PIN4 catalyzes cis-trans isomerization of proline residues, modulating:
Binds to AT-rich DNA sequences via its N-terminal domain.
Associates with preribosomal ribonucleoprotein complexes, suggesting roles in ribosome biogenesis .
Components:
Diagnostic Scenario | PIN4 Stain Result | Interpretation |
---|---|---|
Adenocarcinoma | AMACR+; p63/HMWK- | Malignant |
Benign Mimickers | AMACR-; p63/HMWK+ | Non-malignant |
Atypical Small Acinar Proliferation | AMACR-; p63/HMWK- | Requires further evaluation |
PIN4 knockdown (KD): Reduces HBc protein stability, cccDNA levels, and virion secretion by >70% compared to PIN1 KD .
Key data:
Saccharomyces cerevisiae Pin4: Binds glycolytic and mitochondrial mRNAs under glucose-rich conditions; regulates transcriptional reprogramming during starvation .
PIN4 (Peptidyl-prolyl cis-trans isomerase NIMA-interacting 4) is a human protein encoded by the PIN4 gene that belongs to the PpiC/parvulin rotamase family, specifically the PIN4 subfamily . The protein exists in multiple isoforms, with isoform 1 functioning primarily as a ribosomal RNA processing factor involved in ribosome biogenesis . PIN4 demonstrates the ability to bind to tightly bent AT-rich stretches of double-stranded DNA, suggesting a potential role in DNA replication or gene expression regulation . The protein exhibits peptidyl-prolyl cis-trans isomerase activity, catalyzing the isomerization of proline residues, which can significantly impact protein folding and function in cellular contexts .
PIN4 has multiple isoforms, with isoform 1 (also known as Par14 or hPar14) and isoform 2 (also known as Par17 or hPar17) being the most extensively studied . Isoform 1 functions predominantly as a ribosomal RNA processing factor essential for ribosome biogenesis and binds to particular DNA configurations, specifically tightly bent AT-rich stretches of double-stranded DNA . Isoform 2 has been characterized primarily for its DNA-binding capabilities, interacting with double-stranded DNA without the specificity for AT-rich regions exhibited by isoform 1 . The functional differences between these isoforms are largely attributed to their distinct subcellular localization patterns and post-translational modification states, which regulate their participation in various cellular processes including ribosome assembly and potentially transcriptional regulation .
PIN4 undergoes multiple post-translational modifications that significantly influence its functionality and localization within the cell. The most extensively documented PTMs include:
Site | PTM Type | PTM Enzyme | Functional Implication |
---|---|---|---|
K6, K11, K32, K47, K75 | Acetylation | - | Potentially regulates protein-protein interactions |
S19 | Phosphorylation | CSNK2A1 (Casein Kinase II) | Required for nuclear localization; does not affect PPIase activity |
S18, S24, Y122 | Phosphorylation | - | Potential regulatory functions being investigated |
C45 | S-Nitrosylation | - | May affect protein structure and function under oxidative stress |
K47, K75 | Ubiquitination | - | Likely regulates protein turnover and degradation |
Importantly, the phosphorylation state of isoform 1, particularly at Ser-19, is critical for its subcellular localization but does not affect its peptidyl-prolyl isomerase activity . Dephosphorylation of isoform 1 is a prerequisite for DNA binding, indicating a regulatory mechanism for its interaction with nucleic acids . The unphosphorylated form of isoform 1 associates with pre-rRNP complexes in the nucleus, highlighting the importance of this PTM in regulating PIN4's role in ribosome biogenesis .
To investigate PIN4's function in ribosome biogenesis, researchers should implement a multi-faceted approach combining molecular, cellular, and biochemical techniques. RNA immunoprecipitation followed by sequencing (RIP-seq) provides a powerful method to identify specific RNA targets of PIN4, particularly pre-rRNA species . Co-immunoprecipitation assays with known ribosome assembly factors can elucidate PIN4's interactome within the ribosome biogenesis pathway . For studying localization and dynamics, fluorescence recovery after photobleaching (FRAP) with GFP-tagged PIN4 variants would allow visualization of nucleolar trafficking patterns under various cellular conditions .
Methodologically, researchers should consider using CRISPR-Cas9 gene editing to create PIN4 knockout or phospho-mutant cell lines (particularly S19A and S19E mutations) to assess the specific impact of phosphorylation on ribosome assembly . Ribosome profiling combined with polysome analysis in these modified cell lines would quantitatively measure the effects of PIN4 manipulation on translation efficiency and ribosome assembly rates . Additionally, structural studies using cryo-electron microscopy of PIN4 in complex with pre-ribosomes would provide atomic-level insights into its mechanism of action during ribosome maturation .
Investigating PIN4's DNA binding characteristics requires sophisticated biophysical and molecular techniques tailored to understand both binding specificity and structural interactions. Electrophoretic mobility shift assays (EMSA) with systematically designed DNA sequences varying in AT content provide a foundational approach to quantify binding affinity and specificity . More advanced biophysical techniques such as isothermal titration calorimetry (ITC) and surface plasmon resonance (SPR) offer quantitative measurements of binding kinetics and thermodynamics, essential for understanding the physical basis of PIN4-DNA interactions .
For in-depth structural analysis, a combination of X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy of PIN4 complexed with specific DNA sequences would reveal atomic-level details of the interaction interface . Chromatin immunoprecipitation followed by sequencing (ChIP-seq) can identify genome-wide binding sites of PIN4 in vivo, while DNase I footprinting assays would pinpoint the exact nucleotides protected by PIN4 binding . When designing these experiments, researchers should account for the critical influence of phosphorylation status on DNA binding by using both phosphorylated and dephosphorylated PIN4 preparations, potentially generated through site-directed mutagenesis (S19A to mimic dephosphorylation) or through in vitro phosphatase treatment .
The investigation of PIN4 variants in disease contexts, especially cancer, requires integrated genomic, proteomic, and functional approaches. Mining cancer genomic databases (TCGA, COSMIC) for PIN4 mutations, particularly focusing on the K75N variant associated with lung cancer, provides the foundation for connecting genetic alterations to disease phenotypes . Patient-derived xenograft models harboring PIN4 mutations offer platforms for in vivo functional studies and potential therapeutic interventions .
Methodologically, researchers should employ CRISPR-Cas9 to introduce specific PIN4 mutations (such as K75N) into normal cell lines and assess resulting phenotypic changes in proliferation, migration, and ribosome biogenesis . Comparative proteomics using stable isotope labeling with amino acids in cell culture (SILAC) between wild-type and mutant PIN4-expressing cells would reveal altered protein interaction networks . RNA-seq analysis of these isogenic cell lines could identify transcriptional programs disrupted by PIN4 variants . For translational relevance, immunohistochemical analysis of PIN4 expression and phosphorylation status in cancer tissue microarrays, correlated with clinical outcomes, would establish its potential as a biomarker . Additionally, high-throughput drug screening using PIN4 mutant cells might identify compounds that specifically target cells with aberrant PIN4 function, potentially leading to precision medicine approaches for cancers harboring PIN4 alterations .
When investigating PIN4 phosphorylation and its effects on cellular localization, researchers must implement rigorous control measures to ensure valid interpretations. Phosphorylation-null mutants (S19A) and phosphomimetic mutants (S19E) of PIN4 serve as critical controls for distinguishing phosphorylation-dependent effects . When conducting immunofluorescence or subcellular fractionation studies, researchers should include treatment with phosphatase inhibitors in one condition and active phosphatases in another to maintain or remove phosphorylation, respectively .
For experimental validation, parallel tracking of known nuclear and cytoplasmic markers alongside PIN4 provides essential reference points for localization changes . Time-course experiments following treatment with kinase inhibitors (particularly CSNK2A1 inhibitors) or phosphatase inhibitors are necessary to establish the dynamic relationship between phosphorylation status and localization . To eliminate potential artifacts, researchers should compare multiple detection methods, including immunofluorescence, subcellular fractionation with Western blotting, and live-cell imaging with fluorescently tagged PIN4 . Additionally, mass spectrometry-based quantification of phosphorylation at specific residues provides an essential control to confirm the phosphorylation status in various experimental conditions, ensuring that observed localization changes are indeed correlated with the intended phosphorylation manipulations .
Investigating PIN4's function in ribosomal biogenesis presents several methodological challenges that require careful experimental design. To address the rapid dynamics of ribosome assembly, pulse-chase labeling of rRNA with 5-fluorouracil combined with immunoprecipitation of PIN4 can capture transient interactions . The complexity of distinguishing direct from indirect effects necessitates the use of catalytically inactive PIN4 mutants alongside wild-type controls to differentiate between structural and enzymatic contributions to ribosome formation .
The heterogeneity of pre-ribosomal complexes requires gradient fractionation techniques coupled with mass spectrometry to precisely identify the specific pre-ribosomal particles with which PIN4 associates . To mitigate the challenge of nuclear/nucleolar targeting, researchers should employ nuclear export inhibitors (such as leptomycin B) to prevent cytoplasmic shuttling of PIN4 during the experimental timeframe . When investigating the kinetics of PIN4's role in ribosome maturation, selective inhibition of transcription (with actinomycin D) at various time points, followed by PIN4 immunoprecipitation, allows for temporal mapping of PIN4's engagement with pre-ribosomes . Additionally, researchers should consider implementing parallel studies in different cell types with varying ribosome production rates to determine whether PIN4's function is universally conserved or context-dependent .
The production of functional recombinant PIN4 protein for in vitro studies requires careful optimization of expression and purification conditions to preserve native structure and activity. Expression systems should be selected based on the specific experimental requirements: E. coli systems (as used in available commercial preparations) provide high yields suitable for structural studies, while mammalian or insect cell systems are preferable when post-translational modifications are crucial to the investigation . The inclusion of a cleavable His-tag (MGSSHHHHHHSSGLVPRGSHM) facilitates initial purification while allowing for tag removal to prevent interference with functional assays .
Buffer optimization is critical, with most successful preparations utilizing buffers containing stabilizing agents such as 10% glycerol or low concentrations of reducing agents to prevent oxidation of cysteine residues, particularly C45 which is known to undergo S-nitrosylation . Temperature control during expression (typically 18-25°C) helps prevent inclusion body formation and enhance soluble protein yield . For purification, a multi-step approach combining initial affinity chromatography (Ni-NTA) followed by ion exchange and size exclusion chromatography produces the highest purity preparations (>95%) with minimal endotoxin contamination (<1 EU/μg) .
Quality control measures should include SDS-PAGE, HPLC, and circular dichroism to verify structural integrity, while functional validation through peptidyl-prolyl isomerase activity assays and DNA binding assays confirms that the recombinant protein retains native functionality . For experiments requiring specific phosphorylation states, in vitro kinase treatments (using purified CSNK2A1 for S19 phosphorylation) or phosphatase treatments should be performed, with phosphorylation status confirmed by mass spectrometry or phospho-specific antibodies .
Addressing contradictory findings about PIN4's cellular functions requires systematic meta-analysis and standardized experimental approaches. Researchers should begin by documenting all experimental variables across conflicting studies, including cell types, PIN4 isoforms examined, detection methods, and experimental conditions . Meta-analysis of these variables can often reveal that apparent contradictions stem from context-dependent functions of PIN4 rather than truly contradictory findings .
When differences persist, researchers should design experiments that directly test competing hypotheses within a single experimental system, using multiple complementary techniques to measure the same outcome . For instance, if contradictions exist regarding PIN4's role in ribosome biogenesis, researchers should employ both biochemical ribosome profiling and imaging-based approaches in the same cell lines . Collaboration between laboratories reporting contradictory results, using standardized reagents (such as verified antibodies and cell lines) and protocols, can significantly clarify sources of variation .
For data interpretation, researchers should adopt Bayesian statistical frameworks that can incorporate prior knowledge while evaluating new evidence, allowing for quantitative assessment of competing hypotheses . Additionally, computational modeling of PIN4's interaction networks across different cellular conditions may reconcile seemingly contradictory observations by identifying condition-specific interaction partners or regulatory mechanisms . Finally, considering evolutionary perspectives by examining PIN4 functions across species can provide valuable context for understanding apparently divergent functions as specialized adaptations of a conserved molecular mechanism .
The analysis of PIN4 post-translational modification (PTM) data demands sophisticated statistical approaches tailored to the complexities of PTM dynamics and detection methods. For mass spectrometry-based PTM identification, researchers should implement target-decoy approaches to control false discovery rates, with a threshold typically set at <1% for high-confidence PTM site assignments . When quantifying relative PTM abundances across conditions, normalization methods must account for both total protein abundance changes and potential ionization efficiency differences .
Time-course PTM data should be analyzed using longitudinal statistical models rather than multiple pairwise comparisons, reducing type I errors while capturing temporal trends . For integrating multiple PTM types (phosphorylation, acetylation, ubiquitination) on PIN4, multivariate statistical methods such as principal component analysis or partial least squares discriminant analysis help identify PTM signatures associated with specific cellular states or treatments .
The co-occurrence of multiple PTMs requires analysis of potential crosstalk, using conditional probability models to identify PTM sites that display statistical dependencies . Researchers should employ hierarchical Bayesian models when analyzing PTM data across multiple experimental replicates, as these models appropriately handle both biological and technical variability . For relating PTM patterns to functional outcomes, machine learning approaches such as random forests or support vector machines can identify predictive PTM signatures while accounting for complex non-linear relationships . Additionally, researchers should consider employing structural bioinformatics approaches to map PTM sites onto known protein structures or models, providing context for interpreting statistical associations in terms of mechanistic hypotheses about how specific PTMs affect PIN4 function .
Interpreting PIN4 localization changes under cellular stress requires careful consideration of multiple factors to distinguish specific responses from general cellular perturbations. Researchers should first establish baseline PIN4 localization dynamics in unstressed cells across the cell cycle, as cell cycle-dependent changes could confound stress response interpretations . Time-course experiments capturing both acute and chronic stress responses provide crucial information about the kinetics of PIN4 relocalization and potential adaptation .
Methodologically, researchers should employ multiple complementary techniques to confirm localization changes, including live-cell imaging of fluorescently tagged PIN4, immunofluorescence of endogenous PIN4, and biochemical fractionation followed by Western blotting . Colocalization analysis with markers for specific subcellular compartments (nucleoli, splicing speckles, stress granules) helps precisely define the nature of stress-induced relocalization .
When interpreting results, researchers must discriminate between PIN4-specific responses and global cellular changes by comparing PIN4 dynamics with control proteins that share similar baseline localization but different functions . Phosphorylation status at S19 should be monitored simultaneously with localization, given its established role in regulating nuclear localization . Correlation analysis between stress-induced PIN4 relocalization patterns and functional outcomes (changes in ribosome biogenesis, DNA binding, etc.) helps establish the biological significance of observed localization changes . Additionally, researchers should consider whether observed changes might represent protective adaptations or contribute to pathological processes by correlating PIN4 behavior with cell survival outcomes following stress resolution .
The PIN4 gene was identified through a search of an expressed sequence tag (EST) database using human PIN1 and Escherichia coli parvulin sequences as probes. This led to the isolation of a cDNA encoding PIN4 from a lung cDNA library . The gene encoding PIN4 is located on the X chromosome and is expressed in various tissues, indicating its fundamental role in cellular processes .
PIN4 has a unique structure that includes a peptidyl-prolyl isomerase domain, which is responsible for its enzymatic activity. This domain contains a nucleophilic cysteine residue (Cys113) that is crucial for its function . The enzyme catalyzes the cis/trans isomerization of proline residues in polypeptides, a process that is essential for proper protein folding and function .
PIN4 plays a significant role in several cellular processes, including the cell cycle, chromatin remodeling, and ribosome biogenesis . It is involved in the regulation of various signaling pathways by inducing conformational changes in key signaling molecules following proline-directed phosphorylation . This regulation is critical for maintaining cellular homeostasis and function.
The deregulation of PIN4 has been implicated in various diseases, particularly cancer. Overexpression of PIN4 has been observed in several types of cancer, including breast, cervical, ovarian, and endometrial cancers . This overexpression promotes cell proliferation and transformation, contributing to tumorigenesis . Targeting PIN4 with specific inhibitors has shown promise in reducing tumor progression and increasing survival in preclinical models .
Recent research has focused on developing specific inhibitors for PIN4 to explore its therapeutic potential. One such inhibitor, Sulfopin, has demonstrated high affinity binding to PIN4 and inhibition of its catalytic activity . In preclinical studies, Sulfopin treatment led to tumor regression and increased survival in mouse models of cancer . These findings highlight the potential of PIN4 as a therapeutic target for cancer treatment.