ING2, or Inhibitor of Growth Family Member 2, is a protein that plays a crucial role in various cellular processes, including chromatin remodeling, gene regulation, and cellular growth control. It is part of the ING family of proteins, which are known for their involvement in tumor suppression and cellular aging. ING2 is particularly significant due to its association with cancer, tissue repair, and mitochondrial function.
ING2 contains a PHD (plant homeodomain) finger and a nuclear localization signal (NLS), which are common features among the ING family proteins. The PHD domain is essential for binding to specific histone modifications, such as H3K4me3, which are associated with active chromatin regions . This interaction suggests that ING2 is involved in chromatin remodeling and gene regulation.
PHD Domain: Essential for binding to histone modifications.
NLS: Directs the protein to the nucleus.
C-terminal: Plays a crucial role in the specific regulation of target genes like MMP13 and PAI-1 .
ING2 is known to regulate the expression of several genes, including MMP13 (matrix metalloproteinase 13) and PAI-1 (plasminogen activator inhibitor-1). These genes are involved in processes such as tissue remodeling and fibrinolysis. The regulation of MMP13 and PAI-1 by ING2 is specific to ING1b and ING2 among the ING family proteins .
Gene | Function | Regulation by ING2 |
---|---|---|
MMP13 | Tissue remodeling, collagen degradation | Upregulated |
PAI-1 | Inhibits fibrinolysis, involved in tissue repair | Upregulated |
HSPA1A | Heat shock protein, stress response | Upregulated |
MRPL12 | Mitochondrial ribosomal protein, involved in mtDNA transcription | Regulated through ubiquitination |
Recent studies have shown that ING2 plays a role in modulating mitochondrial respiration. It does so by regulating the ubiquitination of MRPL12, a mitochondrial transcription factor, thereby affecting mtDNA transcription and mitochondrial function . This role highlights the diverse cellular processes influenced by ING2.
ING2 has been implicated in carcinogenesis, particularly in breast and gynecological cancers . It is also involved in tissue repair and regeneration. The protein's role in cancer can vary depending on the type of cancer, sometimes acting as a tumor suppressor .
Cancer Type | Role of ING2 |
---|---|
Breast Cancer | Associated with carcinogenesis |
Gynecological Cancer | Involved in carcinogenesis |
Soft-Tissue Sarcomas | Acts as a tumor suppressor |
ING2 is crucial for spermatogenesis. Mice deficient in ING2 exhibit abnormal spermatogenesis and infertility, highlighting its role in reproductive health .
ING2 (Inhibitor of Growth Family Member 2) is a plant homeodomain (PHD)-containing protein that functions as a putative tumor suppressor. It plays pivotal roles in regulating cellular senescence, apoptosis, DNA damage repair, gene transcription, and chromatin modification. ING2 serves as a subunit of the mSin3A-HDAC1 (histone deacetylase 1) complex and specifically binds to tri-methylated lysine 4 of histone H3 (H3K4me3) via its PHD finger domain .
Methodologically, ING2 functions can be investigated through:
Chromatin immunoprecipitation (ChIP) to identify genomic binding sites
Co-immunoprecipitation to detect protein-protein interactions (particularly with p53 and HDAC1)
Gene expression analysis following ING2 modulation using quantitative PCR or RNA-sequencing
Cell cycle analysis using flow cytometry in ING2-deficient or overexpressing cells
ING2 functionally interplays with the p53 tumor suppressor protein in two distinct manners:
Endogenous ING2 inhibits senescence, and p53-mediated transcriptional repression of ING2 abrogates this inhibition
Overexpressed ING2 enhances p53 acetylation and stability to induce senescence
In experimental settings, researchers can study this interaction by:
Using reporter gene assays with p53-responsive elements
Employing site-directed mutagenesis to determine critical interaction domains
Performing acetylation assays to measure p53 post-translational modifications
Conducting cellular senescence assays using β-galactosidase staining
ING2 shows tissue-specific expression patterns with particularly abundant expression in human testes. Decreased ING2 expression has been associated with male infertility and defective spermatogenesis in humans . Public datasets reveal that ING2 expression is significantly reduced (>7-fold) in testes from patients with Sertoli-cell only Syndrome and in teratozoospermic sperm (>3-fold reduction) .
Methodological approaches to study expression include:
RNA-seq analysis of different human tissues
Immunohistochemistry to visualize protein localization
Western blotting for protein quantification
Single-cell RNA sequencing to identify cell-type specific expression
ING2 functions as a novel mediator of transforming growth factor-β (TGF-β) dependent transcription and cell cycle arrest. Research findings demonstrate that:
ING2 enhances TGF-β-induced transcription in multiple cell lines, including augmenting the expression of reporter genes driven by TGF-β-responsive promoters such as PAI-1
The PHD domain of ING2 is required for its enhancement of TGF-β-dependent transcription
ING2 promotes the transcriptional activity of TGF-β-regulated Smad2 and Smad3
ING2 forms a novel interaction with the transcriptional modulator SnoN, which recruits ING2 to Smad2 to induce TGF-β-dependent transcription
ING2 Construct | Effect on TGF-β Signaling | Mechanism |
---|---|---|
Wild-type ING2 | Enhanced transcription | Promotes Smad2/3 activity |
ING2 (ΔN) | Partial enhancement | N-terminal deletion affects function |
ING2 (ΔC) | Significantly reduced | Loss of PHD domain impairs activity |
ING2 (ΔPHD) | Significantly reduced | Cannot bind to H3K4me3 |
Methodologically, researchers investigating ING2's role in TGF-β signaling should:
Utilize reporter gene assays with TGF-β-responsive elements
Perform co-immunoprecipitation to detect interactions with Smad proteins
Examine endogenous target gene expression through RT-qPCR
Use structure-function analyses with deletion mutants to identify critical domains
ING2 regulates spermatogenesis through both p53-dependent and chromatin-mediated mechanisms. Studies in Ing2-deficient mice revealed:
Male mice lacking Ing2 were infertile, with significantly reduced sperm numbers (~2% of wild type) and motility (~10% of wild type)
Testicular abnormalities included degeneration of seminiferous tubules and meiotic arrest before pachytene stage
Molecular analysis showed incomplete meiotic recombination, p53 induction, and enhanced apoptosis
The phenotype was only partially rescued by concomitant loss of p53, indicating both p53-dependent and p53-independent mechanisms
Arrest of spermatocytes was characterized by lack of HDAC1 accumulation and deregulated chromatin acetylation
Parameter | Wild-type | Ing2-/- | Statistical Significance |
---|---|---|---|
Sperm count | Normal | ~2% of wild-type | P<0.0001 |
Sperm motility | Normal | ~10% of wild-type | P<0.0001 |
Seminiferous tubules | Intact | Degenerated | Qualitative observation |
Meiotic progression | Complete | Arrested before pachytene | Qualitative observation |
Apoptosis | Minimal | Enhanced | Qualitative observation |
Research methodologies to investigate these mechanisms include:
Histological analysis of testicular tissue
TUNEL assays to measure apoptosis
Immunofluorescence to detect stage-specific markers of spermatogenesis
ChIP-seq to identify ING2 binding sites in germline chromatin
RNA-seq to analyze transcriptional changes in ING2-deficient testes
ING2, as a subunit of the mSin3A-HDAC1 complex, specifically binds to tri-methylated lysine 4 of histone H3 (H3K4me3) via its PHD finger domain . This interaction enables ING2 to:
Recognize specific chromatin states associated with active transcription
Recruit histone deacetylase complexes to regulate gene expression
Modify local chromatin structure in response to DNA damage signals
Coordinate stage-specific histone modifications essential for processes like spermatogenesis
The chromatin-binding function of ING2 requires:
An intact PHD finger domain for H3K4me3 recognition
Association with the mSin3A-HDAC1 complex for effector functions
Proper nuclear localization for chromatin access
Methodological approaches to study ING2 chromatin interactions include:
ChIP-seq to map genomic binding sites
ATAC-seq to assess chromatin accessibility changes
Histone modification profiling in ING2-deficient cells
Structure-function analyses of PHD domain mutations
Proximity ligation assays to visualize chromatin interactions in situ
Several experimental models have proven valuable for investigating ING2 function:
Cellular Models:
Animal Models:
Molecular Tools:
When selecting an experimental model, researchers should consider:
The specific aspect of ING2 function under investigation
The evolutionary conservation of pathways between model organisms and humans
The availability of reagents and genetic tools
The physiological relevance to human disease processes
To study ING2's transcriptional regulatory functions, researchers should employ a combination of approaches:
Transcriptomic Analysis:
RNA-seq to identify differentially expressed genes
ChIP-seq to map ING2 binding sites genome-wide
ATAC-seq to assess changes in chromatin accessibility
CUT&RUN for high-resolution mapping of chromatin interactions
Reporter Assays:
Protein-Protein Interactions:
Chromatin Modifications:
ChIP for specific histone marks (e.g., H3K4me3, acetylation)
Sequential ChIP to identify co-occupancy with other factors
Functional assays for HDAC activity in ING2-containing complexes
Transcriptional Assay | Application | Advantage | Challenge |
---|---|---|---|
RNA-seq | Global expression changes | Comprehensive | Indirect ING2 effects |
ChIP-seq | Direct binding sites | Maps all targets | Antibody specificity |
Reporter assays | Specific pathway activation | Direct readout | Artificial system |
Gal4-fusion assays | Transcription factor activity | Isolates specific factors | May not reflect endogenous context |
Though the search results don't directly address therapeutic targeting of ING2, researchers interested in this question could consider these methodological approaches:
Target Validation:
Analyze ING2 expression in patient samples versus healthy controls
Correlate expression with disease progression and outcomes
Perform CRISPR-Cas9 knockout/knockdown in disease models
Utilize patient-derived xenografts to assess relevance
Intervention Approaches:
Develop small molecule inhibitors targeting the PHD domain
Design peptide inhibitors of ING2-protein interactions
Explore antisense oligonucleotides or siRNA for expression modulation
Consider degrader technologies (PROTACs) for protein depletion
Phenotypic Screening:
Establish cellular assays based on ING2-dependent processes
Screen compound libraries for modulators of these processes
Validate hits through target engagement studies
Assess effects on downstream pathways (e.g., TGF-β signaling)
Biomarker Development:
Identify gene expression signatures associated with ING2 activity
Develop assays to measure ING2 complex formation
Correlate chromatin modifications with ING2 function
Establish patient stratification criteria for clinical studies
Research challenges in targeting ING2 would include:
Achieving specificity among PHD domain-containing proteins
Determining appropriate disease indications
Understanding potential side effects (especially on fertility)
Developing appropriate drug delivery methods
While the search results focus specifically on ING2, researchers should note that ING2 belongs to a family of proteins with related but distinct functions:
Structural Comparison:
All ING family members contain a conserved PHD finger domain
ING proteins differ in their N-terminal domains
Different family members associate with distinct protein complexes
Functional Overlap and Distinction:
Different ING proteins may interact with p53 through distinct mechanisms
Family members may show tissue-specific expression patterns
Knockout phenotypes vary between family members
Research Methodology:
Comparative expression analysis across tissues
Domain-swapping experiments to identify functional determinants
Cross-complementation studies in knockout models
Comparative ChIP-seq to map binding site specificities
Research approaches should include:
Side-by-side functional assays with multiple ING family members
Careful antibody validation to ensure specificity
Consideration of compensatory mechanisms in knockout models
Evolutionary analysis to understand functional divergence
Based on the search results, several areas of ING2 research contain apparent contradictions or knowledge gaps that represent opportunities for further investigation:
Dual Role in p53 Signaling:
Role in Cancer:
Mechanistic Questions:
Therapeutic Implications:
Could targeting ING2 have unintended consequences on fertility?
How can specificity be achieved given ING2's involvement in fundamental cellular processes?
Research methodologies to address these contradictions include:
Careful dose-response studies to evaluate concentration-dependent effects
Tissue-specific and inducible knockout models to resolve temporal effects
Single-cell approaches to identify cell-type specific functions
Detailed structure-function analyses to map interaction domains
The Inhibitor of Growth (ING) family comprises 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 proteins are highly conserved across species and are involved in various cellular processes through their interactions with chromatin and other proteins.
The ING2 gene encodes the Inhibitor of Growth Family, Member 2 protein, also known as p33ING2 . This protein is involved in several critical cellular functions, including DNA repair, apoptosis, and cell cycle regulation. ING2 is known to interact with histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes, modulating their activity and influencing chromatin structure and gene expression .
ING2 contains several functional domains that enable its interaction with chromatin and other proteins:
ING2 plays a pivotal role in various biological processes:
As a tumor suppressor, ING2 is often downregulated or mutated in various cancers. Its loss of function can lead to uncontrolled cell proliferation and tumor development. Research has shown that restoring ING2 function in cancer cells can inhibit their growth and induce apoptosis, highlighting its potential as a therapeutic target .