CLU’s dual roles in cell survival and death depend on cellular context:
Anti-apoptotic: Inhibits BAX mitochondrial translocation, activates PI3K/Akt signaling, and suppresses p53-mediated apoptosis .
Pro-apoptotic: Truncated nuclear CLU promotes DNA damage response .
In Alzheimer’s disease (AD), CLU binds amyloid-β (Aβ), modulating fibril formation. While secreted CLU may clear Aβ, excessive levels or altered trafficking promote neurotoxic oligomers .
CLU is the third-largest genetic risk factor for late-onset AD (LOAD) after APOE and BIN1. The rs11136000 polymorphism reduces AD risk in Caucasians but shows no association in Asian populations . In AD brains, CLU levels correlate with Aβ distribution, peaking in cortical regions .
CLU overexpression confers resistance to chemotherapy. Custirsen, an antisense oligonucleotide targeting CLU mRNA, showed promise in clinical trials:
Trial Phase | Cancer Type | Treatment | Outcome | Sources |
---|---|---|---|---|
Phase I | Prostate | Custirsen + surgery | Clusterin suppression, apoptosis increase | |
Phase II | CRPC | Custirsen + docetaxel | Improved OS (23.8 vs. 16.9 months) |
The rs11136000 polymorphism (C→T) is protective in Caucasians (OR: 0.84–0.92) but irrelevant in Asians . This ethnic disparity highlights the need for population-specific therapeutic strategies.
Amyloid Toxicity: CLU’s role in Aβ oligomer formation vs. fibril clearance remains debated. In vivo studies suggest structural differences in amyloid deposits when CLU is absent .
Intracellular vs. Secreted CLU: ER stress-induced cytoplasmic accumulation may mitigate neurotoxicity in ALS models but exacerbate Aβ toxicity in AD .
Metabolic Regulation: The β-chain’s lipid-promoting effects contrast with full-length CLU’s lipid-lowering activity , warranting isoform-specific targeting.
Clusterin is synthesized as a preproprotein derived primarily from mRNA transcript NM_001831.3 containing exons 1-9, with translation typically initiated at the ATG in exon 2. The resulting protein undergoes extensive post-translational modifications in the secretory pathway. Initially, the preproprotein enters the endoplasmic reticulum (ER) where the N-terminal signal peptide is cleaved to produce a 50 kDa immature proprotein. This proprotein undergoes phosphorylation and glycosylation in the ER and Golgi apparatus . Within the Golgi, further processing occurs via cleavage between residues 227 and 228, resulting in alpha and beta chains linked by five disulfide bonds. The mature secreted clusterin is a highly glycosylated heterodimer with a molecular weight of 75-80 kDa, consisting of two 40 kDa chains .
Clusterin exists in two primary forms with distinct cellular localizations and functions:
Secreted Clusterin (sCLU): This is the predominant form, processed through the conventional secretory pathway. sCLU is glycosylated and functions as an extracellular chaperone with cytoprotective properties. It interacts with the BAX-Ku70 complex, stabilizing it and inhibiting BAX translocation to mitochondria, thereby preventing apoptosis .
Nuclear Clusterin (nCLU): This form arises from alternative splicing and non-canonical translation, producing a truncated, non-glycosylated protein that lacks exon 2. Unlike sCLU, nCLU has pro-apoptotic functions. It competes with BAX for binding to Ku70, leading to increased free BAX that can translocate to mitochondria and trigger apoptosis. nCLU also interacts with DNA-PK complexes, inhibiting DNA repair and promoting cell stress and death .
The balance between these isoforms appears crucial for cellular homeostasis, with alterations potentially contributing to pathological states.
Clusterin expression is regulated through multiple mechanisms:
The CLU promoter contains a 14-base pair clusterin element (CLE) that differs from the heat shock element (HSE) by only a single base pair. Under stress conditions, this element becomes bound by heat shock factor 1 (HSF-1), inducing CLU expression . During proteasome inhibition, HSF-2 can also bind to this element .
Additionally, various stress conditions can trigger alternative splicing of CLU mRNA, leading to different protein isoforms with distinct subcellular localizations and functions. This regulated splicing represents an important mechanism for controlling the balance between pro-survival and pro-apoptotic functions of clusterin .
Clusterin (CLU) is considered the third most significant genetic risk factor for late-onset Alzheimer's Disease (LOAD), following APOE and BIN1. This status was established through multiple genome-wide association studies (GWAS) that identified several single nucleotide polymorphisms (SNPs) in the CLU gene as susceptibility loci for AD .
The significance of clusterin in AD is supported by multiple lines of evidence:
Clusterin is upregulated in the hippocampus and cortex of AD brains, where it colocalizes with amyloid-beta (Aβ) plaques .
Elevated clusterin levels are detected in the cerebrospinal fluid (CSF) of AD patients .
Higher plasma clusterin levels correlate with increased hippocampal atrophy and faster clinical progression in AD patients .
Rare AD-associated mutations in CLU alter protein trafficking, resulting in increased intracellular clusterin accumulation and reduced secretion, suggesting that altered clusterin distribution may contribute to AD pathogenesis .
These findings collectively suggest that clusterin plays a critical role in AD pathophysiology, though it remains unclear whether it functions as a causal factor in disease development or contributes to disease progression once initiated.
When investigating clusterin's interaction with amyloid-beta, researchers should consider the following methodological approaches:
Protein-protein interaction assays: Surface plasmon resonance, co-immunoprecipitation, and proximity ligation assays can determine binding affinities and interaction dynamics between clusterin and Aβ under various conditions.
Aggregation studies: Thioflavin T fluorescence assays, electron microscopy, and dynamic light scattering can assess how clusterin influences Aβ aggregation kinetics. Researchers should examine different clusterin:Aβ ratios, as the nature of the interaction appears ratio-dependent .
Cellular models: Primary neurons, neuronal cell lines, and iPSC-derived neurons can be treated with Aβ with or without exogenous clusterin, or with clusterin knockdown/knockout, to assess effects on viability, toxicity, and clearance mechanisms .
In vivo models: Clusterin knockout mice crossed with AD model mice (e.g., APP/PS1) can reveal the impact of clusterin deficiency on Aβ deposition, clearance, and cognitive outcomes.
Trafficking studies: Fluorescently tagged clusterin constructs and subcellular fractionation can track how Aβ exposure alters clusterin trafficking between intracellular compartments and the extracellular space .
Researchers should be mindful that clusterin's effects on Aβ may be concentration-dependent, with clusterin exhibiting neuroprotective properties at certain concentrations and neurotoxic effects at others .
Differentiating between clusterin's protective and pathological roles requires careful experimental design that considers:
Clusterin isoform specificity: Researchers should develop isoform-specific antibodies or expression constructs to distinguish between secreted and intracellular clusterin forms. The secreted form is generally considered neuroprotective, while intracellular accumulation may be pathological .
Clusterin:Aβ ratio analysis: Studies should systematically vary the ratio between clusterin and Aβ, as clusterin's effects on Aβ aggregation and toxicity appear ratio-dependent. At high clusterin:Aβ ratios, clusterin may prevent aggregation, while at low ratios, it might promote the formation of toxic oligomers .
Temporal considerations: Acute versus chronic effects should be distinguished by using inducible expression systems or time-course studies to determine if clusterin's role changes during disease progression.
Mechanistic dissection: Researchers should employ pathway inhibitors or genetic approaches to isolate specific mechanisms. For example, blocking clusterin-mediated Aβ receptors or specific clearance pathways can help determine whether clusterin's predominant effect is promoting clearance or enhancing toxicity .
Cell-type specific analyses: Different neural cell types may respond differently to clusterin-Aβ interactions. Single-cell approaches or cell-type specific manipulations can help resolve these differences .
Recent evidence suggests that clusterin knockdown provides protection from Aβ-induced neurotoxicity in both rodent neurons and iPSC-derived neurons, indicating that clusterin may mediate Aβ toxicity under certain conditions . This highlights the importance of context-dependent evaluation of clusterin's roles.
For effective genetic manipulation of clusterin, researchers should consider:
CRISPR-Cas9 genome editing: This approach allows for precise modification of the CLU gene, including:
Complete knockout to study loss-of-function effects
Introduction of specific SNPs associated with AD risk to study variant effects
Modification of specific exons to selectively alter expression of different clusterin isoforms
Creation of reporter constructs for live tracking of clusterin expression and localization
RNA interference approaches: siRNA or shRNA targeting specific regions of clusterin mRNA can achieve:
Transient knockdown for acute effects assessment
Stable knockdown using inducible systems for temporal control
Isoform-specific knockdown by targeting unique regions of alternatively spliced variants
Antisense oligonucleotides: These can be designed similar to Custirsen (OGX-011), which was developed to target clusterin in cancer contexts. These oligonucleotides can provide specific targeting of secreted versus nuclear forms of clusterin .
Overexpression systems: Using different promoters and inducible expression systems to control:
Expression of wild-type versus mutant forms
Expression of specific clusterin isoforms
Expression with different tags for localization studies
The choice of cell model is crucial, as clusterin's function appears to be context-dependent. iPSC-derived neurons from patients with AD risk variants in CLU offer a physiologically relevant system for studying the effects of genetic manipulation on clusterin function in human neurons .
Accurately distinguishing between intracellular and secreted clusterin requires multiple complementary approaches:
Subcellular fractionation: Sequential extraction of proteins from different cellular compartments, followed by Western blotting with antibodies that recognize specific forms of clusterin. This approach can separate nuclear, cytoplasmic, membrane-bound, and secreted fractions.
Immunocytochemistry with form-specific antibodies: Antibodies targeting unique epitopes present in secreted versus intracellular clusterin can be used for co-localization studies with organelle markers.
Glycosylation analysis: Since secreted clusterin is heavily glycosylated while intracellular forms may be unglycosylated or differently glycosylated, researchers can use:
Enzymatic deglycosylation (PNGase F treatment) followed by Western blotting to detect mobility shifts
Lectins that bind specific glycan structures
Mass spectrometry to characterize glycan profiles
Pulse-chase experiments: Metabolic labeling with radioactive amino acids or click chemistry can track newly synthesized clusterin through the secretory pathway versus retained intracellular forms.
Domain-specific tagging strategies: Genetic constructs with tags positioned to distinguish between different clusterin forms (e.g., N-terminal tags visible only before signal peptide cleavage, internal tags that remain after processing).
Researchers should be aware that stress conditions can alter the normal trafficking of clusterin, causing accumulation of normally secreted forms within the cell . Therefore, careful consideration of experimental conditions is essential for accurate interpretation of results.
For reliable monitoring of clusterin activity, researchers should consider the following biomarkers:
Fluid biomarkers:
Cerebrospinal fluid (CSF) clusterin levels, which are upregulated in AD patients
Plasma/serum clusterin levels, which correlate with hippocampal atrophy and clinical progression in AD
Ratio of different clusterin isoforms in biofluids
Post-translational modifications of clusterin (glycosylation patterns, phosphorylation status)
Tissue biomarkers:
Functional readouts:
Genetic biomarkers:
CLU risk variants associated with AD
Expression levels of specific CLU mRNA isoforms
Epigenetic modifications of the CLU gene locus
An endophenotype-based approach using CSF clusterin levels has been employed to identify novel loci linked to AD pathogenesis through clusterin alterations . This approach represents a promising strategy for developing more specific and sensitive biomarkers.
Clusterin's involvement extends beyond Alzheimer's Disease to several other neurodegenerative conditions, with both similarities and differences in its function:
Similarities across neurodegenerative disorders:
Increased clusterin levels are observed in multiple neurodegenerative diseases, including ALS, multiple sclerosis, transmissible spongiform encephalopathies, and Huntington's disease
In protein aggregation disorders, clusterin's chaperone function appears to play a role in modulating protein aggregation and toxicity
Stress-induced changes in clusterin expression and trafficking are common features
Alpha-synucleinopathies (Parkinson's Disease, Lewy Body Dementia):
Amyotrophic Lateral Sclerosis (ALS):
ER stress in a Drosophila model of ALS results in cytoplasmic accumulation of clusterin
This accumulation reduces TDP-43 protein inclusions and partially rescues the ALS-like phenotype
This suggests a potentially protective role, in contrast to some evidence in AD where intracellular clusterin accumulation may be harmful
Multiple Sclerosis:
The key difference appears to be the specific protein aggregates or pathological processes that clusterin interacts with in each disorder. Researchers investigating clusterin across neurodegenerative diseases should consider these context-specific interactions while recognizing the common underlying mechanisms related to protein aggregation, oxidative stress, and cell survival pathways .
To investigate clusterin's dual role in cancer, researchers should implement these methodological approaches:
Isoform-specific expression analysis:
qRT-PCR with primers specific to alternatively spliced variants
Western blotting with antibodies that distinguish between secreted clusterin (sCLU) and nuclear clusterin (nCLU)
Immunohistochemistry with isoform-specific antibodies to assess subcellular localization
Context-dependent functional assays:
Cell proliferation, invasion, and migration assays with:
Isoform-specific overexpression
Selective knockdown of specific isoforms
Exposure to different stress conditions
Apoptosis assays after various treatments to assess pro-survival (sCLU) versus pro-apoptotic (nCLU) functions
Molecular pathway analysis:
Therapeutic response evaluation:
In vivo studies:
Tumor xenograft models with manipulation of specific clusterin isoforms
Metastasis models to evaluate clusterin's role in different stages of cancer progression
Research from clinical trials with Custirsen (OGX-011), an antisense oligonucleotide targeting clusterin, has provided insights into clusterin's role in cancer therapy resistance, though effects were not better than current regimens alone . These approaches should be applied with careful consideration of cancer type, stage, and microenvironment context, as clusterin's effects appear highly dependent on these factors.
Findings from cardiovascular disease research on clusterin can inform neurodegeneration studies in several key ways:
Protective mechanisms:
In cardiac cells, clusterin appears to be primarily protective during damage and disease
This contrasts with its more complex role in neurodegeneration, where it can be both protective and pathological
Investigating the molecular determinants that specify protective functions in cardiac tissue could reveal strategies to promote clusterin's protective role in neurodegenerative contexts
Stress response pathways:
Both cardiovascular and neurodegenerative diseases involve cellular stress responses where clusterin plays a role
Comparative analysis of stress-induced clusterin expression and trafficking in cardiac versus neuronal cells could identify tissue-specific regulatory mechanisms
Understanding how clusterin responds to oxidative stress, which is common to both disease categories, could reveal shared pathways for therapeutic targeting
Protein quality control systems:
Clusterin's chaperone function is relevant in both cardiovascular and neurodegenerative diseases
Examining how clusterin interacts with misfolded proteins in different cellular environments could inform approaches to enhance its beneficial chaperone activity in neurodegenerative contexts
Vascular contributions to neurodegeneration:
Since clusterin is expressed in the vasculature and many neurodegenerative diseases have vascular components
Research on clusterin's role in vascular health could illuminate mechanisms of cerebrovascular contribution to neurodegenerative diseases
The blood-brain barrier interface represents a critical area where clusterin's function in both domains intersects
Therapeutic approaches:
Strategies that have successfully modulated clusterin's protective functions in cardiovascular models could be adapted for neurodegenerative conditions
Lessons from failed or successful cardiovascular interventions targeting clusterin could inform neurodegeneration-focused therapeutic development
By systematically comparing clusterin's molecular interactions, regulation, and function across these different disease contexts, researchers can identify common principles and disease-specific peculiarities that could lead to more targeted therapeutic approaches .
When designing isoform-specific detection methods for clusterin, researchers should address these critical considerations:
Epitope selection:
Target unique regions present in specific clusterin isoforms
For secreted clusterin (sCLU), epitopes in the signal peptide region or glycosylated domains
For nuclear clusterin (nCLU), epitopes in regions retained after alternative splicing
Avoid epitopes in the alpha and beta chains common to all isoforms unless specifically distinguishing between processed and unprocessed forms
Post-translational modification awareness:
Account for differential glycosylation patterns between isoforms
Consider phosphorylation states that may be isoform-specific
Design detection methods that are either sensitive or insensitive to these modifications, depending on research goals
Subcellular localization techniques:
Combine immunological detection with subcellular fractionation
Use confocal microscopy with co-localization markers for cellular compartments
Employ super-resolution microscopy for precise localization of clusterin variants
mRNA detection strategies:
Design primers spanning exon-exon junctions unique to alternatively spliced variants
Use RNAscope or similar techniques for in situ detection of specific mRNA variants
Consider quantitative PCR approaches with isoform-specific probes
Assay validation:
Use genetic models (knockout, knockdown, overexpression) to confirm specificity
Include appropriate positive and negative controls in all experiments
Cross-validate results using multiple detection methods
Experimental conditions:
These considerations are particularly important given that intracellular clusterin may arise through multiple mechanisms (impaired secretion, reuptake of secreted clusterin, or premature escape from the secretory pathway) . Careful attention to these technical details will enable more accurate interpretation of clusterin's diverse biological functions.
To resolve contradictory findings about clusterin's effects across experimental models, researchers should implement these methodological strategies:
Standardization of experimental conditions:
Define precise clusterin concentrations, as effects may be dose-dependent
Standardize cell types, culture conditions, and treatment durations
Establish consistent stress conditions when studying stress-induced changes in clusterin
Comprehensive characterization of model systems:
Document endogenous clusterin expression levels and isoform distribution
Verify cellular localization of clusterin in each model
Assess expression of key clusterin-interacting partners (e.g., Ku70, BAX, Bcl-xl)
Context-specific analysis:
Systematically vary clusterin:interacting partner ratios (e.g., clusterin:Aβ ratio) which can determine whether clusterin exhibits protective or toxic properties
Consider microenvironmental factors that may influence clusterin function
Evaluate cell-type specific responses, as clusterin's effects may vary between neurons, astrocytes, microglia, etc.
Multi-method validation:
Apply complementary experimental approaches to the same biological question
Use both gain-of-function and loss-of-function approaches
Validate in vitro findings in more complex systems (ex vivo, in vivo)
Temporal considerations:
Distinguish between acute and chronic effects of clusterin
Design time-course experiments to capture dynamic changes
Consider developmental timing in animal models
Systematic review methodology:
Conduct meta-analyses of published data with attention to methodological differences
Develop collaborative standardized protocols across research groups
Establish reporting standards for clusterin research to facilitate comparison
A relevant example is the apparently contradictory findings regarding clusterin's role in Aβ metabolism. Some studies indicate that clusterin alters aggregation and promotes Aβ clearance (neuroprotective), while others suggest clusterin reduces Aβ clearance and mediates neurotoxicity . These contradictions may be resolved by recognizing that the clusterin:Aβ ratio determines the nature of their interaction, highlighting the importance of carefully controlled experimental conditions .
Translating clusterin research from animal models to human applications requires careful consideration of several factors:
Species-specific differences in clusterin biology:
Compare amino acid sequences and post-translational modifications between species
Assess differences in alternative splicing patterns and isoform distribution
Evaluate species-specific promoter regulation and expression patterns
Document differences in clusterin interactome across species
Model selection and validation:
Use multiple animal models to verify findings (rodents, non-human primates)
Complement animal studies with human-derived systems (iPSC neurons, brain organoids)
Consider humanized animal models expressing human clusterin variants
Validate key findings in human post-mortem tissues
Disease modeling fidelity:
Assess how accurately animal models recapitulate human disease phenotypes
Consider differences in disease progression timelines between models and humans
Evaluate whether stress responses involving clusterin are comparable across species
Account for differences in blood-brain barrier properties affecting clusterin distribution
Genetic variability considerations:
Incorporate human genetic variants (e.g., AD-associated CLU SNPs) into animal models
Study effects in both sexes and across different genetic backgrounds
Consider population-specific genetic variation in human translation
Biomarker development strategy:
Validate clusterin-based biomarkers in both animal models and human samples
Develop assays that reliably detect comparable clusterin forms across species
Consider the translational potential of CSF, plasma, or imaging biomarkers
Therapeutic development pathway:
Human iPSC-derived neurons provide a valuable bridge between animal models and human applications, particularly when derived from patients with AD risk variants in CLU . Additionally, CRISPR-based studies for introducing and correcting specific variants are anticipated to be pivotal in understanding the functional consequences of human CLU variants and their potential as therapeutic targets .
Several emerging technologies show exceptional promise for advancing clusterin research:
Single-cell multi-omics approaches:
Single-cell transcriptomics to identify cell-specific clusterin expression patterns
Single-cell proteomics to detect isoform distribution at individual cell level
Spatial transcriptomics to map clusterin expression in complex tissues with preserved architecture
Integration of these datasets to understand cell type-specific clusterin functions
Advanced imaging technologies:
Super-resolution microscopy for precise subcellular localization of clusterin isoforms
Live-cell imaging with genetically encoded sensors to track clusterin trafficking in real-time
Correlative light and electron microscopy to connect clusterin localization with ultrastructural features
Expansion microscopy for enhanced visualization of clusterin-protein interactions
CRISPR-based technologies:
Protein interaction and structural biology approaches:
Cryo-electron microscopy to resolve clusterin-partner complex structures
Hydrogen-deuterium exchange mass spectrometry to map binding interfaces
Proximity labeling techniques (BioID, APEX) to identify context-specific clusterin interactors
AlphaFold and other AI-based structure prediction to model clusterin conformations
Organoid and microphysiological systems:
Brain organoids to study clusterin in complex neural environments
Organ-on-chip technology to model blood-brain barrier interactions with clusterin
Patient-derived organoids to study person-specific clusterin biology
Multi-organ microphysiological systems to study clusterin in systemic contexts
These technologies will be particularly valuable for resolving longstanding questions about clusterin's diverse biological functions and reconciling contradictory findings . The integration of multiple advanced approaches will likely provide the most comprehensive understanding of clusterin biology.
Based on current understanding of clusterin biology, several promising therapeutic strategies are emerging:
Isoform-specific modulation:
Selective inhibition of intracellular clusterin accumulation while preserving secreted clusterin's neuroprotective functions
Antisense oligonucleotides designed to alter the balance of specific clusterin isoforms
Small molecules targeting specific clusterin domains to modify its interaction with partners like Aβ or Ku70
Targeting clusterin-mediated pathways:
Modulation of the clusterin-Ku70-BAX pathway to control apoptotic signaling
Interventions targeting clusterin's role in protein quality control and autophagy
Compounds that alter clusterin's chaperone activity to promote proper protein folding
Enhancing protective functions:
Promoting clusterin-mediated clearance of protein aggregates
Augmenting clusterin's anti-inflammatory properties
Boosting clusterin's ability to protect against oxidative stress
Cell-type specific approaches:
Neuron-targeted delivery of secreted clusterin to enhance neuroprotection
Glial-targeted interventions to modify clusterin's role in neuroinflammation
Vascular-targeted strategies addressing clusterin's function at the blood-brain barrier
Combination therapies:
Clusterin-targeted approaches combined with anti-amyloid or anti-tau strategies
Synergistic interventions targeting multiple aspects of proteostasis
Combining clusterin modulation with neuroinflammation-targeted treatments
Personalized medicine approaches:
Stratification of patients based on CLU genetic variants for targeted interventions
Biomarker-guided therapy selection based on clusterin levels or isoform ratios
iPSC-based drug screening for patient-specific clusterin-targeting compounds
Despite significant advances, several critical questions about clusterin remain unresolved and require innovative research approaches:
Origin and trafficking of intracellular clusterin:
How exactly does intracellular clusterin arise? Is it primarily from impaired secretion, reuptake of secreted clusterin, or premature escape from the secretory pathway?
What cellular mechanisms regulate the balance between secreted and intracellular clusterin?
Structure-function relationships:
What are the structural differences between secreted and intracellular clusterin isoforms?
How do post-translational modifications affect clusterin's various functions?
What structural features determine whether clusterin exhibits protective or pathological effects?
Cell type-specific functions:
How does clusterin function differ between neurons, glia, and vascular cells in the brain?
Are there region-specific differences in clusterin biology within the brain?
How do these differences contribute to selective vulnerability in neurodegenerative diseases?
Causal role in disease:
Interaction with other risk factors:
How does clusterin interact with other major genetic risk factors for AD, such as APOE?
Are there synergistic or antagonistic effects between clusterin and other pathological proteins?
How do environmental factors modulate clusterin function?
Temporal dynamics:
How does clusterin's role change during aging and disease progression?
Are there critical periods during which clusterin modulation would be most effective?
How rapidly do changes in clusterin expression and localization occur in response to pathological triggers?
Addressing these questions will require integration of multiple innovative approaches, including longitudinal studies in human patients and animal models, advanced imaging techniques to track clusterin in real-time, comprehensive genetic studies incorporating multiple risk factors, and systems biology approaches to understand clusterin within complex cellular networks .
The CLU gene contains nine exons and produces several mRNA isoforms, although most are expressed at very low levels. The dominant isoform encodes a secreted form of clusterin, which is a disulfide-linked heterodimeric glycoprotein with an approximate molecular mass of 75-80 kDa . The mature protein consists of two chains, α and β, linked by disulfide bonds. There are also truncated versions of clusterin, such as a 55 kDa form localized to the nucleus, which has pro-apoptotic activities .
Clusterin functions as an extracellular molecular chaperone. It binds to misfolded proteins in body fluids, neutralizing their toxicity and facilitating their cellular uptake through receptor-mediated endocytosis. Once internalized, the complexes are trafficked to lysosomes for degradation . Clusterin is involved in several biological processes, including:
Clusterin is implicated in various diseases, including neurodegenerative diseases, cancers, inflammatory diseases, and aging . Its expression is finely regulated by cytokines, growth factors, and stress-inducing agents, leading to elevated levels in states of cellular disturbance. For example, in the eye, clusterin expression is significantly increased in conditions like age-related macular degeneration and Fuch’s corneal dystrophy .
Recent research has focused on the role of clusterin in tissue remodeling, wound healing, and its potential as a therapeutic target. Its ability to protect cells from apoptosis and its involvement in lipid transport and complement inhibition make it a protein of interest in the study of various pathologies .