PURG antibodies are immunoglobulin molecules that specifically bind to the PURG protein, encoded by the PURG gene (Gene ID: 29942). PURG is a nucleic acid-binding protein critical for regulating gene expression and maintaining genomic stability . Antibodies targeting PURG enable researchers to investigate its localization, expression levels, and functional roles in cellular processes.
PURG antibodies are typically rabbit polyclonal or recombinant IgG molecules. Their validation involves rigorous testing across multiple applications:
Product Code | Host/Isotype | Applications | Reactivity | Dilution Range |
---|---|---|---|---|
84399-3-RR | Rabbit/IgG | WB, IF/ICC, ELISA | Human, Mouse, Rat | WB: 1:5k–1:50k; IF: 1:200–1:800 |
21750-1-AP | Rabbit/IgG | WB, ELISA | Human, Mouse, Rat | WB: 1:500–1:2000 |
Novus Biologicals’ antibody (NBP2-13830) further validates PURG’s nuclear localization in human testis and SH-SY5Y neuroblastoma cells via IHC and ICC .
PURG antibodies are pivotal in:
Western Blotting: Detecting PURG in brain tissues (mouse/rat) and HeLa cells .
Immunofluorescence: Visualizing PURG in nuclear and plasma membrane compartments .
Cancer Research: Anti-PrPc antibodies (related to PURG) inhibit colorectal cancer stem cell migration and tumor growth in mouse models, highlighting therapeutic potential .
Tumor Inhibition: Clone 6 (murine anti-PrPc antibody) reduced tumor growth in nude mice by >50% .
Diagnostic Sensitivity: Humanized antibodies (e.g., 4AA-M) outperformed commercial alternatives (8H4, 3F4) in detecting PrPc in Western blotting and IHC .
Recent guidelines emphasize the need for independent validation of antibodies to ensure specificity and reproducibility . Key steps include:
Application-Specific Testing: Confirm performance in intended assays (e.g., WB, IHC).
Negative Controls: Use knockout cell lines or tissues to rule out off-target binding.
Cross-Validation: Compare results with alternative antibodies or orthogonal methods (e.g., mRNA expression data) .
The Human Protein Atlas highlights PURG’s role in DNA repair and transcriptional regulation, supported by its interaction with proteins involved in chromatin remodeling . AlphaFold-predicted structures reveal conserved domains critical for nucleic acid binding .
PURG (Purine-Rich Element Binding Protein G) is a DNA and RNA binding protein that belongs to the PUR family of transcription factors. It binds to purine-rich elements in DNA and RNA, playing roles in transcriptional regulation, DNA replication, and potentially in RNA processing and transport. PURG has been implicated in various cellular processes including cell differentiation and development, though its specific functions are still being investigated in many contexts . Research methodologies studying PURG often employ multiple detection techniques including Western blotting and immunofluorescence to correlate protein expression with cellular phenotypes.
PURG antibodies are available in multiple formats optimized for different experimental applications:
Antibody Type | Target Region | Host Species | Applications | Reactivity |
---|---|---|---|---|
Unconjugated polyclonal | C-Terminal (AA 225-253) | Rabbit | WB, FACS | Human |
Unconjugated polyclonal | N-Terminal | Rabbit | WB | Human, Mouse, Cow, Horse, Bat, Monkey |
Unconjugated polyclonal | N-Terminal | Rabbit | WB | Human, Mouse, Dog, Rat |
FITC-conjugated | AA 242-345 | Rabbit | FACS | Human |
Biotin-conjugated | AA 242-345 | Rabbit | ELISA | Human |
HRP-conjugated | AA 242-345 | Rabbit | ELISA | Human |
These antibodies are generated using KLH-conjugated synthetic peptides corresponding to specific amino acid sequences from different regions of the human PURG protein . When selecting an antibody, researchers should consider both the target region and the intended experimental application.
For maximizing PURG antibody shelf-life and maintaining consistent performance, store antibodies at 4°C for short-term use (1-2 weeks). For long-term storage, aliquot the antibody solution into smaller volumes and store at -20°C to avoid repeated freeze-thaw cycles which can damage antibody structure and reduce binding efficacy . Most commercial PURG antibodies are provided in buffer solutions containing PBS (pH 7.2) with 40% glycerol and 0.02% sodium azide as preservatives . When preparing working dilutions, use fresh buffer and handle antibodies on ice to minimize protein degradation.
Proper validation of PURG antibodies is critical for ensuring experimental reliability:
Positive and negative controls: Include cell lines or tissues known to express or lack PURG.
Knockdown/knockout validation: Use siRNA or CRISPR-mediated knockdown/knockout of PURG to confirm antibody specificity.
Peptide competition assay: Pre-incubate antibody with excess immunizing peptide to block specific binding.
Cross-reactivity testing: Test reactivity against closely related family members (e.g., PURA, PURB).
Multiple antibody comparison: Use antibodies targeting different epitopes of PURG to confirm consistent detection patterns.
The most comprehensive validation employs a multi-parameter approach combining at least three of these methods. High-quality commercial PURG antibodies typically undergo specificity verification on protein arrays containing the target protein and numerous non-specific proteins . Documentation of validation procedures should be included in research methods sections.
Optimal working concentrations vary by application technique:
Application | Recommended Dilution | Incubation Conditions | Detection System |
---|---|---|---|
Western Blotting | 1:500-1:1000 | Overnight at 4°C or 2 hours at room temperature | HRP-conjugated secondary antibody with ECL detection |
Immunohistochemistry | 1:500-1:1000 | 1 hour at room temperature or overnight at 4°C | DAB or fluorescent detection systems |
Immunocytochemistry | 1-4 μg/ml | 1-2 hours at room temperature | Fluorescent secondary antibodies |
Flow Cytometry | 1:50-1:200 | 30-60 minutes at 4°C | Direct detection (for conjugated antibodies) or fluorescent secondary antibodies |
ELISA | Assay-dependent | 1-2 hours at room temperature | HRP or biotin detection systems |
Always perform titration experiments to determine optimal concentrations for your specific experimental system. Background signal can be minimized by including appropriate blocking reagents (5% BSA or 5% non-fat milk) and using stringent washing protocols with detergent-containing buffers .
PURG can localize to both nuclear and cytoplasmic compartments, requiring specific sample preparation methods:
For nuclear fraction isolation:
Harvest cells and wash in ice-cold PBS
Resuspend in hypotonic buffer (10 mM HEPES pH 7.9, 10 mM KCl, 0.1 mM EDTA)
Add NP-40 to 0.5% final concentration
Centrifuge to separate cytoplasmic (supernatant) and nuclear (pellet) fractions
Resuspend nuclear pellet in high-salt buffer (20 mM HEPES pH 7.9, 400 mM NaCl, 1 mM EDTA)
Process for Western blotting or immunoprecipitation
For immunofluorescence detection of nuclear PURG, permeabilization with 0.1-0.5% Triton X-100 is recommended, while cytoplasmic detection may require milder detergents like 0.1% saponin. Include protease inhibitor cocktails in all buffers to prevent protein degradation during sample preparation .
False Positive Results:
Cross-reactivity with related PUR family proteins (PURA, PURB)
Non-specific binding to highly abundant proteins
Improper blocking leading to background signal
Secondary antibody cross-reactivity
Excessive primary antibody concentration
False Negative Results:
Epitope masking due to protein-protein interactions
Sample preparation methods that denature the recognized epitope
Insufficient antigen retrieval in fixed tissues
Low PURG expression levels below detection threshold
Degradation of target protein during sample preparation
To minimize false results, employ multiple detection methods and include appropriate positive and negative controls in each experiment. For Western blotting applications, verify the molecular weight of detected bands against the predicted size of PURG (approximately 32-35 kDa) .
For successful immunoprecipitation of PURG protein complexes:
Lysis buffer optimization: Use RIPA buffer (150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM Tris pH 8.0) for strong interactions; milder buffers (1% NP-40, 150 mM NaCl, 50 mM Tris pH 7.5) for preserving weak interactions
Pre-clearing: Pre-clear lysates with protein A/G beads to reduce non-specific binding
Antibody binding: Incubate 1-5 μg antibody per 500 μg protein lysate overnight at 4°C
Capture: Add protein A/G magnetic beads for 1-2 hours
Washing: Perform at least 4 washes with decreasing salt concentrations
Elution: Use gentle elution methods (low pH glycine or competition with immunizing peptide) to maintain complex integrity
For co-immunoprecipitation studies investigating PURG interaction partners, consider crosslinking approaches using formaldehyde or DSP (dithiobis(succinimidyl propionate)) to stabilize transient interactions before cell lysis .
Rigorous quantitative analysis using PURG antibodies requires the following controls:
Loading controls: Include housekeeping proteins (β-actin, GAPDH) for normalization
Calibration curves: Generate standard curves using recombinant PURG protein
Technical replicates: Perform at least three technical replicates per biological sample
Biological replicates: Include at least three independent biological samples
Negative controls: Include samples with undetectable PURG expression
Signal linearity verification: Ensure signal intensity correlates linearly with protein concentration
Antibody titration: Confirm that antibody concentration is not limiting detection
For absolute quantification approaches, researchers should consider using synthetic peptide standards labeled with stable isotopes as internal controls .
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) with PURG antibodies can reveal genome-wide binding patterns:
Antibody selection: Choose antibodies validated specifically for ChIP applications (several PURG antibodies are suitable for immunoprecipitation)
Crosslinking optimization: Test both formaldehyde (1-1.5%, 10-15 minutes) and dual crosslinking approaches (DSG followed by formaldehyde)
Sonication conditions: Optimize to achieve 200-500 bp chromatin fragments
IP conditions: Use 3-5 μg antibody per ChIP reaction with overnight incubation
Washing stringency: Include high-salt and LiCl washes to reduce background
Library preparation: Generate sequencing libraries from immunoprecipitated DNA following standard protocols
For validating ChIP-seq peaks, perform ChIP-qPCR on selected targets comparing enrichment to input and IgG controls. Consider parallel experiments using antibodies targeting different epitopes of PURG to increase confidence in identified binding sites .
Several complementary approaches can identify and characterize PURG interaction partners:
Co-immunoprecipitation with mass spectrometry: Pull down PURG complexes and identify partners by LC-MS/MS
Proximity labeling: Use BioID or APEX2 fused to PURG to identify proximal proteins
Yeast two-hybrid screening: Screen for direct binding partners using PURG as bait
FRET/BRET analysis: Study dynamic interactions in living cells
Protein microarrays: Screen for interactions against arrays of purified proteins
Crosslinking mass spectrometry: Map interaction interfaces at amino acid resolution
To validate interactions, employ reciprocal co-IPs and confirm co-localization by immunofluorescence microscopy. When studying RNA-protein interactions involving PURG, RNA immunoprecipitation (RIP) or CLIP (crosslinking immunoprecipitation) approaches can be employed using validated PURG antibodies .
Post-translational modifications (PTMs) can significantly impact antibody recognition of PURG:
Phosphorylation: May alter epitope accessibility, particularly in regulatory domains
Ubiquitination: Can affect protein stability and complex formation
SUMOylation: May influence nuclear localization and DNA binding
Methylation/Acetylation: Can regulate transcriptional activity
To study modified PURG:
Use modification-specific antibodies (when available)
Employ phosphatase or deubiquitinase treatments as controls
Perform 2D gel electrophoresis to separate modified forms
Use mass spectrometry to map modification sites
Generate phospho-mimetic or phospho-dead mutants for functional studies
When studying PTM effects on PURG function, consider using antibodies targeting regions distant from known modification sites to ensure detection of all protein forms .
Recent advances in computational antibody design can enhance PURG antibody development:
Epitope prediction: Machine learning algorithms can identify optimal antigenic determinants within PURG for targeted antibody generation
Sequence optimization: Generative adversarial networks (GANs) can design antibody variable regions with improved specificity and developability
Structural modeling: AI-driven protein structure prediction tools can model antibody-PURG interactions
Cross-reactivity prediction: Computational approaches can assess potential cross-reactivity with related PUR family proteins
Developability assessment: Deep learning models can predict antibody properties like expression, stability, and non-specific binding
Advanced computational pre-screening can reduce the number of candidates requiring experimental validation, accelerating the development of highly specific PURG antibodies with optimal performance characteristics .
When faced with discrepancies between results obtained with different PURG antibodies:
Epitope mapping: Determine precise binding sites of each antibody
Isoform specificity: Verify which PURG isoforms (PURG-A, PURG-B) each antibody recognizes
Validation stringency: Employ knockout/knockdown controls for each antibody
Orthogonal techniques: Confirm results using antibody-independent methods (e.g., mass spectrometry)
Condition sensitivity: Test whether discrepancies are specific to certain experimental conditions
Post-translational modifications: Determine if modifications affect epitope recognition
A systematic analysis comparing multiple antibodies against defined positive and negative controls can help establish a consensus result. Recording and reporting the specific clone or catalog numbers used is essential for result interpretation and reproducibility .
For multiplexed detection involving PURG:
Antibody panel design: Select compatible primary antibodies from different host species
Conjugated antibodies: Use directly labeled PURG antibodies to avoid secondary antibody cross-reactivity
Sequential staining: Employ multiple rounds of staining with stripping between cycles
Spectral unmixing: Use hyperspectral imaging to separate overlapping fluorescence signals
Cyclic immunofluorescence: Perform repeated cycles of staining and signal inactivation
PURG expression shows tissue-specific patterns relevant to experimental design:
Tissue Type | Relative PURG Expression | Detection Method |
---|---|---|
Brain | High | IHC, RNA-seq |
Heart | Moderate | RT-qPCR, WB |
Skeletal Muscle | Moderate | IHC, RNA-seq |
Kidney | Low to moderate | WB, IHC |
Liver | Low | RT-qPCR, WB |
Lung | Variable | IHC, RNA-seq |
Lymphoid tissues | Low | Flow cytometry |
In pathological conditions, PURG expression may be dysregulated. When investigating disease-associated changes, include appropriate control tissues and consider analysis of multiple isoforms. For quantitative comparisons across tissues, normalization to tissue-specific reference genes rather than common housekeeping genes may provide more accurate results .
When analyzing PURG subcellular localization:
Fixation artifacts: Different fixation methods can alter apparent localization
Co-localization controls: Include markers for specific subcellular compartments
Cell cycle dependence: PURG localization may vary throughout the cell cycle
Physiological state: Consider how stress, differentiation, or activation may affect localization
Resolving power limitations: Standard light microscopy cannot resolve structures below ~200 nm
Quantitative analysis: Use digital image analysis for objective quantification of co-localization
Super-resolution microscopy techniques (STED, STORM, SIM) can provide enhanced resolution of PURG localization patterns. When possible, complement immunofluorescence data with biochemical fractionation approaches to confirm subcellular distribution .
For multi-omics integration of PURG-related data:
Transcriptomics correlation: Compare PURG protein levels with RNA-seq data for PURG and related genes
ChIP-seq integration: Correlate PURG binding sites with transcriptional changes
Proteomics validation: Confirm antibody-based findings with mass spectrometry data
Interactome mapping: Connect PURG interactions with functional networks
Epigenomic correlation: Analyze relationships between PURG binding and chromatin modifications
Pathway analysis: Place PURG within regulatory networks using combined data sources
Computational approaches like weighted gene co-expression network analysis (WGCNA) can help identify functional modules associated with PURG. When integrating antibody-based data with omics approaches, careful normalization and statistical analysis are essential to account for technological biases between platforms .