PHF5A (PHD Finger Protein 5A) is a highly conserved nuclear protein encoded by the PHF5A gene in humans. It belongs to the PHD zinc finger protein family and functions as a critical component of the spliceosome, a macromolecular complex essential for pre-mRNA splicing . PHF5A is ubiquitously expressed in eukaryotic cells and plays roles in transcriptional regulation, chromatin remodeling, and embryonic stem cell pluripotency . Recent studies highlight its oncogenic potential, linking its dysregulation to tumor progression and metastasis .
PHF5A is a core subunit of the SF3B complex within the 17S U2 snRNP, facilitating:
Recognition of intron branch sites during pre-mRNA splicing .
Assembly of the "A complex" by anchoring U2 snRNP to pre-mRNA .
Chromatin Remodeling: Regulates histone modifications (e.g., H3K79me2, H3K36me3) via the PAF1 complex, influencing transcriptional elongation .
Embryonic Stem Cell Pluripotency: Maintains self-renewal by stabilizing PAF1C on pluripotency gene loci .
DNA Damage Repair: Modulates histone H2A variant deposition and chromatin integrity .
PHF5A is overexpressed in multiple malignancies, correlating with poor prognosis:
Alternative Splicing: Drives CRC progression by inducing aberrant splicing of MAP3K7 and TRAF6, activating NF-κB signaling .
Epigenetic Regulation: Promoter hyperacetylation (e.g., H3K27ac) elevates PHF5A expression in CRC .
Therapeutic Target: PHF5A-Y36C mutation confers resistance to splicing modulators, highlighting its druggable potential .
Commercial variants are produced for research applications:
CRC Models: PHF5A overexpression in HCT8 cells increased tumor growth and lung metastasis in mice .
Splicing Modulation: PHF5A-SF3B1 forms a binding pocket for splicing inhibitors, validated by cryo-EM .
Diagnostic Utility: PHF5A expression discriminates tumor vs. normal tissues in 12 cancer types (AUC >0.7) .
Immune Correlation: Associates with immunosuppressive microenvironments in BRCA and LUAD .
PHF5A represents a promising target for anticancer therapy due to its dual role in splicing and chromatin regulation. Ongoing research focuses on:
PHF5A contains a characteristic PHD-finger-like domain flanked by highly basic N- and C-terminal regions. Its zinc finger domain features a Cys4-His-Cys3 structure that mediates protein-protein and protein-DNA interactions . The protein shows remarkable evolutionary conservation, with 94% amino acid sequence similarity between humans and rodents (mice/rats) . PHF5A is predominantly localized in the nucleus, where it participates in various nuclear processes including splicing and transcriptional regulation .
PHF5A functions primarily as a component of the splicing factor 3b (SF3b) complex within the U2 small nuclear ribonucleoproteins complex (U2 snRNP) . This complex is essential for pre-mRNA splicing, binding to pre-mRNA upstream of the intron's branch site in a sequence-independent manner, potentially anchoring the U2 snRNP to the pre-mRNA . Beyond splicing, PHF5A participates in chromatin-mediated transcription, cell cycle regulation, DNA damage repair, and maintenance of embryonic stem cell pluripotency . These diverse functions position PHF5A as a multifunctional protein at the intersection of RNA processing and gene regulation .
PHF5A interacts with multiple components of the SF3B complex within the 17S U2 snRNP spliceosome . It directly participates in early spliceosome assembly and mediates recognition of the intron branch site during pre-mRNA splicing . PHF5A serves as a bridge protein between splicing proteins and ATP-dependent RNA helicases . Within the spliceosome, PHF5A contributes to the selection of the pre-mRNA branch-site adenosine, the nucleophile for the first step of splicing , and stabilizes protein-protein interactions essential for spliceosome function.
For investigating PHF5A's splicing functions, researchers should employ RNA-seq with specific computational pipelines for alternative splicing analysis, combined with CLIP-seq to map direct RNA-protein interactions . For chromatin-related functions, ChIP-seq identifies PHF5A binding sites on chromatin, while CUT&RUN provides higher resolution . Proximity labeling techniques like BioID or APEX can identify protein interaction networks in different cellular compartments. Co-immunoprecipitation experiments followed by mass spectrometry help define specific interaction partners in different cellular contexts. Combined approaches integrating RNA-protein crosslinking with chromatin association studies can reveal how PHF5A bridges these two processes.
Given PHF5A's essential role in splicing, complete knockout often results in cellular lethality . Researchers should employ inducible knockdown systems (e.g., doxycycline-regulated shRNA) or conditional knockout models (Cre-loxP systems) to control the timing and extent of PHF5A depletion. CRISPR-Cas9 with multiple guide RNAs targeting conserved regions can be used to create hypomorphic alleles. For studying specific functions, domain-specific mutations or phosphorylation site mutants (e.g., Y36F to prevent phosphorylation) provide insights while maintaining essential activities . Time-course experiments following partial depletion can distinguish primary from secondary effects and identify the most sensitive cellular processes to PHF5A reduction.
PHF5A undergoes key post-translational modifications including phosphorylation at Y36 and hyperacetylation that affect its function . Enrichment techniques such as phosphoprotein purification or acetylated protein immunoprecipitation should be employed before mass spectrometry analysis. Phospho-specific antibodies targeting Y36 phosphorylation can track this modification in various cellular contexts . Site-directed mutagenesis creating phosphomimetic (Y36E) or phospho-deficient (Y36F) mutants helps evaluate functional consequences. Targeted mass spectrometry approaches like parallel reaction monitoring (PRM) or multiple reaction monitoring (MRM) increase sensitivity for detecting specific modifications. Combining these approaches with functional assays links modifications to specific PHF5A activities.
PHF5A functions as a critical component of the SF3B subcomplex within the 17S U2 snRNP, which is essential for branch point recognition during splicing . Mechanistically, PHF5A contributes to the stable binding of U2 snRNP to the branchpoint sequence in pre-mRNA, forming the 'A' complex . Though its binding to pre-mRNA is sequence-independent, it helps anchor the U2 snRNP complex to the correct position . PHF5A works in concert with other SF3B components to ensure proper selection of the branch site adenosine, which serves as the nucleophile for the first catalytic step of splicing . Its positioning within the complex allows it to communicate with both RNA structures and protein components of the spliceosome.
Disruption of PHF5A function leads to widespread alterations in splicing patterns across the transcriptome, with particular effects on genes involved in cell cycle regulation, DNA damage response, and pluripotency maintenance . Studies indicate that PHF5A regulates alternative splicing of genes critical for cancer progression and stem cell function . While the complete repertoire of PHF5A-dependent splicing events remains to be fully characterized, research suggests that genes containing non-canonical or weak branch points may be particularly sensitive to PHF5A perturbation. Detailed transcriptome analysis following PHF5A depletion typically reveals increased intron retention, exon skipping, and utilization of cryptic splice sites.
To identify direct PHF5A-regulated splicing events, researchers should implement integrated methodological approaches. CLIP-seq (Cross-linking immunoprecipitation sequencing) directly maps PHF5A-RNA interactions in vivo . Comparing acute versus chronic depletion effects through time-course experiments helps distinguish primary from secondary effects. Rescue experiments with wildtype versus mutant PHF5A can validate direct targets. Combining these data with motif analysis around affected splice sites can reveal sequence features that confer PHF5A dependency. Integrating results with other SF3B component binding data strengthens evidence for direct regulation. Computational modeling incorporating RNA structure predictions further refines the identification of direct targets versus downstream effects.
PHF5A expression shows significant variation between normal and tumor tissues across multiple cancer types, with generally elevated expression in cancerous tissues . Pan-cancer analysis demonstrates that PHF5A can serve as both a diagnostic and prognostic marker . The expression patterns correlate with clinical outcomes in various cancers, suggesting utility as a biomarker . Cancer-specific expression patterns may reflect differential requirements for PHF5A-mediated splicing regulation in different tumor types. Immunohistochemical analysis of PHF5A in tumor samples can contribute to cancer classification and prognostication . Methodologically, researchers should employ tissue microarrays and quantitative immunohistochemistry to standardize PHF5A evaluation across cancer types.
PHF5A promotes cancer progression through multiple mechanisms. Its aberrant expression drives oncogenic splicing patterns that favor tumor growth and invasion . The phosphorylation of PHF5A at Y36 by the TrkA-ERK1/2-ABL1 cascade regulates centrosome separation, affecting cell division and potentially contributing to genomic instability . Hyperacetylation of PHF5A is associated with enhanced tumor pathogenesis, though the precise mechanisms remain under investigation . In medulloblastoma, the TrkA-ERK1/2-ABL1-PHF5A phosphorylation cascade is hyperactivated, and its inhibition induces senescence and restricts proliferation . PHF5A also plays a critical role in tumor immunity, positioning it as a potential target for anticancer immunotherapy .
Several therapeutic approaches targeting PHF5A are being investigated in preclinical models. Inhibition of the TrkA-ERK1/2-ABL1 cascade that phosphorylates PHF5A at Y36 shows promise in restricting medulloblastoma proliferation by inducing cellular senescence . Small molecule inhibitors disrupting PHF5A's interaction with other SF3B components could selectively impair cancer-specific splicing events. RNA interference approaches directly targeting PHF5A expression demonstrate anti-tumor effects in multiple cancer models . Immunotherapy strategies leveraging PHF5A's role in tumor immunity are emerging as potential treatment modalities . Combination approaches targeting both PHF5A and related splicing factors may overcome resistance mechanisms and enhance therapeutic efficacy.
PHF5A maintains pluripotency through multiple interconnected mechanisms. It recruits and stabilizes the Polymerase Associated Factor 1 Complex (PAF1C) on pluripotency gene loci, regulating their expression . PHF5A influences the deposition of elongation-associated histone modifications, including H3K79me2 and H3K36me3, on PAF1C targets and self-renewal genes . It regulates RNA polymerase II promoter-proximal pause release of PAF1C targets and self-renewal genes, as well as the levels of elongating ('Ser-2' phosphorylated) RNA polymerase II in their gene bodies . These activities collectively preserve the transcriptional programs required for pluripotency maintenance. Experimental approaches tracking these mechanisms include ChIP-seq for histone modifications, PRO-seq for nascent transcription, and genetic interaction studies with pluripotency factors.
To study PHF5A's role in lineage-specific differentiation, researchers should employ: (1) embryonic stem cell differentiation protocols with inducible PHF5A perturbation at defined time points; (2) organoid systems that recapitulate tissue development with spatiotemporal control of PHF5A function; (3) CRISPR-engineered reporter systems tracking lineage markers during differentiation under varying PHF5A conditions; (4) mouse models with conditional PHF5A manipulation in specific tissues; and (5) single-cell sequencing approaches to resolve heterogeneous differentiation trajectories. PHF5A has been specifically implicated in muscle specification, where it regulates adult myoblast differentiation by stabilizing PAF1C on chromatin . Comparative studies across multiple lineages can identify common and tissue-specific roles for PHF5A during differentiation.
This apparent paradox reflects PHF5A's context-dependent functions. Researchers should investigate how post-translational modifications (particularly phosphorylation and acetylation) alter PHF5A activity during state transitions . Protein interaction studies across differentiation time courses can reveal dynamic changes in PHF5A's binding partners. Alternative splicing analysis during differentiation may uncover shifts in PHF5A-regulated isoform patterns that facilitate state transitions. Chromatin occupancy mapping (ChIP-seq or CUT&RUN) at different differentiation stages can track changes in PHF5A genomic targeting. The protein likely serves as a regulatory node that integrates multiple signals to either maintain pluripotency or permit differentiation, depending on the cellular context and modification state.
PHF5A contributes to genome stability through both direct and indirect mechanisms. It participates in DNA damage repair by regulating or stabilizing histone chaperone complexes involved in the deposition of histone H2A variants, including H2AX and H2AZ . These variants are crucial for the recruitment of repair proteins to DNA damage sites and maintenance of chromatin integrity. PHF5A's splicing function ensures proper expression of DNA repair genes, while its non-spliceosomal activities may directly influence chromatin structure at damage sites. To investigate these connections, researchers should employ comet assays, γH2AX foci formation analysis, chromosomal aberration studies, and DNA repair capacity assays following PHF5A manipulation.
The phosphorylation of PHF5A at Y36 by the TrkA-ERK1/2-ABL1 cascade plays a critical role in regulating centrosome separation . Phosphorylated PHF5A promotes the interaction between CEP250 and Nek2A in a spliceosomal-independent manner, leading to premature centrosome separation . This premature separation can remodel microtubule organization and subsequently affect cell proliferation and migration . Dysregulation of this process potentially contributes to chromosomal instability, a hallmark of cancer. Research methodologies should include live-cell imaging of centrosome dynamics, super-resolution microscopy of mitotic spindles, and chromosome segregation analysis following manipulation of PHF5A phosphorylation status through phosphomimetic or phospho-deficient mutations.
To investigate this interplay, researchers should implement: (1) RNA-seq following DNA damage induction in cells with normal versus depleted/mutated PHF5A to identify damage-responsive splicing events; (2) splicing-reporter assays containing DNA repair gene introns to directly assess PHF5A's impact; (3) chromatin proteomics comparing PHF5A-associated proteins before and after DNA damage; (4) CLIP-seq to map changes in PHF5A-RNA interactions following genotoxic stress; (5) functional repair assays (e.g., homologous recombination and non-homologous end joining reporters) following PHF5A perturbation; and (6) real-time tracking of repair factor recruitment in PHF5A-deficient backgrounds. Integration of these approaches can establish whether PHF5A coordinates splicing and repair processes and how this coordination is regulated under genotoxic stress.
PHF5A undergoes multiple post-translational modifications that significantly impact its function. Phosphorylation at Y36 regulates its non-spliceosomal role in centrosome separation through the TrkA-ERK1/2-ABL1 cascade . Hyperacetylation of PHF5A has been linked to enhanced tumor pathogenesis, suggesting a regulatory role in cancer progression . These modifications likely create a "code" that determines which function of PHF5A predominates in a given cellular context. To comprehensively map PHF5A modifications, researchers should apply mass spectrometry-based proteomics following enrichment of post-translationally modified proteins. Functional studies comparing wild-type PHF5A with modification-mimetic or modification-resistant mutants can reveal the biological significance of each modification.
The TrkA-ERK1/2-ABL1 signaling cascade regulates PHF5A phosphorylation at Y36, a modification critical for centrosome separation . This pathway can be experimentally manipulated using: (1) small molecule inhibitors targeting specific kinases in the cascade (TrkA inhibitors, MEK inhibitors for ERK1/2, and ABL1 inhibitors like imatinib); (2) constitutively active or dominant-negative kinase mutants; (3) phosphatase inhibitors to prolong phosphorylation; (4) optogenetic systems for spatiotemporal control of kinase activity; and (5) CRISPR-mediated genomic editing of phosphorylation sites. Monitoring PHF5A phosphorylation status using phospho-specific antibodies or mass spectrometry following these manipulations reveals pathway dynamics. The hyperactivation of this cascade in medulloblastoma suggests therapeutic potential in targeting these pathways .
To analyze PHF5A modifications across cellular compartments, researchers should employ: (1) subcellular fractionation followed by modification-specific western blotting; (2) proximity ligation assays to visualize modified PHF5A interactions in situ; (3) mass spectrometry with SILAC or TMT labeling for quantitative comparison across compartments; (4) immunofluorescence with modification-specific antibodies combined with super-resolution microscopy; (5) FRET-based biosensors to track PHF5A modification dynamics in living cells; and (6) ChIP-seq with modification-specific antibodies to identify genomic regions bound by differently modified PHF5A. For comprehensive mapping, combining these approaches with synchronization protocols can reveal cell cycle-dependent changes in PHF5A modification patterns and localization.
Multi-omics integration provides a comprehensive view of PHF5A function by connecting different layers of regulation. Researchers should combine: (1) transcriptomics (RNA-seq) to identify PHF5A-dependent gene expression and splicing patterns; (2) proteomics to map PHF5A interaction networks and post-translational modifications; (3) epigenomics (ChIP-seq, ATAC-seq) to characterize chromatin states associated with PHF5A binding; (4) genomics to identify genetic variations affecting PHF5A function; and (5) metabolomics to detect downstream metabolic consequences. Computational integration through network analysis, pathway enrichment, and machine learning approaches can identify emergent properties not apparent in single-omics analyses. Time-course studies across these platforms during processes like differentiation or DNA damage response can reveal dynamic regulatory networks centered on PHF5A.
To predict PHF5A-dependent splicing events, researchers should employ: (1) machine learning algorithms trained on CLIP-seq data and known regulated events; (2) RNA structure prediction tools to identify branch points and splicing signals potentially sensitive to PHF5A; (3) splicing-specific analysis software (e.g., rMATS, MISO, VAST-TOOLS) applied to RNA-seq data from PHF5A perturbation experiments; (4) network analysis connecting splicing changes to downstream functional pathways; (5) conservation analysis across species to identify evolutionarily conserved PHF5A-dependent events; and (6) integrative analysis with other splicing factor binding data to identify cooperative or competitive regulation. These computational approaches should be validated through targeted experimental confirmation using minigene splicing reporters or antisense oligonucleotides targeting specific splice sites.
To reconcile contradictory findings, researchers should implement: (1) standardized experimental protocols across multiple cell types and model organisms to directly compare PHF5A function; (2) comprehensive characterization of PHF5A expression levels, isoforms, and modifications in each system; (3) rescue experiments using the same PHF5A constructs across different models; (4) detailed time-course studies to distinguish between immediate and adaptive responses to PHF5A perturbation; (5) genetic interaction screens to identify context-dependent cofactors; and (6) targeted studies addressing specific discrepancies using identical methodologies across systems. Creating a matrix of PHF5A functions across different cellular contexts can help identify conditional factors that determine which PHF5A activities predominate in a given system, potentially explaining seemingly contradictory observations.
PHD Finger Protein 5A (PHF5A) is a highly conserved protein that plays a crucial role in the regulation of gene expression. It is a member of the plant homeodomain (PHD) finger protein family, which is known for its involvement in chromatin remodeling and transcriptional regulation. The PHD finger domain is a specialized zinc finger motif that can bind to specific histone modifications, acting as an “epigenome reader” to mediate the activation or repression of underlying genes .
PHF5A contains a PHD-finger-like domain flanked by highly basic N- and C-termini. This domain is essential for its function as a chromatin-associated protein. PHF5A is a component of the 17S U2 small nuclear ribonucleoprotein (snRNP) complex of the spliceosome, a large ribonucleoprotein complex that removes introns from transcribed pre-mRNAs . Within this complex, PHF5A is part of the SF3B subcomplex, which is required for the assembly of the ‘A’ complex formed by the stable binding of U2 snRNP to the branchpoint sequence in pre-mRNA .
PHF5A is involved in pre-mRNA splicing, a critical process in gene expression. It directly participates in early spliceosome assembly and mediates the recognition of the intron branch site during pre-mRNA splicing by promoting the selection of the pre-mRNA branch-site adenosine, the nucleophile for the first step of splicing . This function is vital for the accurate and efficient removal of introns from pre-mRNA, ensuring the proper expression of genes.
Recent studies have highlighted the role of PHF5A as an oncoprotein in various cancers, including non-small cell lung cancer (NSCLC). PHF5A has been found to be highly upregulated in lung adenocarcinoma (LAC) tissues compared to adjacent non-tumor tissues . Its expression is closely associated with tumor progression and poor patient prognosis . Functional studies have demonstrated that PHF5A knockdown results in reduced cell proliferation, increased cell apoptosis, cell cycle arrest, and suppressed migration and invasion in LAC cells . These findings suggest that PHF5A contributes to LAC progression by regulating multiple signaling pathways and may serve as a potential therapeutic target in NSCLC .