Clusterin’s roles include:
Complement regulation: Inhibits complement-mediated cell lysis .
Apoptosis modulation: Inhibits apoptosis via BAX inhibition and NF-κB pathway modulation .
Lipid transport: Binds lipids, hormones (e.g., leptin), and amyloid-beta (Aβ) .
Chaperone activity: Stabilizes misfolded extracellular proteins .
Aβ Binding: Clusterin binds Aβ, influencing its aggregation and clearance. Recombinant His-tagged clusterin is used to study this interaction in vitro, though glycosylation deficits may alter binding kinetics .
Genetic Risk: CLU polymorphisms (e.g., rs11136000) are linked to AD risk, with clusterin implicated in Aβ toxicity and neurodegeneration .
Survival Signaling: Clusterin inhibits apoptosis via BAX and NF-κB pathways, promoting cancer cell survival .
Therapeutic Target: Silencing CLU enhances chemotherapy efficacy in preclinical models .
Clusterin is expressed in diverse tissues, including:
Note: Expression varies by developmental stage and pathological state .
Expression System: Bacterial expression ensures high yield but lacks mammalian post-translational modifications .
Purification: His-tag enables nickel/nitrilotriacetic acid (Ni-NTA) affinity chromatography .
Parameter | Specification | Source |
---|---|---|
Purity | >95% (SDS-PAGE confirmed) | |
Activity | Functional in binding assays (e.g., Aβ) | |
Storage | -20°C (lyophilized) or -80°C (reconstituted) |
Lack of Glycosylation: Absence of carbohydrate groups may reduce binding affinity to certain ligands (e.g., Aβ) .
Single-Chain Structure: Unlike native heterodimers, the recombinant form may not replicate native conformational dynamics .
Species-Specific Effects: Human clusterin shares ~70–80% homology with rodent variants, necessitating caution in cross-species studies .
Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone that is normally secreted from cells. It serves multiple physiological functions including:
Maintenance of protein homeostasis (proteostasis)
Inhibition of cell death pathways
Modulation of pro-survival signaling
Regulation of transcriptional networks
This multifunctional glycoprotein is upregulated in various pathological conditions, most notably Alzheimer's disease and numerous cancer types. The protein's chaperone activity helps prevent protein aggregation and maintains cellular integrity during stress conditions .
Clusterin exists in multiple forms with distinct subcellular localizations and opposing functions:
Secreted Clusterin (sCLU): The primary form that undergoes glycosylation and is secreted from cells. It demonstrates predominantly pro-survival functions by stabilizing the BAX-Ku70 complex, which inhibits BAX translocation to mitochondria and prevents apoptotic cascades.
Intracellular Clusterin (iCLU): A truncated, non-glycosylated form that accumulates within cells. This variant is associated with pro-apoptotic functions as it competes with BAX for binding to Ku70, releasing BAX to translocate to mitochondria and promote cell death.
The balance between these forms appears to be critical for determining whether cells survive or undergo apoptosis . Research indicates that stress conditions can trigger alternative splicing of CLU mRNA, favoring the production of intracellular forms with distinct functional properties .
Clusterin expression is regulated through multiple complex mechanisms:
Transcriptional regulation: Various transcription factors influence CLU expression in a tissue-specific manner.
Epigenetic modifications: DNA methylation plays a significant role, with hypermethylation often silencing CLU expression. Studies have demonstrated that DNA demethylation by 5-aza-2′-deoxycytidine increases CLU expression in prostate cancer cell lines .
Histone modifications: In colon cancer cell lines, CLU is predominantly regulated by histone modifications such as H3K9me3 and H3K4me3, which can significantly alter nuclear clusterin expression .
Pharmacological modulation: Treatment with HDAC inhibitors like valproic acid and Vorinostat induces CLU expression and increases clusterin secretion in human astrocytes .
This multi-layered regulation indicates that CLU expression is highly context-dependent, responding to diverse intracellular and extracellular signals in a cell- and tissue-specific manner .
Clusterin plays a complex role in Alzheimer's disease (AD) pathophysiology through several mechanisms:
Genetic association: CLU is now recognized as the third greatest genetic risk factor for late-onset Alzheimer's Disease (LOAD), after APOE and BIN1. Multiple single nucleotide polymorphisms (SNPs) have been identified as susceptibility loci in genome-wide association studies .
Amyloid interactions: Clusterin colocalizes with amyloid beta (Aβ) plaques in the AD brain, particularly in the hippocampus and cortex . This interaction may influence Aβ aggregation and clearance.
Altered distribution: AD-associated mutations can alter the distribution of clusterin inside and outside cells, increasing intracellular clusterin while reducing secreted clusterin .
Neurotoxicity mediation: Knockdown studies have demonstrated that CLU silencing provides protection from Aβ-induced neurotoxicity in both rodent neurons and iPSC-derived neurons, suggesting clusterin mediates Aβ toxicity .
Immunohistochemical colocalization studies with amyloid plaques using both monoclonal and polyclonal antibodies for validation
RT-qPCR for quantifying CLU mRNA expression in different brain regions
CRISPR-based studies for introducing or correcting specific variants to establish causality
CSF and plasma biomarker measurements to correlate with disease progression
Researchers should combine these approaches while accounting for the distinct forms of clusterin when investigating its role in AD pathophysiology .
Studying clusterin in traumatic brain injury (TBI) presents several experimental challenges:
Temporal dynamics complexity:
Challenge: Clusterin expression shows variable timing of peak expression across different brain regions after TBI.
Solution: Implement comprehensive time-course studies covering both acute (hours-days) and chronic (weeks-months) phases, with sampling at multiple time points (2-6 hours, 1-3 days, 5 days-1 week, 2 weeks-1 month, 3 months, and 6 months post-injury) .
Contradictory plasma and brain expression patterns:
Challenge: Despite increased brain expression, plasma clusterin levels are acutely down-regulated post-TBI rather than elevated .
Solution: Always pair plasma measurements with brain tissue analysis when possible, and establish appropriate dilution protocols for ELISA assays (1:500-1:2000 dilution has been confirmed as the optimal range) .
Inconsistent cellular localization findings:
Challenge: Reports of clusterin's cellular localization vary between studies, with some showing neuronal/glial colocalization and others finding primarily extracellular distribution .
Solution: Use multiple antibodies (both monoclonal and polyclonal) and analyze colocalization with multiple cellular markers (NeuN, GFAP, CD68, OX-42) to comprehensively assess distribution .
Model-specific differences:
For optimal experimental design, researchers should implement a multi-method approach combining protein expression analysis, mRNA quantification, and careful control for potential confounding factors such as haemolysis in blood samples .
Differentiating between secreted and intracellular clusterin forms presents significant technical challenges but is crucial for understanding CLU's dual functionality. Recommended methodological approaches include:
Antibody selection and validation:
Use antibodies that specifically recognize either the uncleaved, full-length protein (secreted form) or the nuclear/cytoplasmic variants (intracellular forms)
Confirm specificity through western blotting with recombinant proteins and knockout controls
Subcellular fractionation techniques:
Implement differential centrifugation to separate cytosolic, nuclear, and membrane/secretory fractions
Verify fraction purity using established markers for each compartment
Glycosylation analysis:
Utilize endoglycosidase treatments (PNGase F) to distinguish glycosylated (secreted) from non-glycosylated (intracellular) forms
Combine with western blotting to visualize mobility shifts
Cleavage-specific detection:
Design assays targeting the α-β cleavage site present only in mature secreted clusterin
Use antibodies recognizing neoepitopes created by proteolytic processing
Fluorescent tagging strategies:
Generate constructs with different fluorophores tagging N-terminal and C-terminal regions
Monitor intracellular trafficking and secretion through live-cell imaging
These approaches should be used in combination, as relying on a single method may lead to misinterpretation of results, particularly given the stress-induced alternative splicing that can shift production between different clusterin forms .
Current analytical approaches for investigating clusterin in cancer encompass multiple complementary techniques:
Genetic mutation analysis:
Expression correlation studies:
Protein interaction networks:
Comparative transcriptomics:
Cross-cancer comparisons:
Different cancer types show notable variations in CLU's expression patterns and prognostic significance. Specific analytical tools like TIMER2 have been employed to examine correlations between CLU expression and immune infiltrates across all TCGA tumor types. The analyses typically utilize various computational approaches to characterize immunological infiltrates, including algorithms such as TIMER, CIBERSORT, and QUANTISEQ .
For comprehensive analysis, researchers should combine these approaches while accounting for cancer-specific contexts and potential confounding factors.
When designing experiments to study clusterin in cell culture systems, researchers should consider several critical parameters:
Cell line selection:
Endogenous expression: Choose cell lines with appropriate baseline expression of clusterin (high for knockdown studies, low for overexpression)
Tissue relevance: Select cells derived from tissues where clusterin plays established roles (neurons, astrocytes, cancer cell lines)
Response capacity: Ensure cells can modulate clusterin expression under stress conditions
Stress induction protocols:
Oxidative stress: Hydrogen peroxide (100-500 μM) or paraquat (10-100 μM) for 6-24 hours
ER stress: Tunicamycin (1-5 μg/ml) or thapsigargin (0.1-1 μM) for 12-48 hours
Heat shock: 42°C for 30-60 minutes followed by recovery at 37°C
Genotoxic stress: Cisplatin (10-50 μM) or UV irradiation (10-50 J/m²)
Expression modulation:
Detection methods optimization:
Western blotting: Use gradient gels (4-20%) to effectively separate differently processed forms
ELISA: Validate linearity within an appropriate dilution range (1:500-1:2000 for plasma samples)
Immunofluorescence: Include permeabilized and non-permeabilized conditions to distinguish secreted vs. intracellular forms
These parameters should be systematically optimized for each experimental system, with appropriate controls for stress responses and cellular viability.
The apparent contradictory findings regarding clusterin's roles in neuroprotection and neurotoxicity can be resolved through careful experimental design that accounts for several factors:
Form-specific analysis:
Explicitly distinguish between secreted clusterin (generally neuroprotective) and intracellular clusterin (potentially neurotoxic)
Utilize antibodies and detection methods that can differentiate between these forms
Design experiments that manipulate the ratio between forms rather than total clusterin levels
Context-dependent interpretation:
Consider the specific neurodegenerative context (AD, TBI, stroke, etc.)
Account for the temporal progression of pathology
Evaluate the presence of specific binding partners that may alter clusterin function
Concentration-dependent effects:
Implement dose-response studies with physiologically relevant concentration ranges
Correlate clusterin concentrations with functional outcomes
Consider threshold effects where protective functions may shift to detrimental ones
Integration of in vitro and in vivo findings:
Compare results from cell culture, animal models, and human studies
Validate key findings across multiple experimental systems
Develop unified models that explain seemingly contradictory observations
Mechanistic dissection:
By systematically addressing these factors, researchers can develop a more nuanced understanding of how clusterin functions in the nervous system, recognizing that its effects are highly dependent on its form, concentration, cellular context, and disease stage.
Accurate measurement of clusterin in biological fluids requires careful sample preparation, with protocols that must be tailored to the specific fluid being analyzed:
Collection and processing:
Collect blood in EDTA tubes for plasma or serum separator tubes for serum
Process within 2 hours of collection (centrifuge at 2000×g for 10 minutes at 4°C)
Aliquot to avoid freeze-thaw cycles and store at -80°C
Pre-analysis handling:
Quality control:
Collection and processing:
Collect via lumbar puncture using standardized protocols
Centrifuge immediately (2000×g for 10 minutes) to remove cellular components
Transfer to polypropylene tubes to prevent protein binding to tube walls
Pre-analysis handling:
Dilute less than plasma samples (typically 1:10-1:50)
Avoid repeated freeze-thaw cycles
Document time from collection to freezing
Quality control:
Check for blood contamination (measure hemoglobin or cell counts)
Document collection gradient (first vs. last tube)
Consider circadian variations in collection timing
For both fluid types, researchers should validate the detection method, establish reference ranges for their specific population, and document all pre-analytical variables that might influence results.
Interpreting changes in clusterin expression across brain regions following traumatic injury requires a nuanced approach:
Regional and temporal specificity:
Recognize that peak expression timing varies significantly between brain regions
In the perilesional cortex, significant upregulation begins within 1 week post-injury
In the thalamus, clusterin expression peaks at around 3 months
In the dentate gyrus, expression increases gradually from 1 week to 3 months
Correlation with pathological processes:
Localization significance:
Extracellular clusterin (as opposed to neuronal or glial expression) suggests involvement in the extracellular matrix remodeling and debris clearance
Presence in select axonal pathways indicates potential roles in axonal transport or repair mechanisms
Absence in contralateral regions confirms specificity to injury response
Translation between models and humans:
When designing studies or interpreting results, researchers should account for these spatiotemporal patterns and consider how they relate to the specific pathophysiological processes occurring in each region and time point after injury.
When analyzing correlations between clusterin genetic variations and disease outcomes, researchers should employ robust statistical methods tailored to genetic association studies:
Genome-Wide Association Studies (GWAS) analysis:
Use logistic regression models adjusting for age, sex, and population stratification
Implement Bonferroni or false discovery rate (FDR) corrections for multiple testing
Calculate odds ratios (OR) with 95% confidence intervals for risk assessment
Consider advanced approaches like polygenic risk scores that incorporate multiple CLU variants
Survival analysis techniques:
Receiver Operating Characteristic (ROC) analysis:
Calculate area under the ROC curve (AUC) to assess diagnostic potential
Determine optimal cut-off values balancing sensitivity and specificity
Example: For plasma clusterin in TBI, ROC analysis revealed an AUC of 0.851 with a clusterin concentration cut-off of 1.22 resulting in 83% sensitivity and 74% specificity
Correlation analysis for biomarker studies:
Advanced genetic analysis methods:
These statistical approaches should be clearly described in methods sections, with appropriate justification for the chosen tests and transparency regarding any data transformations or outlier handling.
Integrating clusterin expression data with other -omics datasets requires systematic approaches to reveal comprehensive systems-level insights:
Multi-omics correlation analysis:
Network-based integration:
Pathway enrichment approaches:
Machine learning integration models:
Use supervised learning to identify patterns across multi-omics datasets
Implement dimensionality reduction techniques (PCA, t-SNE) for visualization
Develop predictive models incorporating CLU expression with other molecular features
Validate models through cross-validation and external datasets
Data visualization strategies:
By systematically implementing these integration approaches, researchers can move beyond single-gene analyses to understand how clusterin functions within broader molecular networks and biological systems.
Several innovative therapeutic approaches targeting clusterin are emerging in neurodegenerative disease research:
Form-specific targeting strategies:
Antisense oligonucleotides (ASOs): Designed to selectively reduce intracellular clusterin while preserving secreted forms
Small molecule modulators: Compounds that alter the balance between secreted and intracellular clusterin
Antibody-based approaches: Antibodies that recognize specific domains or conformations of clusterin to modulate its function
Pathway-based interventions:
Clusterin-Aβ interaction inhibitors: Molecules that disrupt the binding between clusterin and amyloid beta to prevent co-deposition
BAX-Ku70-clusterin modulators: Compounds that influence apoptotic pathways by targeting this key interaction
DNA-PK complex regulators: Agents that modify clusterin's interaction with DNA repair mechanisms
Gene therapy approaches:
CRISPR-based correction: Targeted editing of disease-associated CLU variants
Viral vector delivery: AAV-mediated expression of beneficial clusterin variants
Exosome-based delivery: Engineered exosomes carrying therapeutic clusterin forms
Biomarker-guided personalized therapies:
These approaches represent promising avenues for therapeutic development, with early evidence suggesting that modulating clusterin's form and function could potentially mitigate neurodegenerative processes by addressing both protein homeostasis disruption and aberrant apoptotic signaling.
Single-cell analytical approaches offer unprecedented opportunities to dissect clusterin's diverse cellular functions across different cell types and disease states:
Single-cell RNA sequencing (scRNA-seq) applications:
Cell type-specific expression profiling: Identifying which cell populations express CLU at baseline and during disease
Pseudotime trajectory analysis: Mapping how CLU expression changes during cellular differentiation or disease progression
Spatial transcriptomics integration: Correlating CLU expression with anatomical location in tissue sections
Alternative splicing detection: Identifying cell populations that preferentially express specific CLU transcript variants
Single-cell proteomics approaches:
Mass cytometry (CyTOF): Simultaneous detection of clusterin and other proteins at single-cell resolution
Imaging mass cytometry: Spatial mapping of clusterin protein variants in tissue sections
Proximity ligation assays: Detecting protein-protein interactions involving clusterin in situ
Single-cell Western blotting: Quantifying different clusterin forms within individual cells
Live-cell imaging strategies:
Fluorescent protein tagging: Monitoring clusterin trafficking between cellular compartments
Photoactivatable probes: Tracking clusterin movement following activation
FRET-based sensors: Detecting clusterin conformational changes and interactions
Optogenetic manipulation: Controlling clusterin function with light-activated domains
Integrative single-cell multi-omics:
CITE-seq: Simultaneous measurement of clusterin mRNA and surface proteins
ATAC-seq with scRNA-seq: Correlating chromatin accessibility with CLU expression
G&T-seq: Linking genomic variants to CLU transcription at single-cell level
REAP-seq: Profiling CLU protein expression alongside surface epitopes
These advanced single-cell approaches will help resolve conflicting findings by revealing how clusterin's expression, localization, and function vary across different cell types and under different pathophysiological conditions.
Advanced computational modeling approaches offer powerful tools for predicting clusterin's interactions with various binding partners across disease contexts:
Structural modeling techniques:
Homology modeling: Generating 3D models of clusterin domains based on related proteins
Molecular dynamics simulations: Examining conformational changes under different conditions
Protein-protein docking: Predicting binding interfaces between clusterin and partners like Aβ, BAX, or Bcl-xl
Disorder prediction: Identifying intrinsically disordered regions that contribute to clusterin's binding promiscuity
Network-based prediction methods:
Interactome mapping: Constructing comprehensive protein-protein interaction networks
Network perturbation analysis: Simulating the effects of clusterin alterations on cellular pathways
Differential network analysis: Comparing interaction networks between healthy and disease states
Module identification algorithms: Detecting functional modules involving clusterin in complex networks
Machine learning prediction approaches:
Sequence-based interaction prediction: Using deep learning to identify potential binding partners from primary sequences
Structure-based binding affinity prediction: Estimating interaction strengths based on structural features
Context-specific interaction models: Training algorithms on tissue or disease-specific datasets
Transfer learning models: Applying knowledge from well-characterized interactions to predict novel ones
Multi-scale modeling frameworks:
Integrative modeling: Combining data from multiple experimental techniques to build comprehensive models
Coarse-grained simulations: Modeling larger systems and longer timescales than possible with atomic detail
Systems biology models: Incorporating clusterin into broader cellular pathway simulations
Agent-based modeling: Simulating emergent behaviors resulting from clusterin interactions
These computational approaches can generate testable hypotheses about clusterin's binding partners and functional impacts in different disease contexts, guiding experimental design and potentially identifying novel therapeutic targets.
Ensuring the quality of recombinant clusterin is essential for experimental reproducibility. Critical quality control parameters include:
Purity assessment:
SDS-PAGE with Coomassie staining (>95% purity recommended)
Size-exclusion chromatography to confirm monodispersity
Mass spectrometry to verify molecular weight and detect contaminating proteins
Endotoxin testing (<1.0 EU/μg protein) to prevent confounding inflammatory effects
Structural integrity verification:
Circular dichroism to confirm secondary structure elements
Thermal shift assays to assess protein stability
Native PAGE to evaluate oligomeric state
Dynamic light scattering to detect aggregation
Functional validation:
Chaperone activity assays using model substrate proteins
Binding assays with known interaction partners (e.g., Aβ, complement proteins)
Cell-based activity assessments (e.g., cytoprotection against stress)
Comparison with native human clusterin for functional equivalence
Post-translational modification analysis:
Glycosylation profiling (for secreted clusterin variants)
Phosphorylation status determination
Disulfide bond mapping
Confirmation of proper α-β chain processing if applicable
Storage stability monitoring:
Freeze-thaw stability testing (avoid more than 3 cycles)
Long-term stability assessment at -80°C
Optimal buffer composition determination
Aggregation monitoring during storage
When using His-tagged clusterin specifically, additional quality control should include verification of tag accessibility, assessment of whether the tag affects function, and confirmation that endotoxin levels remain within acceptable ranges after metal affinity purification steps.
Selecting appropriate antibodies for clusterin detection requires careful consideration of their specificity and sensitivity profiles:
Antibody Type | Epitope Region | Form Specificity | Applications | Advantages | Limitations |
---|---|---|---|---|---|
Monoclonal anti-α chain | N-terminal domain | Primarily secreted form | WB, IHC, ELISA | High specificity, low background | May miss truncated forms |
Monoclonal anti-β chain | C-terminal domain | Both secreted and intracellular | WB, IHC, ELISA, IP | Detects multiple variants | Cannot distinguish forms alone |
Polyclonal (full-length) | Multiple epitopes | All forms | WB, IHC, IP | High sensitivity, robust signal | Lower specificity |
Anti-nuclear form | N-terminal truncated region | Intracellular variant | IHC, IF | Specifically detects nuclear form | Limited to localization studies |
Anti-neoepitope | α-β junction | Uncleaved precursor | WB, ELISA | Specific to immature form | Narrow application range |
For brain tissue studies, paired monoclonal and polyclonal antibodies provide complementary information about clusterin distribution, as demonstrated in TBI studies
For distinguishing between secreted and intracellular forms, combinations of antibodies targeting different domains are recommended
Validation using both positive controls (recombinant protein) and negative controls (CLU knockout samples) is essential
Pre-absorption controls should be performed to confirm specificity in immunohistochemistry applications
The choice of antibody should be guided by the specific research question, with particular attention to whether form-specific detection is required.
Designing effective genetic manipulation experiments for clusterin requires careful consideration of several critical factors:
Target specificity considerations:
Isoform selectivity: Design manipulations that target all variants or specific splice variants
Off-target effects: Thoroughly validate guide RNAs or siRNAs for specificity
Regulatory element targeting: Consider manipulating promoters or enhancers versus coding regions
Compensatory mechanisms: Assess upregulation of related chaperones following CLU manipulation
Technology selection guidelines:
CRISPR-Cas9: Optimal for stable knockout, knock-in of specific variants, or promoter modification
siRNA/shRNA: Suitable for transient knockdown studies or graded expression reduction
Overexpression systems: Consider inducible systems to control expression timing and magnitude
Base editors: Appropriate for introducing specific point mutations found in disease variants
Delivery method optimization:
Viral vectors: Select appropriate serotype based on target cell type
Lipid nanoparticles: Consider for in vivo delivery to specific tissues
Electroporation: Optimize parameters for cell type-specific delivery
Transgenic approaches: For whole-organism studies with tissue-specific promoters
Validation requirements:
Transcript level verification: RT-qPCR with primers spanning multiple exons
Protein level confirmation: Western blotting with antibodies targeting different regions
Functional assessment: Chaperone activity assays, stress response tests
Phenotypic evaluation: Cell viability, morphology, and function assessments
Control design:
Rescue experiments: Re-expressing CLU to verify phenotype specificity
Scrambled guides/siRNAs: Controlling for non-specific effects
Empty vector controls: For overexpression studies
Wild-type comparisons: Including non-manipulated cells alongside all experiments
By carefully addressing these considerations, researchers can design robust genetic manipulation experiments that provide meaningful insights into clusterin's function in normal physiology and disease contexts.
Apolipoprotein-J, also known as Clusterin, is a multifunctional glycoprotein involved in various physiological processes. The recombinant form of Apolipoprotein-J tagged with a His-tag is widely used in research for its ease of purification and detection.
Apolipoprotein-J (ApoJ) is synthesized as a 427 amino acid polypeptide that is post-translationally cleaved into two subunits, designated as ApoJ α (residues 1-205) and ApoJ β (residues 206-427). These subunits are associated through disulfide bonds . The mature protein is a disulfide-linked heterodimeric glycoprotein with an approximate molecular mass of 75-80 kDa .
Apolipoprotein-J is an extracellular molecular chaperone that binds to misfolded proteins in body fluids, neutralizing their toxicity and mediating their cellular uptake by receptor-mediated endocytosis. Once internalized, these complexes are trafficked to lysosomes for degradation . ApoJ is involved in lipid transport, membrane recycling, cell adhesion, programmed cell death, and complement-mediated cell lysis . It has been implicated in various diseases, including neurodegenerative disorders, cancers, inflammatory diseases, and aging .
The His-tag, also known as a polyhistidine tag, is an amino acid motif consisting of at least six histidine residues, often added to the N- or C-terminus of recombinant proteins. This tag facilitates the purification and detection of the protein through immobilized metal ion affinity chromatography (IMAC), where the histidine residues chelate metal ions like nickel, cobalt, or copper . The His-tag allows for the selective isolation of the protein of interest, making it a valuable tool in protein research .
Recombinant Apolipoprotein-J with a His-tag is used in various research applications, including studies on lipid metabolism, neurodegenerative diseases, and cancer. Its ability to bind and neutralize misfolded proteins makes it a useful model for understanding protein aggregation and clearance mechanisms. Additionally, its role in lipid transport and cell adhesion provides insights into cardiovascular and metabolic diseases.