxygen-regulated, with preferential binding during normoxia . Experiments should compare normoxic and hypoxic conditions (typically 1% O₂).

  • Cell lysis buffers:

    • Use buffers containing 0.1% NP-40 or similar mild detergents

    • Include protease inhibitors to prevent degradation

    • Consider phosphatase inhibitors to preserve post-translational modifications

    • Maintain cold temperatures throughout processing

  • Binding detection methods:

    • Co-immunoprecipitation works well for endogenous interactions

    • For weaker interactions, crosslinking prior to lysis may help

    • In vitro binding assays require properly folded recombinant proteins

    • Consider both forward and reverse co-IP approaches

  • Controls:

    • Include prolyl hydroxylase inhibitors (e.g., DFO) as controls

    • VHL-HIF interactions serve as positive controls for oxygen-regulated binding

    • Include domain mutants to confirm binding specificity

  • Detection reagents:

    • Use high-quality antibodies validated for immunoprecipitation

    • For tagged proteins, verify tag does not interfere with interaction

These optimized conditions help overcome challenges in detecting physiologically relevant COMMD1-HIF interactions, which can be transient and regulated by cellular oxygen status .

How can researchers effectively model COMMD1 deficiency in experimental systems?

Multiple approaches can be used to model COMMD1 deficiency, each with specific advantages:

  • RNA interference approaches:

    • siRNA for transient knockdown studies (3-5 days)

    • shRNA for stable knockdown models

    • Benefits: Relatively quick, can be used in many cell types

    • Limitations: Incomplete knockdown, potential off-target effects

  • CRISPR/Cas9 gene editing:

    • Complete knockout cell lines

    • Introduction of specific mutations

    • Benefits: Complete elimination of protein, models genetic conditions

    • Limitations: More time-consuming, potential for compensatory mechanisms

  • Mouse models:

    • Conditional knockout using Cre-lox system (e.g., Commd1ᶠ/ᶠ)

    • Tissue-specific deletion models

    • Benefits: In vivo relevance, study systemic effects

    • Limitations: Expensive, time-consuming, potential developmental effects

  • Complementation studies:

    • Re-expression of COMMD1 in deficient systems

    • Domain mutants to dissect specific functions

    • Benefits: Validates specificity, identifies critical domains

    • Approach: Use rescue experiments to confirm phenotype specificity

The choice of model depends on the research question, with consideration of:

  • Time frame of the study

  • Need for complete versus partial loss of function

  • Specific pathways being investigated

  • Physiological relevance required

For cancer studies, patient-derived xenografts with naturally occurring COMMD1 suppression provide particularly relevant models .

What analytical techniques can differentiate between COMMD1's effects on protein stability versus transcriptional regulation?

Distinguishing between COMMD1's effects on protein stability and transcriptional regulation requires a multi-faceted approach:

For protein stability assessment:

  • Cycloheximide chase assays: Track protein degradation rates with protein synthesis blocked

  • Pulse-chase experiments: Monitor protein turnover using metabolic labeling

  • Proteasome inhibition studies: Determine if protein accumulation under COMMD1 deficiency is proteasome-dependent

  • Ubiquitination assays: Assess changes in substrate protein ubiquitination status

  • Half-life calculations: Quantify protein stability mathematically from degradation curves

For transcriptional regulation assessment:

  • RT-qPCR: Measure mRNA levels of target genes

  • Luciferase reporter assays: Directly assess transcriptional activity using reporter constructs with response elements

  • Chromatin immunoprecipitation (ChIP): Determine transcription factor binding to target gene promoters

  • DNA binding assays: Evaluate the ability of transcription factors to bind DNA elements in the presence/absence of COMMD1

  • Nuclear run-on assays: Measure actual transcription rates rather than steady-state mRNA levels

Integrated approaches:

  • Compare protein levels with corresponding mRNA levels

  • Use translation inhibitors alongside proteasome inhibitors

  • Employ mathematical modeling to distinguish contributions of synthesis versus degradation

  • Consider time-course experiments to capture dynamic effects

For example, when studying COMMD1's effect on HIF pathway, researchers demonstrated through luciferase reporters and DNA binding assays that COMMD1 primarily affects transcriptional activity rather than HIF-1α stability , while confirming unchanged HIF1A mRNA levels through RT-qPCR .

How do researchers reconcile COMMD1's opposing effects on different aggregation-prone proteins?

The paradoxical effects of COMMD1 on different aggregation-prone proteins present a significant research challenge:

  • Conflicting observations: COMMD1 enhances mutant SOD1 aggregation in ALS models but decreases mutant Parkin inclusions in Parkinson's disease models, while having minimal effect on Huntingtin aggregation .

  • Potential explanations:

    • Client-specific recognition: COMMD1 likely recognizes specific structural features unique to each misfolded protein rather than general aggregation propensity .

    • Differential binding modes: The COMMD1 interaction may engage different domains of various client proteins, resulting in distinct conformational changes.

    • Context-dependent cofactors: Different cellular environments and cofactors may modify COMMD1's effect on specific substrates.

    • Subcellular localization differences: The aggregation process for different proteins occurs in distinct cellular compartments where COMMD1 may have varied functions.

    • Aggregation mechanism diversity: SOD1, Parkin, and Huntingtin likely aggregate through different mechanisms, which may explain differential sensitivity to COMMD1.

Methodological approaches to resolve these contradictions:

  • Structural studies of COMMD1-client protein complexes

  • Creation of chimeric aggregation-prone proteins to identify critical domains

  • Proteomic identification of context-specific COMMD1 binding partners

  • Cellular mapping of interaction locations using advanced microscopy

Researchers should design experiments that directly compare multiple client proteins under identical conditions to properly characterize this client-specific behavior, as was done in the seminal study identifying this phenomenon .

What explains the variation in COMMD1's effects under different oxygen conditions?

COMMD1's relationship with oxygen-regulated pathways shows interesting complexities:

  • Paradoxical observations:

    • COMMD1 preferentially binds HIF-1α under normoxic conditions

    • Yet this binding is independent of prolyl hydroxylation (unlike VHL binding to HIF-1α)

    • COMMD1 deficiency has greater effects on HIF activity during normoxia than hypoxia

  • Potential mechanisms explaining oxygen-dependent effects:

    • Oxygen-sensitive conformational changes: Oxygen levels may induce structural changes in HIF-1α that affect COMMD1 binding independently of hydroxylation.

    • Competition with oxygen-regulated factors: Under hypoxia, other proteins may compete more effectively with COMMD1 for HIF-1α binding.

    • Post-translational modifications: Oxygen-dependent modifications beyond prolyl hydroxylation might affect interaction surfaces.

    • Subcellular relocalization: Oxygen levels may change the cellular distribution of COMMD1 or its partners.

    • Indirect regulation through other pathways: Oxygen might affect COMMD1 function through parallel cellular pathways.

Experimental approaches to clarify this relationship:

  • Studies with HIF-1α mutants lacking specific modification sites

  • Protein structure analysis under different oxygen conditions

  • Comprehensive mapping of oxygen-dependent post-translational modifications

  • Investigation of COMMD1 localization under varying oxygen levels

The observed oxygen sensitivity without dependence on the canonical prolyl hydroxylation pathway suggests novel regulatory mechanisms worthy of further investigation .

What therapeutic strategies could target the COMMD1-HIF axis in cancer?

Several promising therapeutic strategies could exploit the COMMD1-HIF relationship in cancer:

  • COMMD1 replacement/enhancement approaches:

    • Gene therapy to restore COMMD1 expression in cancers where it's suppressed

    • Small molecules that stabilize endogenous COMMD1 protein

    • Peptide mimetics that replicate COMMD1's binding to HIF-1α

  • Disruption of HIF-1α/β dimerization:

    • Small molecules targeting the HIF-1α bHLH/PAS domains identified as COMMD1 binding regions

    • Peptides derived from COMMD1's COMM domain that compete with HIF-1β

    • Allosteric modulators that prevent HIF heterodimer formation

  • Combination approaches:

    • Pairing COMMD1-targeted therapies with established anti-angiogenic drugs

    • Combining with therapies targeting other HIF-regulated pathways

    • Sequential treatment strategies based on oxygen conditions in the tumor

  • Biomarker development:

    • COMMD1 expression as a predictive marker for therapy response

    • HIF activity signatures to identify patients most likely to benefit

    • Monitoring COMMD1/HIF status during treatment to detect resistance

These approaches would likely be most effective in cancers demonstrating both COMMD1 suppression and HIF dependence. Careful consideration of potential systemic effects is needed, given COMMD1's roles in copper homeostasis and other pathways .

How might advanced imaging techniques enhance our understanding of COMMD1 dynamics?

Advanced imaging technologies offer powerful approaches to elucidate COMMD1's dynamic behaviors:

  • Super-resolution microscopy techniques:

    • Stimulated emission depletion (STED) microscopy to visualize COMMD1 in specific subcellular compartments

    • Photoactivated localization microscopy (PALM) to track single COMMD1 molecules

    • Stochastic optical reconstruction microscopy (STORM) for precise localization of COMMD1-protein interactions

  • Live-cell imaging applications:

    • FRAP (Fluorescence Recovery After Photobleaching) to measure COMMD1 mobility

    • FRET (Förster Resonance Energy Transfer) to detect real-time interactions with binding partners

    • Optogenetic approaches to control COMMD1 localization or activity with light

  • Multi-color imaging strategies:

    • Simultaneous tracking of COMMD1 with partners like HIF-1α and HIF-1β

    • Visualization of COMMD1 in relation to aggregation-prone proteins

    • Correlation with markers of specific cellular compartments

  • Correlative microscopy:

    • Combining fluorescence imaging with electron microscopy for ultrastructural context

    • Integrated analysis of COMMD1 localization with functional readouts

Such techniques could address critical questions including:

  • How oxygen levels affect COMMD1's subcellular distribution

  • The dynamics of COMMD1 recruitment to protein aggregates

  • Real-time visualization of competition between COMMD1 and HIF-1β for HIF-1α binding

  • Changes in COMMD1 behavior during cancer progression

Early studies showing COMMD1 co-localization with mutant SOD1 inclusions and LDLR demonstrate the value of imaging approaches, which can now be expanded with these advanced technologies.

What systems biology approaches could integrate COMMD1's diverse cellular functions?

Systems biology approaches offer powerful frameworks to understand COMMD1's multifunctional nature:

  • Multi-omics integration:

    • Combine proteomics, transcriptomics, and metabolomics data from COMMD1-deficient models

    • Integrate copper metabolism, HIF pathway, and protein quality control networks

    • Map post-translational modification landscapes affected by COMMD1

  • Network analysis methodologies:

    • Protein-protein interaction network construction centered on COMMD1

    • Pathway enrichment analysis to identify overrepresented functions

    • Network perturbation modeling to predict effects of COMMD1 modulation

  • Computational modeling approaches:

    • Mathematical models of COMMD1's competing interactions with multiple partners

    • Simulation of oxygen-dependent dynamics of COMMD1-HIF interactions

    • Integration of structural data to predict binding interfaces and competition

  • Single-cell technologies:

    • Single-cell transcriptomics to capture cell-specific responses to COMMD1 manipulation

    • Spatial transcriptomics to map COMMD1 effects in tissue contexts

    • Cell-to-cell variability analysis of COMMD1 function

  • Translational bioinformatics:

    • Mining cancer genomics databases for COMMD1 alterations and correlations

    • Patient stratification based on COMMD1 pathway signatures

    • Drug repurposing strategies targeting COMMD1-related pathways

These approaches could help resolve how one protein participates in seemingly disparate functions like copper homeostasis, transcriptional regulation, and protein quality control. The integration of client-specific effects in protein aggregation with pathway-specific effects in cancer progression would be particularly valuable for developing a unified model of COMMD1 function.

Product Science Overview

Introduction

Copper Metabolism Domain Containing 1 (COMMD1) is a protein encoded by the COMMD1 gene in humans. This protein plays a crucial role in various physiological processes, particularly in the regulation of copper metabolism. The recombinant form of COMMD1 is produced using recombinant DNA technology, which allows for the expression of the human COMMD1 protein in a host organism, such as bacteria or yeast.

Gene and Protein Structure

The COMMD1 gene is located on chromosome 2 and is also known by several aliases, including MURR1 and C2orf5 . The protein encoded by this gene contains a conserved COMM domain, which is essential for its function. The COMM domain is involved in protein-protein interactions and is critical for the regulation of various cellular processes .

Function and Mechanism

COMMD1 is a multifunctional protein that is involved in several key cellular processes:

  1. Copper Homeostasis: COMMD1 plays a significant role in maintaining copper homeostasis within cells. It is involved in the regulation of copper transport and storage, ensuring that copper levels remain within a physiologically optimal range .

  2. Ubiquitination and Protein Degradation: COMMD1 is a component of the Cullin-RING ubiquitin ligase (CRL) complexes. It promotes the ubiquitination and subsequent proteasomal degradation of specific target proteins, including the NF-kappa-B subunit RELA . This regulation is crucial for controlling the activity of NF-kappa-B, a transcription factor involved in inflammatory and immune responses .

  3. Membrane Protein Regulation: COMMD1 is involved in the regulation of membrane protein expression and ubiquitination. For example, it regulates the membrane expression of the sodium-potassium-chloride cotransporter SLC12A2 .

Clinical Significance

Mutations or dysregulation of the COMMD1 gene have been associated with several diseases:

  1. Wilson Disease: Wilson disease is a genetic disorder characterized by excessive accumulation of copper in the liver, brain, and other vital organs. COMMD1 is implicated in the pathophysiology of this disease due to its role in copper metabolism .

  2. Ichthyosis, Congenital, Autosomal Recessive 13: This is a rare genetic skin disorder characterized by dry, scaly skin. Mutations in the COMMD1 gene have been linked to this condition .

Research and Therapeutic Potential

Research on COMMD1 has provided insights into its potential therapeutic applications:

  1. Cancer Therapy: The role of COMMD1 in regulating NF-kappa-B activity has implications for cancer therapy. NF-kappa-B is known to promote cell survival and proliferation, and its dysregulation is associated with various cancers. Targeting COMMD1 to modulate NF-kappa-B activity could be a potential therapeutic strategy .

  2. Copper-Related Disorders: Understanding the mechanisms by which COMMD1 regulates copper homeostasis can lead to the development of therapies for copper-related disorders, such as Wilson disease .

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 2024 Thebiotek. All Rights Reserved.