PFDN2 is upregulated in multiple cancers and correlates with poor prognosis:
Alzheimer’s Disease: Upregulated in brain tissues, potentially compensating for amyloid-β toxicity .
Huntington’s Disease: Knockdown increases aggregation of pathogenic huntingtin protein .
PFDN2 drives G1/S transition by enhancing MYBL2 transcription through hnRNPD nuclear translocation. Overexpression accelerates gastric cancer proliferation, while silencing induces G1 arrest .
Prefoldin subunits: PFDN1, PFDN3, PFDN4, PFDN5, PFDN6.
Transcription regulators: UXT, URI1, POLR2E.
Chaperones: PDRG1, WDR92.
Production: Expressed in E. coli with a His-tag; molecular weight 18.8 kDa .
Applications: Used in studies of chaperone mechanisms, cancer pathways, and drug discovery .
Biomarker Potential: High urinary PFDN2 levels in bladder cancer and serum anti-PFDN2 antibodies in type 2 diabetes .
Therapeutic Target: Silencing PFDN2 inhibits tumor growth in preclinical models, suggesting utility in targeted therapies .
Mechanistic Studies: Elucidate PFDN2’s role in taxane resistance and neurodegenerative proteinopathies.
Clinical Trials: Validate PFDN2 as a prognostic marker in multicenter cohorts.
MGSSHHHHHH SSGLVPRGSH MAENSGRAGK SSGSGAGKGA VSAEQVIAGF NRLRQEQRGL ASKAAELEME LNEHSLVIDT LKEVDETRKC YRMVGGVLVE RTVKEVLPAL ENNKEQIQKI IETLTQQLQA KGKELNEFRE KHNIRLMGED EKPAAKENSE GAGAKASSAG VLVS.
PFDN2 (Prefoldin subunit 2) is a component of the heterohexameric prefoldin complex conserved from archaea to humans. It functions as a cochaperone during co-translational folding of proteins, particularly actin and tubulin monomers. PFDN2 binds specifically to cytosolic chaperonin (c-CPN) and transfers target proteins to it, promoting proper folding in environments with competing pathways for nonnative proteins . Notably, PFDN2 is present in both the canonical prefoldin complex and the Uri/prefoldin-like complex, distinguishing it from subunits like PFDN5 that appear only in the canonical complex .
Human PFDN2 consists of 154 amino acids (Met1-Ser154) in its mature form. The recombinant human PFDN2 with a polyhistidine tag has 169 amino acids with a predicted molecular mass of 18.5 KDa, typically migrating as an 18-20 KDa band in SDS-PAGE under reducing conditions . The protein is encoded by the PFDN2 gene (also known as PFD2, HSPC231) and functions as part of the jellyfish-like structure of the prefoldin complex, which contains six subunits divided into α and β classes based on sequence similarity .
PFDN2 plays a significant role in transcriptional regulation through multiple mechanisms. Research has demonstrated that prefoldin perturbations cause transcriptional alterations across the human genome, with PFDN2 depletion affecting gene expression particularly under conditions of transcriptional regulation such as serum stimulation . The following table summarizes key findings regarding PFDN2's impact on gene expression:
Gene Expression Parameter | Effect of PFDN2 Depletion | Effect of PFDN5 Depletion |
---|---|---|
Significantly affected genes before serum stimulation | Moderate number | Higher number |
Significantly affected genes after serum stimulation | Increased compared to starved state | Increased compared to starved state |
Serum-induced genes (log2(FC) > 1, FDR < 0.05) | 44 genes impaired | 82 genes impaired |
Overlap between affected genes | Significant overlap with PFDN5-dependent genes | Significant overlap with PFDN2-dependent genes |
Direction of gene expression changes | Consistent with PFDN5 depletion | Consistent with PFDN2 depletion |
The significant overlap and consistent direction of gene expression changes between PFDN2 and PFDN5 depletions suggest these subunits share important functions in transcriptional regulation .
Investigating PFDN2's role in co-transcriptional splicing requires a multi-faceted methodological approach:
RNA-seq analysis: Used to detect changes in exon-intron ratios and alternative splicing events using computational tools like SUPPA
RT-qPCR: For measuring pre-mRNA levels for specific introns in control versus PFDN2-depleted cells
Chromatin immunoprecipitation (ChIP): To analyze PFDN2 binding to chromatin and its association with RNA polymerase II
ChIP-seq analysis: For mapping genome-wide binding patterns of prefoldin components
RNase treatment experiments: To determine if prefoldin binding to chromatin is RNA-dependent
Phosphorylation analysis: Western blotting and ChIP to assess RNA polymerase II CTD phosphorylation status
These complementary approaches enable researchers to comprehensively characterize PFDN2's role in the complex process of co-transcriptional splicing.
PFDN2 depletion leads to severe pre-mRNA splicing defects, particularly affecting long genes with numerous introns. The experimental data reveals several mechanistic insights:
Parameter | Control Cells | PFDN2-depleted Cells | PFDN5-depleted Cells |
---|---|---|---|
Exon ratio before serum stimulation | Lower | Higher | Higher |
Exon ratio after serum stimulation | Higher | Lower | Lower |
Pre-mRNA levels in model genes | Baseline | Increased | Increased |
RNA pol II CTD Ser2 phosphorylation | Normal | Decreased | Decreased |
RNA pol II CTD Ser5 phosphorylation | Normal | Decreased | Decreased |
PRP19 recruitment to chromatin | Normal | Reduced | Reduced |
U2AF65 recruitment to chromatin | Normal | Reduced | Reduced |
CDK9 recruitment to transcribed genes | Normal | Reduced | Reduced |
The mechanism involves reduced phosphorylation of RNA polymerase II's carboxy-terminal domain, which compromises the recruitment of splicing factors like PRP19 and U2AF65. This reduces co-transcriptional splicing efficiency, resulting in increased pre-mRNA retention .
Creating reliable PFDN2-deficient cell models requires careful technical considerations:
siRNA-mediated knockdown: Researchers have achieved approximately 67% efficiency in reducing PFDN2 protein using specific siRNAs (sequence: CAGCCUAGUGAUCGAUACA) after 72 hours of treatment . Transfection typically uses Oligofectamine reagent (8 μl with 15 μl siRNA at 20 nM) .
CRISPR-Cas9 gene editing: While the search results don't specifically detail CRISPR for PFDN2, the protocol described for PFDN5 can be adapted, including gRNA design, co-transfection with hCas9, clone selection by dilution, and validation through sequencing .
Experimental timeline: For serum stimulation experiments, cells are typically transfected with siRNAs for 24 hours, serum-starved for 48 hours, then stimulated by adding serum, with samples collected before and 90 minutes after stimulation .
Cell viability considerations: Researchers should monitor potential cytotoxicity, as prefoldin depletion may affect cell viability, though serum starvation appears to mitigate some viability issues with PFDN5 siRNA .
Analysis of PFDN2's genome-wide binding employs several sophisticated approaches:
ChIP-seq protocol: After crosslinking with formaldehyde, cells are sonicated and immunoprecipitated with PFDN2-specific antibodies. The DNA is purified, sequenced, and mapped to the reference genome .
Data processing workflow:
Binding pattern analysis: Prefoldin binding correlates with RNA polymerase II occupancy, showing the highest signals around transcription start sites .
Functional correlation: A key finding is the negative correlation between PFDN2 binding intensity and the effect of its depletion on gene expression - genes with higher PFDN2 occupancy show greater dependence on PFDN2 for their expression .
PFDN2 significantly impacts alternative pre-mRNA processing through its role in co-transcriptional splicing:
Approximately 20% of genes show altered alternative processing events upon prefoldin depletion
These events include both suppressed (≥10% less frequent) and enhanced (≥10% more frequent) processing compared to control cells
No specific class of alternative processing event is particularly affected, suggesting a broad influence on splicing mechanisms
Analysis uses computational tools like SUPPA to identify and classify alternative processing events from RNA-seq data
The exon ratio (exonic reads/total reads) calculation provides a quantitative measure of splicing efficiency before and after stimulation
The systematic analysis of these events reveals PFDN2's broad impact on the splicing machinery rather than affecting specific types of alternative processing.
Studying PFDN2-RNA polymerase II interactions requires precise methodology:
Chromatin Immunoprecipitation (ChIP):
Co-immunoprecipitation:
Phosphorylation analysis:
These approaches have revealed that PFDN2 depletion reduces CTD phosphorylation and CDK9 recruitment, providing mechanistic insight into how PFDN2 influences transcription and splicing.
When analyzing differences between PFDN2 and PFDN5 depletion effects, researchers should consider:
Complex-specific functions: PFDN2 exists in both canonical prefoldin and Uri/prefoldin-like complexes, while PFDN5 is only in the canonical complex. Effects seen with PFDN2 depletion but not with PFDN5 depletion likely reflect Uri/prefoldin-like complex functions .
Depletion efficiency: PFDN2 depletion (67% reduction) versus PFDN5 depletion (55% reduction) may account for some differences in experimental outcomes .
Protein abundance considerations: The higher baseline levels of PFDN2 compared to PFDN5 in human cells may explain the lower impact of PFDN2 depletion on some phenotypes .
Redundancy mechanisms: The prefoldin complex architecture may allow some functional redundancy between subunits, potentially masking certain phenotypes.
Experimental validation: Key findings should be validated using multiple approaches:
Understanding these factors helps researchers distinguish technical artifacts from biologically meaningful differences in subunit functions.
PFDN2's role in co-transcriptional splicing has significant implications for understanding splicing-related diseases:
Mechanistic insights: The finding that PFDN2 influences RNA polymerase II CTD phosphorylation and subsequent splicing factor recruitment provides a molecular mechanism that may be dysregulated in disease contexts .
Long gene susceptibility: The observation that PFDN2 depletion particularly affects long genes with numerous introns suggests a potential vulnerability relevant to neurodevelopmental disorders, where long genes are often affected .
Alternative splicing regulation: PFDN2's broad impact on alternative processing events (affecting ~20% of genes) indicates it may contribute to splicing dysregulation in diseases characterized by aberrant splicing patterns .
Experimental approaches: Researchers investigating splicing-related diseases could:
Analyze PFDN2 expression or mutations in patient samples
Examine PFDN2 binding patterns at disease-relevant genomic loci
Test whether PFDN2 overexpression can rescue splicing defects in disease models
To dissect PFDN2's dual roles, researchers should consider:
Compartment-specific analysis:
Nuclear versus cytoplasmic fractionation to separate transcriptional from protein folding functions
Fluorescent tagging of PFDN2 to track localization during different cellular processes
Domain-specific mutations:
Engineer PFDN2 variants with mutations in domains required for:
Chaperonin binding (protein folding function)
Chromatin association (transcriptional function)
Test these variants in rescue experiments
Temporal dynamics:
Synchronized cell studies to examine PFDN2 function during different cell cycle phases
Pulse-chase experiments to distinguish immediate versus sustained effects
Interaction partner analysis:
BioID or proximity labeling to identify compartment-specific interaction partners
Compare interaction networks under conditions that primarily engage one function versus the other
Integrated multi-omics approach:
Combine proteomics (for folding function) with transcriptomics and genomics (for transcriptional function)
Correlate protein folding defects with transcriptional alterations in the same experimental system
This comprehensive approach would help delineate PFDN2's functions in different cellular contexts and reveal potential crosstalk between its roles.
When working with recombinant human PFDN2 protein, implement these quality control measures:
Purity verification: Confirm >95% purity via SDS-PAGE analysis
Molecular weight confirmation: Verify migration at 18-20 KDa on SDS-PAGE, consistent with its predicted 18.5 KDa mass
Functional assays:
Test binding to cytosolic chaperonin
Verify ability to assist in protein folding of model substrates
Assess complex formation with other prefoldin subunits
Storage optimization:
Determine optimal buffer conditions to prevent aggregation
Validate protein stability after freeze-thaw cycles
Monitor activity retention during storage
Batch consistency:
Compare lot-to-lot variation in activity assays
Ensure reproducible results across different preparations
Expression system considerations:
These measures ensure that experimental outcomes reflect genuine PFDN2 biology rather than artifacts from improperly prepared protein.
The prefoldin complex is a heterohexameric structure composed of six subunits, including two alpha and four beta subunits. PFDN2 belongs to the beta subunit family . The complex has a unique jellyfish-like structure with tentacle-like extensions that capture unfolded protein substrates and transfer them to group II chaperonins for proper folding .
Prefoldin operates in an ATP-independent manner, which makes it particularly useful during cellular stress conditions where ATP levels might be compromised . This ATP-independent mechanism allows prefoldin to act as a holdase, stabilizing unfolded proteins and preventing their aggregation until they can be properly folded by chaperonins .
Prefoldin plays a crucial role in maintaining protein homeostasis, also known as proteostasis. It assists in the folding of approximately 10% of newly synthesized polypeptides downstream of the translation machinery . This includes cytoskeletal proteins such as actin and tubulin, which are essential for maintaining cell structure and function .
Recent studies have linked prefoldin to mitochondrial function. Specifically, PFDN2 has been shown to support mitochondrial morphology and the abundance of some respiratory chain complexes . It interacts with Tom70, a receptor for mitochondrial precursor proteins, suggesting a role in the quality control of proteins destined for the mitochondria .
Prefoldin and its subunits have been implicated in various diseases, particularly neurodegenerative disorders such as Alzheimer’s, Parkinson’s, and Huntington’s diseases . The complex’s ability to prevent protein aggregation is crucial in these conditions, where misfolded proteins form toxic aggregates that damage cells.
Additionally, abnormal expression of prefoldin subunits has been observed in different types of tumors. For instance, the c-Myc binding protein MM-1, a prefoldin subunit, can inhibit the activity of c-Myc, a protein involved in cell proliferation and tumor growth . This highlights the potential of prefoldin subunits as therapeutic targets in cancer treatment.