PIAS4, also known as PIASy, PIASG, or E3 SUMO-protein ligase PIAS4, belongs to the PIAS family of proteins that function as transcriptional coregulators in various cellular pathways, including STAT signaling, p53 pathway, and steroid hormone signaling . PIAS4 primarily functions as an E3 SUMO ligase, facilitating the conjugation of small ubiquitin-like modifier (SUMO) proteins to target substrates .
Recent research has revealed PIAS4's significance in:
Intrinsic antiviral immunity against HSV-1 infection, representing a novel aspect of PIAS protein function
Hypoxia signaling regulation through VHL suppression in pancreatic cancer
DNA damage response pathways by mediating sumoylation of proteins like PARP1 and KAT5
These diverse functions make PIAS4 an important research target for understanding fundamental cellular processes and disease mechanisms.
The choice between monoclonal and polyclonal PIAS4 antibodies depends on the specific research objectives:
Polyclonal PIAS4 antibodies (such as those from Proteintech and Rockland):
Recognize multiple epitopes, providing stronger signals in applications like WB and IHC
Useful for detecting native proteins in various applications
Better for detecting proteins with post-translational modifications or when protein conformation may vary
Monoclonal PIAS4 antibodies (such as Cell Signaling's D2F12):
Provide superior lot-to-lot consistency and specificity
Often preferred for quantitative applications requiring reproducible results
The D2F12 antibody specifically recognizes regions surrounding Lys59 of human PIAS4
Particularly useful when specific isoforms or domains need to be distinguished
For exploratory research or when dealing with samples where PIAS4 may be modified or present at low levels, polyclonal antibodies may provide better detection. For standardized assays or when absolute specificity is required, monoclonal antibodies are preferable.
PIAS4 localizes to both nuclear and cytoplasmic compartments, with its distribution changing depending on cellular conditions and stimuli. Effective sample preparation is crucial for accurate detection:
Nuclear fraction enrichment:
Use specialized nuclear extraction buffers containing 10-20 mM HEPES (pH 7.9), 0.4 M NaCl, 1 mM EDTA, and 10% glycerol
Include SUMO protease inhibitors (20 mM N-ethylmaleimide) to preserve sumoylated forms
Add phosphatase inhibitors when studying PIAS4's roles in signaling pathways
Following HSV-1 infection, modified extraction techniques may be necessary to capture PIAS4 in viral replication compartments
Cytoplasmic detection:
Use lower salt buffers (150 mM NaCl) supplemented with 0.5% NP-40 or Triton X-100
When studying PIAS4's interaction with membrane-associated proteins, consider digitonin-based fractionation
Whole cell lysates:
RIPA buffer supplementation with 1% SDS can improve detection of PIAS4, which often appears at 57-70 kDa
Sonication is recommended to shear DNA and release chromatin-bound PIAS4
When analyzing PIAS4 in pancreatic cancer samples, researchers should note that detection can be improved using specialized extraction methods, as PIAS4 expression is significantly elevated in these tissues compared to normal pancreatic epithelium .
Rigorous validation of PIAS4 antibodies requires multiple controls:
Positive controls:
Verified cell lines with confirmed PIAS4 expression (K-562, HL-60, Panc0327, and Panc1005 cells)
Recombinant PIAS4 protein (full-length or the specific domain to which the antibody was raised)
Mouse ovary or pancreas tissue, which show reliable PIAS4 expression
Negative controls:
PIAS4 knockdown/knockout samples using validated siRNAs targeting exon 2 or exon 6
Primary cells with naturally low PIAS4 expression (such as AsPc1 and BxPc3)
Peptide competition assays using the immunizing peptide to confirm binding specificity
Specificity controls:
Cross-reactivity testing with other PIAS family members (PIAS1, PIAS2, PIAS3) to ensure specificity
Secondary antibody-only controls to rule out non-specific binding
Isotype controls matched to the host species and antibody class
In published studies, siRNA-mediated knockdown has been particularly effective for validating PIAS4 antibody specificity, with researchers successfully targeting PIAS4 using both pooled siRNAs and individual siRNAs against specific exons .
Optimal immunofluorescence protocols for PIAS4 detection vary depending on the cellular context and research question:
Standard cultured cells (e.g., HeLa):
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.2% Triton X-100 for 10 minutes
Block with 5% BSA or 10% serum for 1 hour
Incubate with primary antibody at dilutions of 1:50-1:500 overnight at 4°C
Use appropriate fluorophore-conjugated secondary antibodies (1:500-1:1000)
Counterstain nuclei with DAPI to visualize nuclear localization
Virus-infected cells (e.g., HSV-1 infection):
Fix with 1.8% formaldehyde in PBS containing 2% sucrose
Permeabilize with 0.5% NP-40, 10% sucrose in PBS
For detecting PIAS4 at sites of viral genome entry, image acquisition within 1-2 hours post-infection is critical
For replication compartment localization, imaging at 4-6 hours post-infection is recommended
Tissue sections:
Use heat-induced epitope retrieval (citrate buffer pH 6.0)
Longer primary antibody incubation (overnight at 4°C)
For pancreatic tissue, background reduction with 0.3% hydrogen peroxide is recommended
When co-staining with markers like HIF1α, sequential staining protocols may be necessary
For all applications, including a detergent in wash buffers (0.05% Tween-20) helps reduce background. When studying PIAS4's SUMO E3 ligase activity, co-staining with SUMO1/2/3 antibodies can provide valuable functional information.
PIAS4 plays a crucial role in hypoxia signaling through its interaction with VHL (von Hippel-Lindau) protein and subsequent impact on HIF1α, making it particularly relevant in cancer research . To effectively study this pathway:
Immunoprecipitation approach:
Use anti-PIAS4 antibodies (1:100 dilution) for immunoprecipitation from cell lysates
Co-immunoprecipitate with anti-VHL antibodies to detect direct interaction
Probe for sumoylated forms of VHL using anti-SUMO2/3 antibodies
Compare normoxic and hypoxic conditions (1% O₂, 48 hours) to detect differential interactions
Chromatin immunoprecipitation (ChIP):
Use PIAS4 antibodies to identify genomic regions where PIAS4 functions as a transcriptional coregulator
Cross-link proteins to DNA using 1% formaldehyde for 10 minutes
Sonicate to generate 200-500 bp DNA fragments
Immunoprecipitate with 5 μg of anti-PIAS4 antibody
Analyze enrichment at HIF1α target genes like VEGF and JMJD1A by qPCR
In published studies, researchers have demonstrated that PIAS4 siRNA suppresses the expression of HIF1α in pancreatic cancer cells, and when these cells were exposed to hypoxia for 48 hours, the induction of HIF1α downstream targets (VEGF and JMJD1A) was also attenuated . This methodological approach provides valuable insights into PIAS4's role in hypoxia signaling.
Recent research has revealed a novel role for PIAS4 as an intrinsic antiviral factor against herpes simplex virus 1 (HSV-1) infection . To investigate this function:
Subcellular localization during viral infection:
Infect cells with HSV-1 at MOI of 0.1-10
At different time points post-infection (1, 2, 4, 8 hours), fix and immunostain with anti-PIAS4 antibodies
Co-stain with viral proteins (e.g., ICP0, ICP4) to track infection progression
Use confocal microscopy to determine PIAS4's localization to viral genome entry sites or replication compartments
Functional studies:
Deplete PIAS4 using validated siRNAs targeting exon 2 or exon 6
Infect control and PIAS4-depleted cells with ICP0-null mutant HSV-1
Quantify viral replication by plaque assays or qPCR of viral genes
Compare with PML (promyelocytic leukemia protein) depletion to assess synergistic effects
Domain-specific recruitment analysis:
Generate constructs expressing PIAS4 mutants lacking specific domains:
SAP domain (DNA binding)
PINIT domain (substrate recognition)
SP-RING domain (SUMO ligase activity)
SIM motifs (SUMO-interaction)
Assess recruitment to viral genome entry sites and replication compartments using IF
Use live-cell imaging with fluorescently tagged PIAS4 to track dynamics during infection
Research has shown that PIAS4 relocalizes to sites associated with viral DNA throughout infection, with SIM-dependent mechanisms at genome entry sites and SIM-independent mechanisms in replication compartments . These methodological approaches can reveal the mechanisms of PIAS4's antiviral activities.
PIAS4 functions as an E3 SUMO ligase, mediating the sumoylation of various substrate proteins. To analyze this enzymatic activity:
In vitro sumoylation assays:
Express and purify recombinant PIAS4 protein
Set up reaction mixtures containing:
E1 enzyme (SAE1/SAE2)
E2 enzyme (UBC9)
SUMO1, SUMO2, or SUMO3 proteins
ATP regeneration system
Purified substrate protein (e.g., GATA2, VHL, p53)
Incubate at 30°C for 1-3 hours
Analyze by Western blot using anti-SUMO and substrate-specific antibodies
Cellular sumoylation studies:
Co-transfect cells with:
Tagged SUMO constructs (His6-SUMO or HA-SUMO)
PIAS4 expression vector or siRNA
Substrate protein
Lyse cells in denaturing conditions (8M urea buffer)
For His-tagged SUMO, perform Ni-NTA pulldown
For HA-tagged SUMO, perform anti-HA immunoprecipitation
Detect sumoylated substrates by Western blot
Domain mutant analysis:
Generate PIAS4 mutants:
C342A (SP-RING domain mutant that abolishes E3 ligase activity)
Lys35 mutant (affects auto-sumoylation site)
Compare wild-type and mutant PIAS4 in sumoylation assays
Use CRISPR/Cas9 to generate endogenous PIAS4 domain mutants
In pancreatic cancer research, investigators have shown that PIAS4-mediated sumoylation of VHL leads to its oligomerization and reduced tumor suppressor activity against HIF1α . Similar methodological approaches can be applied to study PIAS4's E3 ligase activity toward other substrates in different biological contexts.
Researchers often encounter technical issues when working with PIAS4 antibodies. Here are methodological solutions to common problems:
Multiple bands in Western blot:
PIAS4 can appear at 57-70 kDa due to post-translational modifications, particularly sumoylation
Include N-ethylmaleimide (20 mM) in lysis buffers to preserve SUMO modifications
Use gradient gels (4-15%) to better resolve different modified forms
Validate bands using PIAS4 knockdown/knockout controls
Consider that the 57 kDa band corresponds to the calculated molecular weight, while higher bands may represent modified forms
Weak or no signal in immunostaining:
Optimize fixation (try 4% PFA and methanol fixation in parallel)
Test different permeabilization methods (0.1-0.5% Triton X-100, 0.1% SDS, or methanol)
Increase antibody concentration to 1:50 for initial optimization
Use tyramide signal amplification for low-abundance detection
Try heat-mediated antigen retrieval even for cell samples (citrate buffer pH 6.0)
High background in immunoprecipitation:
Pre-clear lysates with protein A/G beads for 1 hour at 4°C
Use more stringent wash buffers containing 250-500 mM NaCl
Include 0.1% SDS in wash buffers to reduce non-specific binding
Cross-link antibodies to beads to prevent heavy chain interference in blotting
Use TrueBlot secondary antibodies to detect only native immunoglobulins
Inconsistent results between experiments:
Standardize cell culture conditions, as PIAS4 expression can vary with cell density
For hypoxia studies, tightly control O₂ levels and exposure times
Maintain consistent protein loading (30-50 μg total protein per lane)
Include positive control samples in each experiment (K-562 or HL-60 cells)
Studies have reported seemingly contradictory roles for PIAS4 in different cancer types, particularly regarding its effects on hypoxia signaling. Here's how to methodically approach and interpret such conflicting data:
Methodological approach to conflicting data:
Carefully analyze experimental conditions:
Examine substrate specificity:
Consider context-dependent interactions:
Validate with multiple approaches:
Use both gain-of-function (overexpression) and loss-of-function (siRNA) studies
Confirm findings with at least two different antibodies
Employ both in vitro and in vivo models when possible
Research has shown that in colon cancer, high PIAS4 expression was associated with enhanced HIF1α sumoylation and deactivation, while in pancreatic cancer, PIAS4 induction positively correlated with HIF1α activity through VHL suppression . These differences highlight the importance of cellular context in PIAS4 function.
Accurate quantification of PIAS4 requires appropriate statistical methods depending on the experimental technique:
Western blot quantification:
Use densitometry software (ImageJ, Image Lab) to measure band intensity
Normalize to appropriate loading controls (β-actin for whole cell, Lamin B for nuclear fractions)
For comparing multiple conditions:
Perform one-way ANOVA with Tukey's post-hoc test for multiple comparisons
Use paired t-tests for before/after treatments on the same samples
Present data as fold-change relative to control conditions
For time-course experiments, use repeated measures ANOVA
Immunofluorescence quantification:
For subcellular localization analysis:
Measure nuclear/cytoplasmic intensity ratio in at least 50-100 cells per condition
Use Pearson's correlation coefficient to quantify colocalization with other proteins
Apply Mann-Whitney U test for non-parametric data comparison
For focal structures (e.g., PIAS4 at viral genome entry sites):
Count foci number per cell
Measure foci intensity and size
Use Poisson distribution-based statistics for count data
RT-qPCR expression analysis:
Use 2^(-ΔΔCt) method with appropriate reference genes
Employ multiple reference genes (GAPDH, β-actin, 18S rRNA)
For pancreatic cancer studies, include at least 10-12 tumor samples and 6+ normal tissues
Apply non-parametric tests (Wilcoxon signed-rank) for clinical sample comparisons
Tissue microarray analysis:
Use H-score or Allred scoring systems for IHC quantification
Employ blinded scoring by at least two independent pathologists
Calculate inter-observer agreement using Cohen's kappa
Correlate with patient data using Kaplan-Meier survival analysis and Cox regression
Research has shown that PIAS4 mRNA levels were elevated approximately three-fold in pancreatic cancer cell lines and six-fold in fresh pancreatic tumors compared to normal pancreas (P=0.0154) , demonstrating the importance of rigorous statistical analysis in PIAS4 research.
PIAS4 presents an intriguing research target due to its seemingly contradictory roles in immune responses. While PIAS proteins typically suppress innate immune signaling, PIAS4 has been identified as a positive regulator of intrinsic antiviral immunity . Investigating this duality requires sophisticated methodological approaches:
Comparative viral infection models:
Challenge cells with different virus types:
DNA viruses (HSV-1, CMV, adenovirus)
RNA viruses (influenza, SARS-CoV-2)
Retroviruses (HIV)
Monitor PIAS4 localization and activity using IF and biochemical approaches
Generate time-course data to distinguish between early (intrinsic) and late (innate) immune responses
Pathway-specific dissection:
Simultaneously monitor multiple immune pathways in PIAS4-depleted cells:
cGAS-STING (DNA sensing)
RIG-I/MDA5 (RNA sensing)
IFN receptor signaling
NF-κB activation
Use reporter cell lines expressing luciferase under pathway-specific promoters
Employ multiplexed cytokine profiling to assess downstream effects
Domain-specific mutant analysis:
Create PIAS4 constructs with mutations in different functional domains:
SAP domain (DNA binding)
SIM motifs (SUMO interaction)
SP-RING domain (E3 ligase activity)
Assess each mutant's ability to:
Restrict viral replication
Suppress or enhance IFN signaling
Localize to different subcellular compartments
Identify domains responsible for pathway-specific functions
Research has demonstrated that PIAS4 is recruited to sites associated with HSV-1 genome entry through SIM-dependent mechanisms and accumulates in replication compartments through SIM-independent mechanisms . Understanding how the same protein can both enhance intrinsic immunity while suppressing innate immunity represents a significant research opportunity.
PIAS4 functions in DNA damage response pathways by mediating sumoylation of repair proteins, potentially influencing cancer therapy resistance. To investigate this aspect:
DNA damage induction models:
Treat cells with different DNA-damaging agents:
Ionizing radiation (Double-strand breaks)
Cisplatin (Interstrand crosslinks)
UV radiation (Pyrimidine dimers)
Hydroxyurea (Replication stress)
Track PIAS4 recruitment to damage sites using live-cell imaging
Quantify retention kinetics at damage sites in the presence/absence of key repair proteins
Repair pathway-specific analysis:
Measure repair efficiency in PIAS4-depleted cells using:
Comet assay for global DNA damage
DR-GFP assay for homologous recombination
NHEJ reporter assay for non-homologous end joining
Identify PIAS4-dependent sumoylation targets in each pathway using SUMO proteomics
Create cellular systems with inducible expression of PIAS4 mutants during DNA damage
Therapeutic resistance studies:
Generate isogenic cell lines with PIAS4 knockdown/overexpression
Assess sensitivity to:
PARP inhibitors (olaparib)
Platinum compounds
Radiotherapy
Topoisomerase inhibitors
Combine PIAS4 inhibition with established therapies to test for synergistic effects
PIAS4 has been implicated in the DNA damage response pathway and is thought to work in combination with PIAS1 for the productive association of repair factors like 53BP1, BRCA1, and RNF168 . Methodical investigation of these interactions may reveal PIAS4 as a potential therapeutic target for overcoming treatment resistance.
Recent findings suggest PIAS4 may play roles in neurodegenerative diseases through its regulation of protein homeostasis and stress responses . To investigate these emerging functions:
Protein aggregation models:
Express neurodegenerative disease-associated proteins in cellular models:
Tau (Alzheimer's disease)
α-synuclein (Parkinson's disease)
Huntingtin (Huntington's disease)
Manipulate PIAS4 levels using siRNA or overexpression
Quantify aggregate formation using filter trap assays and microscopy
Assess whether PIAS4-mediated sumoylation alters protein solubility and toxicity
Neuronal stress response studies:
Subject primary neurons or differentiated iPSCs to:
Oxidative stress (H₂O₂, paraquat)
ER stress (tunicamycin, thapsigargin)
Proteasome inhibition (MG132)
Monitor PIAS4 expression, localization, and activity changes
Determine if PIAS4 modulation can protect against stress-induced neuronal death
In vivo neurodegeneration models:
Generate conditional PIAS4 knockout mice using neuron-specific Cre lines
Cross with established neurodegenerative disease models
Assess:
Behavioral phenotypes
Histopathological markers
Protein aggregation burden
Neuroinflammatory signatures
The potential role of PIAS4 in neurodegenerative diseases represents an emerging research area. PIAS4 can interact with proteins like PARK7/DJ-1 involved in Parkinson's disease , and its SUMO E3 ligase activity may influence protein aggregation and clearance mechanisms central to neurodegeneration.