CLIC4 exhibits diverse roles across cellular systems:
Forms redox-regulated chloride channels in lipid bilayers (30 pS conductance) .
Membrane insertion is enhanced under oxidizing conditions, enabling ion transport .
Modulates RhoA and Rac1 GTPase activity in endothelial cells, influencing vascular barrier function .
Binds dynamin I, tubulin, actin, and 14-3-3 isoforms, linking it to cytoskeletal dynamics and vesicular trafficking .
Critical for renal tubule luminogenesis; CLIC4-null mice show dilated proximal tubules and defective apical vesicle fusion .
Enriched in oligodendrocytes, astrocytes, and microglia, with roles in ethanol-induced neuroadaptation .
CLIC4 Human is utilized in studies spanning neuroscience, vascular biology, and cancer:
CLIC4 induction in oligodendrocytes correlates with ethanol sensitivity .
Linked to schizophrenia and addiction via dopamine signaling pathways .
Regulates pulmonary artery hypertension through Arf6 GTPase interaction .
Protects against ischemia-reperfusion injury in cardiac tissue .
CLIC4 is ubiquitously expressed but shows tissue-specific roles:
MGSSHHHHHH SSGLVPRGSH MALSMPLNGL KEEDKEPLIE LFVKAGSDGE SIGNCPFSQR LFMILWLKGV VFSVTTVDLK RKPADLQNLA PGTHPPFITF NSEVKTDVNK IEEFLEEVLC PPKYLKLSPK HPESNTAGMD IFAKFSAYIK NSRPEANEAL ERGLLKTLQK LDEYLNSPLP DEIDENSMED IKFSTRKFLD GNEMTLADCN LLPKLHIVKV VAKKYRNFDI PKEMTGIWRY LTNAYSRDEF TNTCPSDKEV EIAYSDVAKR LTK.
CLIC4 shows preferential localization to mitochondrial-associated membranes (MAMs) rather than pure mitochondrial fractions. Quantitative colocalization studies demonstrate CLIC4 has higher association with mitochondria (41.2 ± 0.6%, n=40 cells) and ER-mitochondria contact site proteins, including mitofusin 2 (34 ± 1%, n=25 cells) and ACSL4 (31 ± 2%, n=25 cells). In isolated crude mitochondria, CLIC4 shows 58 ± 10% association with mitochondria, while in neonatal cardiomyocytes it shows 40 ± 4% (n=28) association with MitoTracker-loaded mitochondria .
CLIC4 demonstrates significantly higher mitochondrial colocalization compared to its paralog CLIC1. Specifically, CLIC1 shows lower colocalization to crude mitochondria (39 ± 8%, P=0.03) and neonatal cardiomyocytes (29 ± 3%, P=0.03, n=28) in comparison to CLIC4. Unlike CLIC1, which is primarily localized in the endoplasmic reticulum, CLIC4's predominant localization to MAMs appears to give it a distinct role in modulating mitochondrial physiology and cellular response to injury .
For precise CLIC4 localization studies, a multi-technique approach is recommended. The most reliable methodology involves:
Immunofluorescence with colocalization analysis using confocal microscopy (with organelle-specific markers like MitoTracker for mitochondria)
Subcellular fractionation using differential centrifugation
Purification of mitochondria using 30% Percoll gradient centrifugation to separate pure mitochondria (M3 fraction) from MAM fractions
Western blot analysis of the various fractions using organelle-specific markers (e.g., GRP78 for ER/MAM)
This combined approach has revealed CLIC4's negligible presence in ultrapure mitochondrial fractions but significant presence in MAMs, establishing it as a MAM-specific Cl⁻ channel .
The recommended approach for generating CLIC4 constructs involves:
Using the pCDNA3.1-clic4 vector encoding N-terminal FLAG as a template
Implementing site-directed mutagenesis via the "Quick change" method with specific primers (see table below)
Using DH5α competent cells for transformation
Screening positive colonies by Sanger sequencing
For protein expression and purification, researchers have successfully used:
Human CLIC4 gene cloned into a pET-28a vector with N-terminal hexahistidine tag and TEV cleavage site
Expression in C43 E.coli strain using M9 minimal media
CLIC4 functions as a tumor suppressor involved in processes including growth arrest, differentiation, and apoptosis. Its expression is diminished in tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms. Importantly, this downregulation is not driven by genomic alterations but instead by post-transcriptional mechanisms. Screening and functional assays have identified miR-142-3p as a key regulator of CLIC4. Expression analysis reveals that CLIC4 and miR-142-3p are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers .
For researchers studying the complex tumor microenvironment, a combination of advanced techniques is required:
In situ hybridization to localize miR-142-3p expression within tissue sections
Single-cell RNA-sequencing to identify cell-type specific expression patterns
Immunohistochemical analysis with cell-type specific markers
These combined approaches have revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in normal epithelium, providing crucial insights into the cell-type specific regulation of CLIC4 in cancer progression .
Sex-specific differences in CLIC4 response to ethanol have been documented using qRT-PCR analysis of the medial prefrontal cortex (mPFC) in C57BL/6J mice. Following acute ethanol exposure, female mice show a rapid increase in Clic4 mRNA expression that is notably absent in male mice. This sex-specific response suggests potential differences in how male and female brains process acute ethanol exposure at the molecular level .
Immunohistochemical and stereological techniques have revealed that CLIC4 protein expression in brain tissue follows this pattern of abundance:
Oligodendrocytes (most abundant)
Microglia
Astrocytes
Neurons (minimal expression)
For comprehensive analysis of CLIC4-expressing cell populations in brain tissue, researchers should employ:
Dual immunohistochemical labeling with cell-type specific markers
Stereological counting methods for quantification
qRT-PCR for mRNA expression analysis before and after experimental interventions
This methodological approach has established that oligodendrocytes likely play a key role in mediating CLIC4's response to ethanol in the brain .
CLIC4 plays a critical cardioprotective role through its localization to mitochondrial-associated membranes (MAMs). Experimental evidence using CLIC4 null cardiomyocytes demonstrated increased apoptosis and mitochondrial dysfunction when subjected to hypoxia-reoxygenation injury compared to wild-type cardiomyocytes. This indicates CLIC4's essential role in maintaining mitochondrial function during cardiac stress conditions .
Heart failure induces complex changes in CLIC4 expression patterns that require sophisticated analysis techniques:
These observations suggest that reduced internalization of soluble CLIC4 to organelle membranes like MAMs may contribute to decreased functional activity in the intracellular membrane fraction, potentially contributing to heart failure pathology .
The crystal structure availability of soluble CLIC4 allows for sophisticated structure-based drug discovery approaches. A comprehensive methodology includes:
Utilizing multiple predictive algorithms to identify consensus allosteric sites:
SiteMap
DeepSite
FTMap
DogSiteScore
ProBIS
Characterizing potential binding pockets based on:
Surface exposure
Hydrophobicity
Hydrophilicity
Druggability (measured by Halgren's and DogSite scores)
Performing virtual screening with blind docking using AutoDock Vina integrated in high-performance computing clusters
Filtering compounds based on binding energies, non-preferential binding to GSH sites, and pharmacological relevance
For rigorous validation of potential CLIC4 inhibitors identified through computational screening, researchers should implement:
Recombinant protein expression in appropriate bacterial systems (e.g., C43 E.coli strain)
Protein purification via affinity chromatography with Ni IMAC
In vitro binding and functional assays to confirm inhibitor interaction and efficacy
GPU-accelerated molecular dynamics simulations to investigate allosteric inhibitory mechanisms
This combined computational and experimental approach has successfully identified new molecules with improved specificity compared to traditional non-selective CLIC inhibitors like IAA94 and A9C .
Chloride ion channels, including CLIC4, play critical roles in modulating immunological interactions. Research indicates that chloride flux may utilize distinct signaling pathways to execute specific functions in cellular context-dependent manners. The CLIC family located in various cellular organelles is involved in diverse physiological functions and pathological conditions, including immune response regulation .
While specific experimental models aren't detailed in the search results, emerging research suggests that studying CLIC4's role in immunomodulation requires:
Cell-type specific conditional knockout models
Flow cytometry analysis of immune cell populations
Cytokine profiling in response to inflammatory stimuli
Examination of CLIC4 expression in immune cell subsets under various inflammatory conditions
Understanding CLIC4's role in immune signaling pathways may provide insights into novel therapeutic approaches for inflammatory and immune-mediated diseases .
Chloride Intracellular Channel 4 (CLIC4), also known as p64H1, is a member of the chloride intracellular channel (CLIC) family. These proteins are unique in that they can exist both in soluble and membrane-bound forms, allowing them to participate in a variety of cellular processes. CLIC4 is encoded by the CLIC4 gene and is expressed in many tissues throughout the body .
CLIC4 is a protein consisting of 253 amino acids . It has a single transmembrane domain, which allows it to insert into cellular membranes and form ion channels. This insertion is redox-regulated, meaning it occurs under oxidizing conditions . In its soluble state, CLIC4 can catalyze thiol-disulfide exchange reactions, similar to glutaredoxins, using reduced glutathione as an electron donor .
CLIC4 plays several critical roles within cells:
CLIC4 is associated with various diseases and conditions: