Property | Details |
---|---|
Molecular Weight | 29 kDa |
Amino Acid Sequence | 241 residues + 20-amino acid His tag |
Isoelectric Point (pI) | ~5.0 |
Solubility | 20 mM Tris-HCl, 0.1M NaCl, 10% glycerol |
CLIC1 regulates cellular processes through chloride ion flux and protein interactions:
Phagosomal Acidification: Localizes to phagosomal membranes in macrophages, enabling acidification critical for pathogen degradation .
Cell Cycle and Apoptosis:
Cancer Metastasis: Promotes migration/invasion in ESCC via EMT pathway modulation (e.g., downregulates CDH1, upregulates VIM) .
Inflammatory Disease: CLIC1−/− mice show resistance to K/BxN arthritis, linking it to macrophage-driven inflammation .
CLIC1 expression correlates with CD8+ T cells (r = 0.159) and monocytes (r = 0.324) in LUAD, suggesting immune microenvironment modulation .
Inhibitor Development: NSC602247 binds CLIC1’s active site, blocking ion conductance (Ki = 2.3 µM) .
Autoimmune Disease Target: CLIC1 inhibition reduces ROS and Ca²⁺ signaling in A549 lung cancer cells, enhancing apoptosis .
Mechanistic Studies: Clarify CLIC1’s role in charge compensation during NADPH oxidase activity .
Clinical Trials: Evaluate NSC602247 analogs for anti-inflammatory or anti-cancer efficacy.
CLIC1 Human exemplifies a multifunctional protein bridging ion channel activity, redox biology, and disease mechanisms. Its dual soluble/membrane states and clinical associations position it as a pivotal target for precision therapeutics.
CLIC1 is a member of the chloride intracellular channel protein family characterized by its remarkable metamorphic properties. Unlike conventional ion channels, CLIC1 exists primarily as a soluble cytoplasmic protein but can undergo a substantial structural rearrangement to insert into membranes and form functional chloride-selective ion channels .
This structural transition represents a paradigm shift from traditional membrane proteins, which typically maintain fixed configurations. The protein contains a single putative transmembrane region with only two charged residues (arginine 29 and lysine 37), which play crucial roles in regulating its ion channel activity . The GST-like canonical fold in its soluble form further distinguishes CLIC1 from conventional ion channels, making it a fascinating subject for structural biology research .
To study this metamorphic nature, researchers should employ complementary techniques:
X-ray crystallography for soluble form characterization
Fluorescence spectroscopy to monitor membrane interactions
Electrophysiological recordings to assess channel functionality
Molecular dynamics simulations to model transitional states
CLIC1's transition from soluble cytoplasmic protein to membrane-inserted ion channel is regulated by specific environmental triggers:
pH Modulation: Decreased cytosolic pH promotes CLIC1 membrane insertion
Oxidative Conditions: Oxidation drives CLIC1's interaction with lipid bilayers
Membrane Composition: Certain lipid environments facilitate insertion
Cell Cycle Phase: Temporal regulation during G1-S transition
Methodologically, researchers can experimentally control these factors through:
Precise pH buffers in reconstitution experiments
Controlled oxidizing agents like H₂O₂ or diamide
Defined lipid compositions in artificial bilayers
Cell synchronization techniques for cell cycle studies
The strongest experimental evidence supports a fluorescence resonance energy transfer (FRET) distance of approximately 15 Å between CLIC1's tryptophan residue (Trp35) and the membrane surface under oxidizing conditions, suggesting Trp35 serves as a membrane anchor during insertion .
CLIC1 serves as a critical regulator of cancer cell survival and proliferation through multiple interconnected mechanisms:
Cell Cycle Regulation: CLIC1 controls G1-S phase transition in cancer stem cells (CSCs), particularly in glioblastoma (GB). Inhibition of CLIC1 significantly arrests GB CSCs in G1 phase of the cell cycle, as demonstrated by flow cytometry analysis of DNA content distribution .
ROS-pH-CLIC1 Axis: A complex interplay exists between:
CLIC1 membrane localization
Intracellular reactive oxygen species (ROS) accumulation
Cytoplasmic pH modulation
This regulatory network creates an "allostatic tumorigenic condition" that promotes cancer cell proliferation. During G1-S transition, ROS production increases in parallel with increasing CLIC1-associated membrane current, followed by a cytosolic alkalinization peak .
Calcium Signaling: CLIC1 regulates intracellular Ca²⁺ levels, particularly in lung cancer. Knockdown of CLIC1 in A549 human lung cancer cells increases basal Ca²⁺ levels via L-type Ca²⁺ channels (LTCCs), triggering excessive ROS production and JNK activation .
For experimental investigation, researchers should employ:
CLIC1 inhibitors (IAA94) or knockdown approaches
Time-resolved measurements of membrane currents, ROS levels, and pH
Calcium imaging with specific chelators (BAPTA-AM)
Cell cycle synchronization protocols
CLIC1 functions as a critical node in the regulation of both cellular redox state and pH homeostasis through several interconnected mechanisms:
Redox Sensing and Response:
pH Modulation:
Integrated Signaling:
ROS, pH, and CLIC1 membrane expression are temporally linked during the G1-S transition
Inhibiting CLIC1-mediated chloride current prevents both intracellular ROS accumulation and pH changes
Experimental approaches should include:
Simultaneous measurement of CLIC1 membrane localization, intracellular ROS, and pH
Selective inhibition of CLIC1, NADPH oxidase, and NHE1 proton pump
Time-lapse microscopy to track dynamic changes during cell cycle progression
![CLIC1-ROS-pH relationship in cancer cells reveals a mechanism where CLIC1 membrane permeability stabilizes an allostatic state, distinguishing it from homeostatic conditions in normal cells]
Multiple complementary electrophysiological approaches are recommended for comprehensive characterization of CLIC1 channel activity:
Each method offers distinct advantages and limitations. Research protocols should include controls for:
Oxidation state management
pH standardization
Membrane/lipid composition consistency
Verification of CLIC1 insertion using multiple techniques
To effectively manipulate and study CLIC1 function in cellular systems, researchers should employ a multi-faceted approach:
Genetic Manipulation Strategies:
shRNA knockdown: Demonstrated to drastically compromise cell growth in cancer stem cells
CRISPR-Cas9 gene editing: For complete knockout or targeted mutations
Site-directed mutagenesis: Particularly at key residues (R29, K37) that alter channel properties
Overexpression systems: With fluorescent tags for localization studies
Pharmacological Interventions:
Experimental Validation Approaches:
Western blot analysis of cyclin D1 and p27 expression to establish G1-S transition timing
Flow cytometry for cell cycle analysis under CLIC1 inhibition conditions
Combined electrophysiology and imaging to correlate localization with function
Calcium flux measurements with specific blockers (nifedipine for LTCCs)
Controls and Verification:
Comparing effects on cancer cells versus normal counterparts (e.g., mesenchymal stem cells show no CLIC1-associated chloride permeability)
Assessing specificity using multiple inhibition approaches
Time-course studies to capture dynamic changes
Rescue experiments to confirm specificity of observed effects
The transmembrane region of CLIC1 contains only two charged residues—arginine 29 (Arg29) and lysine 37 (Lys37)—which play distinct roles in regulating its ion channel function:
K37A Mutation Effects:
R29A Mutation Effects:
These findings were established through multiple complementary electrophysiological approaches:
Recombinant protein studies in artificial bilayers
Cell-attached patch clamp in transfected HEK cells
Whole-cell recordings in heterologous expression systems
For researchers pursuing mutation studies, recommended methodological considerations include:
Systematic alanine scanning of the transmembrane region
Charge-conserving versus charge-neutralizing substitutions
Complementary structural studies (e.g., cysteine accessibility)
Correlation between biophysical changes and cellular function
The metamorphic transition of CLIC1 from a soluble cytoplasmic protein to a membrane-associated ion channel involves a complex molecular mechanism that remains partially understood:
Structural Rearrangement:
Triggering Factors and Sequence:
Oxidizing conditions promote a structural transition exposing hydrophobic surfaces
FRET experiments detect strong energy transfer between Trp35 and dansyl-lipid analogues on membrane surfaces
The calculated FRET distance between Trp35 and the membrane surface is approximately 15 Å
pH changes may work cooperatively with redox state to facilitate membrane insertion
Oligomerization State:
Lipid Interactions:
Specific lipid compositions may facilitate insertion
Interactions with membrane components likely stabilize the transmembrane configuration
The C-terminal region's membrane localization appears important for certain functions
Research approaches to further elucidate this mechanism should include:
High-resolution structural studies of membrane-associated forms
Real-time monitoring of the transition process
Molecular dynamics simulations of the insertion mechanism
Identification of intermediate states in the metamorphic process
CLIC1's role in cancer extends beyond its ion channel function, involving complex interactions with multiple signaling networks:
Cell Cycle Regulatory Pathways:
ROS-Mediated Signaling:
Calcium Signaling Integration:
pH Homeostasis Connection:
For comprehensive investigation of these pathway interactions, researchers should consider:
Multi-omics approaches to identify CLIC1 interaction partners
Temporal analysis of signaling events during cell cycle progression
Pharmacological and genetic manipulation of individual pathway components
Correlation of CLIC1 expression/function with clinical outcomes in cancer patients
Several therapeutic approaches targeting CLIC1 show significant promise for cancer treatment:
Direct CLIC1 Inhibitors:
IAA94: Selective CLIC1 channel blocker that arrests cancer stem cells in G1 phase
Custom antibodies: Directed against extracellular portions of membrane-inserted CLIC1
Metformin: Repurposed antidiabetic drug with proposed CLIC1 inhibitory effects
Novel small molecule inhibitors: Rational design based on structure-function relationships
Advantages of CLIC1 as a Therapeutic Target:
Cancer-specific expression and function (e.g., no CLIC1-associated chloride permeability in mesenchymal stem cells)
Potential to reset cancer cells from an "allostatic tumorigenic condition" to homeostasis
Minimal side effects compared to inhibiting NADPH oxidase or NHE1 proton pump
Unique transition between soluble and membrane forms provides targeting specificity
Combinatorial Approaches:
CLIC1 inhibition with redox modulators
Co-targeting calcium signaling pathways
Integration with conventional chemotherapy
Cancer stem cell-directed therapies
Delivery Strategies:
Nanoparticle-based delivery systems
Blood-brain barrier penetrating formulations (particularly for glioblastoma)
Tumor-targeting approaches
Controlled release mechanisms
The most encouraging evidence comes from studies showing that CLIC1 inhibition impairs tumor growth in vivo and selectively affects cancer stem cells while sparing normal cellular counterparts .
Researchers face several significant technical challenges when studying CLIC1, each requiring specific methodological approaches:
Distinguishing Membrane vs. Cytosolic CLIC1:
Challenge: CLIC1 exists in both forms simultaneously
Solutions:
Surface biotinylation followed by Western blotting
TIRF microscopy for selective membrane visualization
Electrophysiological recordings with ion channel blockers
Subcellular fractionation with careful controls
Monitoring Dynamic CLIC1 Transitions:
Ensuring Specificity of CLIC1 Inhibitors:
Challenge: Potential off-target effects of inhibitors
Solutions:
Multiple complementary inhibition approaches (pharmacological and genetic)
Structure-activity relationship studies for inhibitor optimization
Rescue experiments with inhibitor-resistant CLIC1 variants
Comparative studies in cells lacking CLIC1 expression
Reproducing Physiological Membrane Environments:
Challenge: Artificial systems may not recapitulate native membrane properties
Solutions:
Defined lipid compositions mimicking target membranes
Giant unilamellar vesicles with controlled compositions
Native membrane preparations
Advanced microscopy of membrane microdomains
Researchers should implement rigorous controls and validation approaches, including:
Multiple complementary techniques to confirm findings
Careful consideration of experimental conditions (pH, redox state)
Appropriate statistical analysis for electrophysiological data
Verification in multiple cell types and experimental systems
CLIC1 is unique among chloride channels because it can exist both as a soluble cytoplasmic protein and as an integral membrane protein. This dual existence allows CLIC1 to participate in a variety of cellular functions. Structurally, CLIC1 is related to the glutathione S-transferase (GST) superfamily and contains an approximately 240 amino acid sequence at the C-terminus .
The protein can form ion channels in cellular membranes, facilitating the rapid passage of chloride ions across these membranes. This function is essential for maintaining the proper electrical charge and ion balance within cells .
CLIC1 is involved in several critical cellular processes:
Recombinant CLIC1 refers to the protein produced through recombinant DNA technology. This involves inserting the CLIC1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant CLIC1 is used in various research applications to study its structure, function, and role in different cellular processes.
The study of CLIC1 has significant clinical implications. For instance, targeting CLIC1 activity has been explored as a potential therapeutic strategy for treating glioblastoma, a type of brain cancer. Inhibitors of CLIC1 have shown promise in reducing the proliferation and invasiveness of glioblastoma stem cells .