ING5 phosphorylation at Thr152 by CDK2/cyclin complexes regulates its activity during S-phase progression. Knockdown experiments demonstrate:
63% reduction in HCT116 colorectal cancer cell proliferation
41% increase in apoptosis (annexin V+ cells) within 48 hours
| Cancer Type | Proliferation | Apoptosis | Metastasis | Key Pathways |
|---|---|---|---|---|
| Gastric | ↓ 58% | ↓ 37% | ↓ 72% | β-catenin, NF-κB, PI3K/Akt |
| Glioblastoma | ↑ 29%* | - | - | Calcium signaling, FSH receptor |
| Lung | ↓ 41% | ↑ 53% | ↓ 68% | EMT inhibition |
*Contrasting pro-proliferative role in glioblastoma stem cells
While ING5 suppresses gastric cancer growth (tumor size reduction: 64% in xenografts) , it promotes:
Chemoresistance through overexpression of:
Stem cell maintenance in glioblastoma via:
Clinical correlations reveal:
Inverse association with gastric cancer stage (r = -0.67, p<0.01)
Prognostic value in lung adenocarcinoma (HR = 1.89, 95% CI 1.2-2.9)
83% cytoplasmic mislocalization in colorectal tumors vs. normal tissue
The antibody's ability to detect both nuclear and cytoplasmic ING5 makes it valuable for studying subcellular localization changes during malignant transformation. Recent work highlights its utility in identifying ING5-containing chromatin complexes (HBO1, MOZ/MORF) through IP applications .
ING5 (Inhibitor of Growth family, member 5) is a 28-32 kDa nuclear protein characterized by a PHD-type zinc finger domain that plays crucial roles in chromatin remodeling and transcriptional regulation. It belongs to the ING family of tumor suppressors and functions as a component of the HBO1 complex with histone H4-specific acetyltransferase activity .
ING5 has emerged as an important research target because:
It regulates cell proliferation, apoptosis, and differentiation
It demonstrates tumor suppressive functions in multiple cancer types
Its decreased nuclear expression correlates with cancer progression
It inhibits epithelial-to-mesenchymal transition (EMT) in lung cancer
ING5 knockout mice show increased susceptibility to developing diffuse large B-cell lymphomas
ING5 antibodies have been validated for multiple applications based on the search results:
Research applications include studying ING5's role in tumor suppression, chromatin remodeling, cell cycle regulation, and protein-protein interactions .
For optimal ING5 detection in tissue samples by IHC:
Antigen retrieval: Use TE buffer at pH 9.0 (primary recommendation) or citrate buffer at pH 6.0 (alternative)
Recommended dilution: 1:20-1:200, with optimization suggested for each testing system
Positive control tissues: Human colon cancer tissue and human ovary tumor tissue have been validated
Detection system: Standard secondary antibody approach with appropriate chromogenic detection
Counterstaining: Hematoxylin for nuclear visualization
Researchers should note that ING5 shows both nuclear and cytoplasmic localization, with the nuclear-to-cytoplasmic ratio having potential clinical significance in cancer progression .
Proper controls are essential for ING5 antibody experiments:
Positive controls:
Cell lines with confirmed ING5 expression (validated in Jurkat, PC-3, HEK-293 cells)
Tissues with known ING5 expression (e.g., mouse kidney tissue)
Negative controls:
Primary antibody omission controls
Isotype controls using matched IgG species and subclass
Validation controls:
Protein overexpression systems for specificity testing
Peptide blocking experiments
Multiple antibodies targeting different epitopes
The literature shows seemingly contradictory findings regarding ING5's effects on cell proliferation, with some studies reporting inhibitory effects while others show proliferation promotion . To resolve these discrepancies:
Cell type considerations: ING5 functions in a cell type-specific manner. In lung cancer cells like A549, ING5 overexpression suppresses proliferation , while in other contexts, ING5 knockdown has inhibited cell proliferation .
p53 status evaluation: While ING5 was initially thought to function exclusively through p53, research has demonstrated both p53-dependent and p53-independent functions. Experiments in A549 (p53 wild-type) and H1299 (p53-null) cells showed that ING5's anti-proliferative effects occurred regardless of p53 status .
Subcellular localization analysis: The nuclear-to-cytoplasmic ratio of ING5 affects its function. Nuclear ING5 often correlates with tumor suppression, while cytoplasmic localization may have different effects . Use fractionation approaches followed by Western blotting to assess localization patterns.
Pathway activation status: Determine whether PI3K/Akt or β-catenin/TCF-4 pathways are activated, as ING5 can influence these pathways, affecting proliferation outcomes .
Experimental design recommendations:
Include both overexpression and knockdown approaches
Conduct time-course experiments to capture temporal effects
Assess multiple proliferation parameters (cell counting, EdU incorporation, colony formation)
Consider three-dimensional culture systems
ING5 functions through protein-protein interactions with chromatin modifiers and transcription factors. The following methodologies are optimal for studying these interactions:
Co-immunoprecipitation (Co-IP):
Chromatin Immunoprecipitation (ChIP):
Electrophoretic Mobility Shift Assay (EMSA):
Prepare biotin-labeled probes containing binding sites of interest
Include appropriate controls (label-free cold competition, mutant probes)
Purify recombinant proteins using His-tagged expression systems
Use 5-10 μg protein in DNA-protein binding reactions
Proximity Ligation Assay (PLA):
Particularly useful for studying in situ protein interactions
Can detect endogenous protein complexes without overexpression
Provides spatial information about interaction locations
Mass Spectrometry-Based Approaches:
The research by Tctp has identified Ing5's interaction with the PHD domain using GST-tagged Ing5 protein fragments, providing a model for domain-specific interaction studies .
CRISPR/Cas9 knockout models provide the gold standard for antibody validation. The following protocol outlines how to validate ING5 antibodies using these models:
Generation of knockout models:
Validation protocol:
Process wild-type and knockout samples identically
Run Western blot using the antibody being validated
True specific antibodies will show bands at expected molecular weight (28-32 kDa) in wild-type samples and no band in knockout samples
Include positive control protein detection on the same membrane
Tissue-specific knockouts for in vivo validation:
Validate ING5 antibodies in tissue-specific knockout models
As demonstrated in recent research, mating ING5 mutant mice with tissue-specific Cre recombinase-expressing mice (e.g., Atp4b-cre, Capn8-cre, PGC-cre, K19-cre, and Pdx1-cre) creates models for antibody validation
Process tissues from wild-type and conditional knockout mice for IHC validation
Additional validation approaches:
Confirm using multiple antibodies recognizing different epitopes
Include overexpression controls alongside knockout controls
Perform peptide competition assays
As reported in recent literature, the KO validation approach has been successfully used for ING5 antibody validation, confirming specificity for the 28 kDa ING5 protein .
ING5 functions as an epigenetic reader that interacts with histone marks and influences chromatin structure. To study these functions:
ChIP-seq analysis:
Use validated ING5 antibodies for chromatin immunoprecipitation
Sequence immunoprecipitated DNA to identify ING5 binding sites
Analyze how ING5 binding correlates with gene expression
Recent studies demonstrate >50% overlap between ING5 and H3K4me3 peaks, indicating ING5 binds primarily at transcription start sites of active genes
Co-immunoprecipitation of histone modifiers:
Investigate ING5 interactions with histone acetyltransferases (HATs) like HBO1
Analyze complex formation using size-exclusion chromatography
Compare ING5 binding to different histone marks using modified histone peptide arrays
Histone modification analysis:
Use antibodies against specific histone modifications (H3K4me3, acetylated H3 and H4)
Perform Western blotting of acid-extracted histones
Compare modification patterns between wild-type and ING5-depleted cells
Consider ChIP-seq for genome-wide analysis of histone modification changes
Gene expression correlation:
PHD domain functional studies:
Studies have reported that the subcellular localization of ING5 has important clinical implications. The following guidelines help interpret such findings:
To investigate ING5's function in epithelial-to-mesenchymal transition (EMT) and metastasis:
In vitro assessment of EMT markers:
Establish stable ING5 overexpression and knockdown cell lines in appropriate cancer models
Analyze EMT markers (E-cadherin, N-cadherin, Snail, Slug) by qRT-PCR and Western blot
Examine morphological changes using phase-contrast microscopy
Perform migration and invasion assays (wound healing, transwell)
Mechanistic studies:
In vivo metastasis models:
Use both subcutaneous and intravenous mouse xenograft models
For lung metastasis studies, inject cells into tail veins and analyze lung nodules
Published studies showed that ING5 overexpression reduced lung metastasis in mouse models
Quantify both the incidence (number of mice with metastasis) and burden (number of metastatic nodules)
Clinical correlation:
Rescue experiments:
Co-express ING5 with EMT-inducing factors to test for functional antagonism
Use EMT-inducing treatments (TGF-β, hypoxia) to determine if ING5 can prevent induced EMT
ING5 plays roles in both normal stem cell maintenance and cancer stem cell properties. To differentiate these functions:
Stem cell model systems:
Use both normal stem cells (e.g., epidermal stem cells) and cancer stem cell populations
Isolate cancer stem cells using established markers (CD133, CD44, ALDH activity)
Compare the effects of ING5 manipulation in matched normal and cancer stem cell populations
Functional assays:
Sphere formation assays (tumorspheres for cancer stem cells, neurospheres for neural stem cells)
Serial dilution transplantation assays for in vivo stem cell potential
Lineage tracing experiments in ING5 conditional knockout mice
Self-renewal vs. differentiation analysis
Molecular characterization:
Expression analysis of stemness markers (OCT4, SOX2, NANOG)
Chromatin landscape analysis (ATAC-seq, ChIP-seq for histone modifications)
Comparison of ING5-bound genomic regions in normal vs. cancer stem cells
In vivo stem cell analysis:
Translational relevance:
Correlation of ING5 expression with stemness markers in patient samples
Analysis of therapy resistance related to cancer stem cell properties
Targeting approaches that specifically affect ING5 function in cancer stem cells but not normal stem cells
Researchers often encounter variability in staining results. To address this:
Antibody selection considerations:
Sample preparation factors:
Protocol optimization:
Biological variables:
Post-translational modifications affecting epitope recognition
Protein complex formation masking antibody binding sites
Subcellular localization changes affecting accessibility
Isoform expression differences
Validation approaches:
Use multiple antibodies recognizing different epitopes
Include positive and negative control samples in each experiment
Consider phosphatase treatment if phosphorylation affects binding
Use recombinant ING5 protein as a standardization control
Tissue microarray (TMA) analysis requires careful methodology:
TMA design principles:
Standardized staining protocol:
Analysis considerations:
Use digital pathology for quantitative assessment when possible
Employ multiple independent scorers to ensure reproducibility
Consider the nuclear-to-cytoplasmic ratio as well as absolute expression levels
Correlate with clinicopathological parameters using appropriate statistical tests
Interpretation guidelines:
Validation approaches:
Confirm key findings with full tissue sections
Validate at the protein level with other methods (Western blot)
Consider correlation with mRNA expression data when available
Researchers sometimes observe discrepancies between antibody-based detection and genetic manipulation results. To reconcile these:
Antibody validation assessment:
Confirm antibody specificity using knockout controls
Test for cross-reactivity with other ING family members
Evaluate if the antibody recognizes all relevant ING5 isoforms
Consider epitope masking in specific contexts
Knockdown efficiency evaluation:
Quantify knockdown at both mRNA and protein levels
Use multiple siRNA/shRNA sequences to control for off-target effects
Consider compensatory upregulation of other ING family members
Research shows two different shRNA constructs (shING5-1 and shING5-2) with different knockdown efficiencies can produce consistent biological effects
Time-course considerations:
Acute vs. chronic knockdown may yield different results
Transient transfection vs. stable cell lines may affect outcomes
Consider inducible systems for temporal control
Functional redundancy analysis:
Test for compensatory mechanisms involving other ING family members
Consider combination knockdown approaches
Analyze expression changes in related pathway components
Experimental design recommendations:
Use multiple methodological approaches (antibody detection, genetic manipulation, functional assays)
Include appropriate controls for each method
Consider cell type-specific effects
Report conflicting results transparently and discuss potential explanations
Post-translational modifications (PTMs) regulate ING5 function:
Phosphorylation analysis:
ING5 is phosphorylated by CDK2 and controls cell proliferation and G2/M arrest
Use phospho-specific antibodies for detection of known sites
Perform IP followed by Western blot with anti-phospho antibodies
Consider phosphatase treatment as a control
Use mass spectrometry for unbiased phosphorylation site mapping
Ubiquitination studies:
Perform immunoprecipitation under denaturing conditions
Blot with anti-ubiquitin antibodies
Use proteasome inhibitors (MG132) to stabilize ubiquitinated forms
Consider targeted mass spectrometry approaches for ubiquitination site identification
Acetylation detection:
Given ING5's role in histone acetylation complexes, acetylation may regulate its function
Use anti-acetyl-lysine antibodies following IP
Treat cells with HDAC inhibitors (e.g., SAHA) to enhance acetylation
Perform site-directed mutagenesis of putative acetylation sites
SUMOylation analysis:
IP followed by Western blot with anti-SUMO antibodies
SUMO-site prediction followed by targeted mutation
Use SUMO-specific proteases as controls
Functional impact studies:
Generate phosphomimetic and phospho-deficient mutants
Analyze subcellular localization changes in response to PTMs
Study protein-protein interaction changes using Co-IP
Assess chromatin binding differences using ChIP
For complex co-localization studies:
Antibody compatibility assessment:
Test primary antibody host species to avoid cross-reactivity
Validate each antibody individually before multiplexing
Consider using directly conjugated primary antibodies when possible
Ensure secondary antibodies have minimal cross-reactivity
Multiplexing strategies:
Sequential staining with careful stripping between rounds
Tyramide signal amplification for weak signals
Spectral unmixing for overlapping fluorophores
Consider specialized multiplexed platforms (Vectra, Codex)
Optimal marker combinations:
Co-stain ING5 with EMT markers (E-cadherin, Vimentin) for transition studies
Combine with proliferation markers (Ki-67) for cell cycle studies
Co-localize with chromatin marks (H3K4me3) for epigenetic studies
Include stem cell markers (CD44, CD133) for stemness correlation
Controls for multiplexed staining:
Single-color controls for spectral unmixing
Isotype controls for each primary antibody species
Fluorescence-minus-one (FMO) controls
Biological controls with known expression patterns
Analysis considerations:
Use specialized software for co-localization quantification
Consider 3D confocal imaging for volumetric co-localization
Perform correlation analysis between markers
Use machine learning approaches for complex pattern recognition
To investigate ING5's roles in cell cycle regulation and apoptosis:
Cell cycle analysis approaches:
Synchronize cells using serum starvation or chemical inhibitors
Perform flow cytometry with PI staining to assess cell cycle distribution
Research shows ING5 overexpression induces G2/M arrest in glioma cells
Co-stain for ING5 and cell cycle markers (cyclins, CDKs) by immunofluorescence
Use EdU incorporation assays for S-phase analysis
Apoptosis detection methods:
Molecular pathway analysis:
In vivo approaches:
Technical considerations:
Include appropriate positive controls for apoptosis (e.g., staurosporine treatment)
Consider time-course experiments to capture transient effects
Use flow cytometry for quantitative analysis of large cell populations
Complement protein-level studies with mRNA expression analysis
To investigate ING5's tumor suppressor functions:
Cross-cancer expression analysis:
Use tissue microarrays containing multiple cancer types
Standardize IHC protocols for consistent cross-study comparison
Focus on nuclear vs. cytoplasmic localization patterns
Studies show ING5 downregulation or cytoplasmic translocation in various cancers including gastric, lung, and head and neck squamous cell carcinoma
Correlation with clinical parameters:
Functional validation approaches:
Mechanism investigation across cancer types:
Study EMT inhibition in epithelial cancers
Analyze cell cycle regulation and apoptosis
Examine chromatin remodeling effects
Compare p53-dependent vs. p53-independent mechanisms
Therapeutic implication studies:
Test combination with chemotherapeutic agents
Analyze synthetic lethality relationships
Explore epigenetic drug interactions
Consider ING5 as a biomarker for treatment response
To investigate ING5's relationship with other tumor suppressors:
Protein-protein interaction studies:
Co-immunoprecipitation of ING5 with p53 and other tumor suppressors
Proximity ligation assay for in situ interaction detection
Use deletion mutants to map interaction domains
Consider split reporter systems (BiFC, FRET) for dynamic interaction studies
Functional cooperation analysis:
Transcriptional regulation studies:
ChIP-seq to identify co-occupied genomic regions
Luciferase reporter assays with p53-responsive promoters
Gene expression analysis after manipulation of ING5, p53, or both
qRT-PCR validation of key target genes
Post-translational modification interactions:
Analyze how modifications of ING5 affect p53 binding
Study if ING5 alters p53 stability or modifications
Consider how stress conditions affect their interaction
In vivo tumor model approaches:
Create compound mouse models (ING5/p53 double knockouts)
Analyze tumor onset, progression, and histology
Study therapeutic responses in different genetic backgrounds
For investigating ING5's epigenetic functions:
Chromatin binding studies:
Histone modification analysis:
Western blot of acid-extracted histones
ChIP-seq for histone marks in ING5 manipulated cells
Focus on H3/H4 acetylation changes
Mass spectrometry-based histone modification analysis
Chromatin remodeling complex studies:
Co-IP to detect ING5 interactions with HBO1 complex components
Size-exclusion chromatography to analyze complex integrity
Density gradient fractionation of nuclear extracts
Analyze effects of ING5 knockdown on complex assembly
Functional genomics approaches:
ATAC-seq to assess chromatin accessibility changes
RNA-seq to correlate with gene expression changes
CUT&RUN for higher resolution chromatin binding
Integrate multi-omics data using computational approaches
Domain function analysis:
For translational research applications:
Sample collection and processing:
Standardize fixation protocols (typically 10% neutral buffered formalin for 24h)
Control pre-analytical variables (ischemia time, fixation duration)
Use tissue microarrays for high-throughput screening
Consider matched normal-tumor pairs when possible
Antibody validation for clinical use:
Perform extensive validation using multiple approaches
Compare multiple antibodies targeting different epitopes
Confirm specificity using knockout controls
Validate in the specific tissue types being studied
Scoring and interpretation systems:
Develop standardized scoring systems for nuclear and cytoplasmic staining
Consider automated digital pathology for quantification
Train multiple pathologists and assess inter-observer agreement
Research shows that nuclear ING5 negatively correlates with clinical stage and lymph node metastasis, while high nuclear ING5 associates with better prognosis
Statistical analysis approaches:
Determine appropriate sample sizes for adequate statistical power
Use multivariate analysis to account for confounding factors
Consider survival analysis methods (Kaplan-Meier, Cox regression)
Test in independent validation cohorts
Integration with other biomarkers:
Analyze ING5 in combination with established markers
Consider multiplex IHC approaches
Develop prediction models incorporating multiple variables
Assess added value beyond standard clinicopathological parameters
By following these methodological guidelines, researchers can more effectively utilize ING5 antibodies in their investigations of this important tumor suppressor protein and potentially develop new diagnostic or therapeutic approaches for cancer patients.