ING2 antibody is a specialized immunoglobulin designed to detect and quantify the ING2 protein, a member of the inhibitor of growth (ING) family. ING2 is a tumor suppressor involved in chromatin remodeling, cell cycle regulation, and mitochondrial function . Antibodies against ING2 are critical tools for studying its role in cancer, developmental biology, and cellular stress responses.
ING2 antibodies have enabled critical insights into ING2’s biological roles:
Colon Cancer: ING2 upregulation correlates with increased MMP13 expression, enhancing invasion and metastasis .
Tumor Suppression: ING2 interacts with p53 to regulate apoptosis and senescence .
Mitochondrial Respiration: ING2 stabilizes MRPL12, a mitochondrial ribosomal protein, to regulate mtDNA transcription and respiration .
Acute Kidney Injury (AKI): ING2 overexpression mitigates ischemic AKI by preserving tubular epithelial cell (TEC) mitochondrial function .
TGF-β Pathway: ING2 enhances TGF-β-induced transcription by interacting with SnoN and Smad2, promoting cell cycle arrest .
ING2 (Inhibitor of Growth Family, Member 2) is a member of the ING tumor suppressor family that associates with and modulates the activity of histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes. ING2 functions in critical cellular processes including DNA repair, apoptosis, and cell cycle regulation. Research has demonstrated its role in chromatin binding, autophagy, and negative regulation of intrinsic apoptotic signaling . It has gained significance as a research target due to its demonstrated function as a mediator of Transforming Growth Factor-β (TGF-β) signaling, where it promotes TGF-β-induced transcription and cell cycle arrest . These characteristics make ING2 an important subject for cancer research, epigenetic studies, and cellular signaling investigations.
When selecting an ING2 antibody, consider these key technical parameters:
Target species reactivity: Verify that the antibody has been validated for your species of interest. Available ING2 antibodies may react with human, mouse, rat, and sometimes multiple species including horse, rabbit, cow, dog, guinea pig, zebrafish, monkey, and pig .
Clonality: Determine whether polyclonal or monoclonal antibodies better suit your experiment:
Host species: Consider the host species (rabbit, mouse, goat) in relation to your secondary detection system and to avoid cross-reactivity in your experimental system .
Applications: Verify validation for your specific application (Western blotting, ELISA, immunohistochemistry, immunofluorescence, immunoprecipitation) .
Epitope location: Some antibodies target specific regions (N-terminal, C-terminal, or particular amino acid sequences like AA 1-280, AA 112-141, AA 25-74), which may be important depending on your research question .
Always review validation data and literature citations when available to ensure the antibody has demonstrated performance in applications similar to yours.
For optimal Western blotting results with ING2 antibodies:
Sample preparation:
Use RIPA or NP-40 buffer with protease inhibitors for cell/tissue lysis
Include phosphatase inhibitors if studying phosphorylation states of ING2
Heat samples at 95°C for 5 minutes in reducing SDS sample buffer
Gel selection and transfer:
Antibody dilutions and incubation:
Detection considerations:
Expected results:
Wild-type ING2 should appear at approximately 33 kDa
Verify antibody specificity using positive controls and ING2 knockdown samples
To optimize ING2 antibody performance in immunohistochemistry:
Fixation and antigen retrieval:
For FFPE tissues, use citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) for antigen retrieval
Heat-induced epitope retrieval (HIER) at 95-98°C for 15-20 minutes typically yields better results than enzymatic methods
Freshly fixed tissues (24-48 hours) often provide optimal antigen preservation
Antibody concentration and incubation:
Signal amplification and detection:
For low expression levels, consider biotin-streptavidin or tyramide signal amplification
For co-localization studies, use fluorescent secondary antibodies with appropriate controls
Validation strategies:
Compare staining pattern across multiple ING2 antibodies targeting different epitopes
Include positive control tissues known to express ING2
For critical studies, confirm specificity using ING2 knockdown tissues/cells
Expected localization:
To investigate ING2's role in TGF-β signaling:
Reporter gene assays:
Utilize TGF-β-responsive reporters such as the 3TP-luciferase reporter containing Smad binding elements (SBEs)
Co-transfect cells with the reporter construct and ING2 expression plasmids
Treat with TGF-β ligand (typically 100-200 pM) for 16-24 hours before assessing luciferase activity
Include appropriate controls (empty vector, TGF-β receptor inhibitors)
Domain function analysis:
Generate ING2 mutants lacking specific domains:
Assess these mutants in reporter assays to determine domain-specific contributions to TGF-β signaling
Protein-protein interaction studies:
Gene expression analysis:
Monitor TGF-β target genes (e.g., PAI-1) by qRT-PCR or Western blot
Compare responses in cells with ING2 overexpression, knockdown, or mutant expression
Include time-course analysis (2-48 hours) to capture early and late TGF-β responses
Cell proliferation assays:
To investigate ING2's chromatin remodeling functions:
Chromatin immunoprecipitation (ChIP):
Perform ChIP with ING2 antibodies to identify genomic binding sites
Couple with sequencing (ChIP-seq) for genome-wide binding profiles
Look for co-occupancy with histone marks (H3K4me3, H3K27ac) and TGF-β-regulated transcription factors
Compare binding patterns before and after TGF-β stimulation
Histone modification analysis:
Examine how ING2 overexpression or knockdown affects:
H3K4 methylation
Histone acetylation at TGF-β target genes
Use Western blotting with histone modification-specific antibodies
Perform ChIP for histone marks at specific promoters
Chromatin accessibility assays:
Use ATAC-seq or DNase-seq to determine how ING2 affects chromatin accessibility
Focus on TGF-β responsive genomic regions
Compare wild-type ING2 versus PHD domain mutants
Protein complex identification:
Perform tandem affinity purification of ING2 followed by mass spectrometry
Identify components of ING2-containing HAT or HDAC complexes
Verify interactions with co-immunoprecipitation
Determine how TGF-β signaling affects complex composition
Live-cell imaging:
Use fluorescently tagged ING2 to track dynamics at chromatin
Perform fluorescence recovery after photobleaching (FRAP) to measure residence time on chromatin
Compare dynamics before and after TGF-β stimulation
To address specificity concerns:
Validation controls:
Genetic approaches: Use ING2 knockout/knockdown cells or tissues as negative controls
Peptide competition: Pre-incubate antibody with the immunizing peptide before application
Multiple antibodies: Use antibodies targeting different ING2 epitopes and compare staining patterns
Heterologous expression: Test antibody on cells overexpressing tagged ING2
Cross-reactivity assessment:
Test for cross-reactivity with other ING family members (ING1, ING3-5)
For polyclonal antibodies, consider affinity purification against recombinant ING2
When detecting endogenous ING2, include positive control samples with known expression
Technical approaches to improve specificity:
Optimize antibody concentration (use titration experiments)
Increase stringency of washing steps (higher salt concentration, longer washes)
Use more selective blocking agents (specific for your application)
Consider monoclonal antibodies for higher specificity in challenging applications
Application-specific considerations:
Common challenges and solutions:
Transient or weak interactions:
Use chemical crosslinking (formaldehyde, DSS, or BS3) before lysis
Try proximity ligation assay (PLA) for detecting in situ protein interactions
Consider bimolecular fluorescence complementation (BiFC) for live cell analysis
Buffer composition challenges:
Test multiple lysis buffers (RIPA vs. NP-40 vs. digitonin-based)
Adjust salt concentration to optimize interaction stability
Include phosphatase inhibitors when studying phosphorylation-dependent interactions
Consider the presence/absence of detergents that might disrupt certain interactions
Nuclear protein extraction issues:
Use specialized nuclear extraction protocols for efficient ING2 isolation
Consider DNase/RNase treatment to release chromatin-bound proteins
For histone-associated interactions, include histone deacetylase inhibitors
Post-translational modifications:
For studying interactions with SnoN and Smad2, consider how TGF-β treatment affects:
Phosphorylation states
Protein stability
Nuclear-cytoplasmic distribution
Time course experiments may be necessary to capture dynamic interactions
Detection sensitivity:
For endogenous interactions, optimize antibody combinations that don't cross-react
Consider using more sensitive detection methods (e.g., proximity-based assays)
For mass spectrometry, use SILAC or TMT labeling to distinguish specific from non-specific interactions
When faced with conflicting results:
Epitope considerations:
Determine the epitopes recognized by each antibody (N-terminal, C-terminal, internal)
Conflicting results may reflect detection of different:
ING2 isoforms
Post-translational modifications
Protein-protein interaction states that mask certain epitopes
Methodological approach:
Compare antibody performance across multiple techniques:
If conflict exists only in one technique (e.g., IHC), it may reflect fixation/processing sensitivity
If conflict exists across techniques, consider fundamental specificity issues
Test antibodies on recombinant ING2 constructs with known modifications/truncations
Validation strategies:
Use genetic models (CRISPR knockout, siRNA) to confirm specificity
Perform rescue experiments with ING2 reexpression
Consider advanced approaches like mass spectrometry to verify antibody targets
Interpretation framework:
Create a decision tree based on:
Antibody validation documentation
Published literature using each antibody
Technical controls performed in your experiments
Weight evidence based on rigor of controls and consistency across techniques
Reporting guidelines:
Document all antibodies used (catalog numbers, dilutions, lots)
Clearly describe conflicting results in publications
Consider showing results from multiple antibodies when conflicts exist
Discuss potential biological explanations for differences
To differentiate direct from indirect effects:
Temporal analysis:
Perform detailed time-course experiments:
Immediate early responses (0-2 hours) more likely reflect direct effects
Delayed responses (>4 hours) may indicate secondary effects
Use protein synthesis inhibitors (cycloheximide) to block secondary responses requiring new protein synthesis
Domain mutation approaches:
Chromatin occupancy analysis:
Perform ChIP-seq for ING2 and Smad proteins
Sites co-occupied by both factors more likely represent direct regulatory targets
Integrate with RNA-seq after short TGF-β treatment to identify immediate transcriptional effects
Biochemical interaction assays:
Use in vitro binding assays with purified components to confirm direct interactions
Reconstitute minimal systems to determine sufficiency for specific effects
Consider in vitro transcription systems to test direct transcriptional regulation
Genetic epistasis experiments:
Perform double knockdown/knockout experiments:
ING2 + Smad2/3
ING2 + SnoN
Compare phenotypes to determine pathway relationships
Use rescue experiments with mutant proteins resistant to siRNA targeting
Table 1: Experimental Approaches to Distinguish Direct vs. Indirect ING2 Effects
Approach | Direct Effect Evidence | Indirect Effect Evidence | Technical Considerations |
---|---|---|---|
Temporal | Rapid response (0-2h) | Delayed response (>4h) | Include protein synthesis inhibitors |
Domain Analysis | Dependent on specific domains (e.g., PHD) | Independent of characterized domains | Use multiple domain mutants |
Genomic Occupancy | Co-occupancy with Smads | Occupancy at secondary response genes | Integrate with gene expression data |
Biochemical | In vitro reconstitution sufficient | Requires additional factors | Control for post-translational modifications |
Genetic | Direct epistatic relationship | Complex genetic interactions | Account for compensatory mechanisms |
Appropriate statistical approaches depend on the experimental design:
Comparative expression analysis:
For comparing ING2 levels across conditions:
Use t-tests for two-group comparisons (if normally distributed)
Use ANOVA followed by post-hoc tests for multiple group comparisons
Consider non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) for non-normal distributions
Calculate appropriate effect sizes (Cohen's d, fold change)
Report both raw p-values and adjusted p-values for multiple comparisons
Correlation analysis:
For examining relationships between ING2 and other variables:
Use Pearson correlation for linear relationships (if normally distributed)
Use Spearman correlation for non-parametric or non-linear relationships
Consider partial correlations to control for confounding variables
Report both correlation coefficients and confidence intervals
Functional assays:
For reporter gene assays:
For proliferation assays:
Consider area-under-curve (AUC) analysis for growth curves
Calculate EC50 values to compare sensitivities to TGF-β
Omics data integration:
For ChIP-seq or RNA-seq:
Apply multiple testing correction (FDR, Bonferroni)
Use enrichment analyses for pathway/GO term identification
Consider integrated approaches (GSEA, network analysis)
Report both statistical significance and biological significance (fold change)
Power and sample size:
Conduct a priori power analysis to determine appropriate sample sizes
Report effect sizes alongside p-values
Consider biological replicates (independent experiments) vs. technical replicates
To investigate this dual role:
Cell context-dependent studies:
Molecular mechanism dissection:
Analyze ING2 interactions with specific Smad complexes:
Growth-inhibitory complexes (Smad3/4 with p15, p21 promoters)
EMT-promoting complexes (Smad3/4 with SNAI1, ZEB1 promoters)
Determine if ING2 differentially regulates these distinct transcriptional programs
In vivo models:
Generate tissue-specific ING2 knockout or overexpression mouse models
Study effects on:
Primary tumor growth
Metastatic potential
TGF-β responsiveness in different tumor stages
Use genetic crosses with established TGF-β pathway mutant models
Epigenetic landscape analysis:
Perform integrated epigenomic profiling:
ChIP-seq for ING2, Smads, and histone modifications
ATAC-seq for chromatin accessibility
DNA methylation analysis
Compare patterns between normal and transformed states
Identify switches in ING2 genomic targeting during malignant progression
Therapeutic implications:
Test ING2 modulation in combination with TGF-β pathway inhibitors
Assess stage-specific responses to determine optimal intervention points
Evaluate biomarkers that predict context-dependent functions
Emerging technologies with potential applications:
Proximity-based protein interaction methods:
BioID or TurboID fusion proteins to identify ING2 proximity partners
APEX2-based proximity labeling for subcellular compartment-specific interactors
Split-BioID to capture condition-specific interactions (e.g., only upon TGF-β stimulation)
Advanced microscopy approaches:
Super-resolution microscopy (STORM, PALM, SIM) to visualize ING2 nuclear distribution
Lattice light-sheet microscopy for dynamic tracking of ING2 in living cells
Single-molecule tracking to measure ING2-chromatin binding kinetics
CRISPR-based technologies:
CUT&RUN or CUT&Tag as antibody-based alternatives to traditional ChIP
CRISPR activation/inhibition of ING2 for functional genomics
CRISPR base editing to introduce specific mutations in endogenous ING2
Antibody engineering approaches:
Nanobodies against ING2 for improved penetration and reduced background
Intrabodies for tracking and manipulating ING2 in living cells
Bispecific antibodies to detect ING2 in complex with specific partners
Mass cytometry and spatial proteomics:
CyTOF with ING2 antibodies for single-cell protein quantification
Imaging mass cytometry or CODEX for spatial context of ING2 expression
Hyperplexed immunofluorescence to map ING2 in relation to multiple markers
Table 2: Comparative Analysis of Emerging Technologies for ING2 Research
Technology | Key Advantage | Technical Challenge | Best Application Scenario |
---|---|---|---|
Proximity Labeling | Captures transient/weak interactions | Requires genetic engineering | Identifying complete ING2 interactome |
Super-Resolution Microscopy | 10-20nm resolution of nuclear structures | Specialized equipment needed | Visualizing ING2 in chromatin domains |
CUT&RUN/CUT&Tag | Lower background than ChIP | Optimization for each antibody | Mapping ING2 genomic binding sites |
Nanobodies | Smaller size, better penetration | Limited commercial availability | Live-cell tracking of ING2 |
Spatial Proteomics | Preserves tissue architecture | Complex data analysis | Analyzing ING2 in tumor microenvironment |
The Inhibitor of Growth (ING) family consists of tumor suppressor genes that play crucial roles in regulating cell proliferation, apoptosis, and cellular senescence. The family includes five members: ING1, ING2, ING3, ING4, and ING5. These genes are highly conserved across species, including humans and mice .
The ING2 gene encodes a protein that is involved in various cellular processes, including DNA repair, apoptosis, and chromatin remodeling. The ING2 protein is known to interact with histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes, which are essential for regulating gene expression through epigenetic mechanisms .
ING2, like other members of the ING family, functions as a tumor suppressor. It is involved in the activation of the p53 pathway, which is crucial for inducing apoptosis in response to DNA damage. The loss or mutation of ING2 has been associated with various types of cancer, highlighting its importance in maintaining cellular homeostasis and preventing malignant transformation .
The mouse anti-human ING2 antibody is a monoclonal antibody used in research to study the expression and function of the ING2 protein in human cells. This antibody is valuable for various applications, including Western blotting, immunohistochemistry, and immunoprecipitation. By targeting the ING2 protein, researchers can gain insights into its role in different cellular processes and its involvement in cancer development .