ING2 Human

Inhibitor of Growth Family, Member 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ING2 Human

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.

Structure and Function of ING2

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.

Key Features of ING2 Structure:

  • 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 .

Role of ING2 in Gene Regulation

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 .

Table: Genes Regulated by ING2

GeneFunctionRegulation by ING2
MMP13Tissue remodeling, collagen degradationUpregulated
PAI-1Inhibits fibrinolysis, involved in tissue repairUpregulated
HSPA1AHeat shock protein, stress responseUpregulated
MRPL12Mitochondrial ribosomal protein, involved in mtDNA transcriptionRegulated through ubiquitination

ING2 and Mitochondrial Function

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 in Cancer and Tissue Repair

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 .

Table: ING2's Role in Cancer and Tissue Repair

Cancer TypeRole of ING2
Breast CancerAssociated with carcinogenesis
Gynecological CancerInvolved in carcinogenesis
Soft-Tissue SarcomasActs as a tumor suppressor

ING2 in Spermatogenesis

ING2 is crucial for spermatogenesis. Mice deficient in ING2 exhibit abnormal spermatogenesis and infertility, highlighting its role in reproductive health .

Product Specs

Introduction
As a member of the inhibitor of growth (ING) family, Inhibitor of Growth Family, Member 2 (ING2) interacts with and regulates the activity of histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes. These complexes play crucial roles in DNA repair and apoptosis. ING2 is believed to be involved in p53/TP53 activation and p53/TP53-dependent apoptotic pathways, potentially by enhancing the acetylation of p53/TP53. It is a component of an mSin3A-like corepressor complex, which is likely involved in the deacetylation of nucleosomal histones. The activity of ING2 is modulated by its binding to phosphoinositides (PtdInsPs).
Description
ING2 Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 303 amino acids (specifically, amino acids 1-280). It possesses a molecular mass of 35.2 kDa. A 23 amino acid His-tag is fused to the N-terminus of ING2, and purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The ING2 protein solution (1 mg/ml) contains 20mM Tris-HCl buffer with a pH of 8.0, 0.4M UREA, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep the vial at 4°C. For longer periods, store frozen at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. It is important to avoid repeated freeze-thaw cycles.
Purity
SDS-PAGE analysis confirms a purity greater than 85.0%.
Synonyms
Inhibitor Of Growth Family Member 2, ING1L, Inhibitor Of Growth 1-Like Protein, P33ING2, ING1Lp, P32, Inhibitor Of Growth Family Member 1-Like, Inhibitor Of Growth Protein 2, ING2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMLGQQQQ QLYSSAALLT GERSRLLTCY VQDYLECVES LPHDMQRNVS VLRELDNKYQ ETLKEIDDVY EKYKKEDDLN QKKRLQQLLQ RALINSQELG DEKIQIVTQM LELVENRARQ MELHSQCFQD PAESERASDK AKMDSSQPER SSRRPRRQRT SESRDLCHMA NGIEDCDDQP PKEKKSKSAK KKKRSKAKQE REASPVEFAI DPNEPTYCLC NQVSYGEMIG CDNEQCPIEW FHFSCVSLTY KPKGKWYCPK CRGDNEKTMD KSTEKTKKDR RSR.

Q&A

What is ING2 and what are its primary functions in human cells?

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

How does ING2 interact with tumor suppressor pathways?

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

What is known about ING2 expression patterns in human tissues?

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

How does ING2 contribute to TGF-β signaling pathways?

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 ConstructEffect on TGF-β SignalingMechanism
Wild-type ING2Enhanced transcriptionPromotes Smad2/3 activity
ING2 (ΔN)Partial enhancementN-terminal deletion affects function
ING2 (ΔC)Significantly reducedLoss of PHD domain impairs activity
ING2 (ΔPHD)Significantly reducedCannot 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

What mechanisms underlie ING2-mediated regulation of spermatogenesis?

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

ParameterWild-typeIng2-/-Statistical Significance
Sperm countNormal~2% of wild-typeP<0.0001
Sperm motilityNormal~10% of wild-typeP<0.0001
Seminiferous tubulesIntactDegeneratedQualitative observation
Meiotic progressionCompleteArrested before pachyteneQualitative observation
ApoptosisMinimalEnhancedQualitative 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

How do the chromatin-binding properties of ING2 influence gene expression?

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

  • Stage-specific regulation of chromatin acetylation

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

What are the optimal experimental models for studying ING2 function?

Several experimental models have proven valuable for investigating ING2 function:

  • Cellular Models:

    • Human cell lines (HepG2, Mv1Lu) for studying TGF-β signaling

    • Primary testicular cells for spermatogenesis studies

    • Inducible expression systems to control ING2 levels temporally

  • Animal Models:

    • Ing2 knockout mice provide insights into developmental functions

    • Conditional knockout models to study tissue-specific effects

    • Double knockout models (e.g., Ing2/p53) to dissect pathway interactions

  • Molecular Tools:

    • Expression vectors for wild-type and mutant ING2 proteins

    • Reporter constructs with TGF-β or p53-responsive elements

    • Domain deletion constructs to map functional regions

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

How can researchers effectively investigate ING2's role in transcriptional regulation?

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:

    • Luciferase reporter systems with specific promoters (e.g., PAI-1)

    • Gal4-fusion heterologous transcription assays to measure Smad2/3 activity

    • Mutational analysis of response elements

  • Protein-Protein Interactions:

    • Co-immunoprecipitation to identify transcription factor complexes

    • Proximity ligation assays for in situ visualization

    • Domain mapping through deletion mutants

    • Mass spectrometry to identify novel interaction partners

  • 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 AssayApplicationAdvantageChallenge
RNA-seqGlobal expression changesComprehensiveIndirect ING2 effects
ChIP-seqDirect binding sitesMaps all targetsAntibody specificity
Reporter assaysSpecific pathway activationDirect readoutArtificial system
Gal4-fusion assaysTranscription factor activityIsolates specific factorsMay not reflect endogenous context

What strategies can be employed to investigate the therapeutic potential of targeting ING2?

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

How does ING2 function compare to other ING family members?

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

What contradictions or unresolved questions exist in ING2 research?

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:

    • Endogenous ING2 inhibits senescence, yet overexpressed ING2 enhances p53-driven senescence

    • This apparent contradiction suggests context-dependent functions requiring further elucidation

  • Role in Cancer:

    • Despite functioning as a putative tumor suppressor, "spontaneous tumor incidence and spectrum were similar in wild-type and Ing2-deficient mice"

    • This unexpected finding requires reconciliation with in vitro studies

  • Mechanistic Questions:

    • How does ING2 coordinate with SnoN to activate TGF-β responses when SnoN is commonly thought to act as a transcriptional corepressor?

    • What determines whether ING2 functions in gene activation versus repression?

  • 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

Product Science Overview

Introduction

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.

ING2 Gene and Protein

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 .

Functional Domains

ING2 contains several functional domains that enable its interaction with chromatin and other proteins:

  • Plant Homeodomain (PHD) Finger: This domain allows ING2 to bind to methylated histones, particularly histone H3 trimethylated at lysine 4 (H3K4me3), which is associated with active transcription.
  • Leucine Zipper-like Domain: This domain facilitates protein-protein interactions, including those with p53, a key regulator of the cell cycle and apoptosis .
Biological Functions

ING2 plays a pivotal role in various biological processes:

  • DNA Repair: ING2 is involved in the DNA damage response, enhancing the acetylation of p53 and promoting its activation. This leads to the transcription of genes involved in DNA repair and apoptosis .
  • Apoptosis: By modulating p53 activity, ING2 can induce apoptosis in response to cellular stress or DNA damage .
  • Cell Cycle Regulation: ING2 helps regulate the cell cycle by influencing the expression of genes involved in cell proliferation and growth arrest .
Role in Cancer

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 .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.