IER3 Human

Immediate Early Response 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Production

IER3 is encoded by the IER3 gene located on chromosome 6p21. The recombinant human IER3 protein (Catalogue No. PRO-1490) is produced in E. coli as a non-glycosylated polypeptide containing 105 amino acids (1–82 residues). Key features include:

PropertyDetails
Molecular Mass11.4 kDa
Post-Translational ModificationsN-terminal His-tag (23 aa) for purification; no glycosylation
Formulation0.25 mg/ml solution in 20 mM Tris-HCl (pH 8.0), 0.2M NaCl, 50% glycerol, 2 mM DTT
SourceE. coli

Functional Roles in Cellular Processes

IER3 modulates apoptosis, proliferation, and DNA repair through interactions with key signaling pathways:

Apoptosis Regulation

  • Protection from Fas/TNFA-induced apoptosis: IER3 inhibits dephosphorylation of ERK by PP2A-PPP2R5C, promoting survival .

  • BCL-2 Family Interaction: Binds MCL-1 and suppresses BCL-2/BCL-xL, inducing apoptosis in a context-dependent manner .

  • TAp73β Dependency: In cervical cancer, IER3 mediates TAp73β-induced apoptosis via BIM, which is essential for mitochondrial apoptosis .

Cell Cycle Control

  • NF-κB Pathway Modulation: Interacts with RelA/p65 to upregulate anti-apoptotic genes, influencing cell cycle progression .

  • ERK and PI3K/Akt Pathways: Regulates ERK phosphorylation and Akt signaling, impacting genomic stability and stress responses .

Stress Response

  • Induction by Stimuli: Rapidly upregulated by growth factors, cytokines, ionizing radiation, and viral infections .

  • Genomic Stability: Knockout mice show impaired DNA repair and susceptibility to hypertension .

Cancer-Dependent Dual Roles

Cancer TypeRoleMechanism
Cervical CancerTumor suppressorTAp73β → IER3 → BIM → Mitochondrial apoptosis
Pancreatic AdenocarcinomaOncogeneERK survival signaling; resistance to starvation-induced stress
Bladder CancerOncogene (cytoplasmic overexpression)Correlates with nodal metastasis and poor survival
Myelodysplastic Syndrome (MDS)DysregulationChromosomal rearrangements (t(6;9)) and amplifications linked to disease progression

Expression Patterns

IER3 mRNA is widely expressed in epithelial tissues with high turnover, including skin, gastrointestinal tract, and lungs . Protein expression is detected in diverse cell lines, such as cervical (HeLa), hematopoietic (MDS), and bladder cancer cells .

Tissue/Cell LineExpression LevelSource
Epithelial TissuesHigh
HeLa (Cervical Cancer)Inducible (TAp73β-dependent)
MDS MarrowDysregulated (35% up, 26% down)
Bladder CancerCytoplasmic overexpression

Clinical Implications

  • Cancer Therapy: IER3’s dual role complicates therapeutic targeting. In cervical cancer, enhancing TAp73β → IER3 signaling may improve apoptosis, while inhibiting it in pancreatic cancer could reduce stress resistance .

  • Biomarker Potential: IER3 rearrangements in MDS and cytoplasmic overexpression in bladder cancer highlight its utility as a prognostic marker .

  • Hypertension and Immunity: Knockout mice exhibit hypertension and immune dysregulation, suggesting IER3’s role in cardiovascular and inflammatory diseases .

Future Directions

  • Mechanistic Studies: Elucidating context-specific interactions (e.g., ERK vs. NF-κB pathways) to refine therapeutic strategies.

  • Clinical Trials: Investigating IER3 as a predictive biomarker in cancers with HPV or stress-induced pathogenesis.

  • Synthetic Biology: Engineering IER3 variants to modulate survival signaling in targeted therapies.

References

  1. Prospec Bio. IER3 Protein Human Recombinant. Link

  2. OceanRep. Role of IER3 in Cell Cycle and Apoptosis. Link

  3. Nature. IER3 Mediates TAp73β-Induced Apoptosis in Cervical Cancer. Link

  4. Maayan Lab. IER3 Functional Associations. Link

  5. Cancer Genetics Web. IER3 in Oncogenesis. Link

  6. Wikipedia. IER3 Gene. Link

  7. PubMed. IER3’s Role in Immune and Cardiovascular Functions. Link

  8. Cancer Research. IER3 Rearrangements in MDS. Link

  9. PMC. IER3 in Bladder Cancer. Link

Product Specs

Introduction
The Immediate Early Response 3 (IER3) protein plays a crucial role in safeguarding cells against apoptosis triggered by Fas or tumor necrosis factor type alpha. It participates in the ERK signaling pathway by preventing the dephosphorylation of ERK by the phosphatase PP2A-PPP2R5C holoenzyme. Additionally, IER3 acts as an effector downstream of ERK, mediating cell survival. As a component of the NUPR1/RELB/IER3 survival pathway, IER3 confers significant resistance to pancreatic ductal adenocarcinoma cells against stress conditions such as starvation.
Description
Recombinant human IER3, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 105 amino acids (specifically, amino acids 1-82). It has a molecular mass of 11.4 kDa. A 23 amino acid His-tag is fused to the N-terminus of IER3. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
A clear solution, sterilized by filtration.
Formulation
The IER3 protein solution (0.25 mg/ml) is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 50% glycerol, and 2mM DTT.
Stability
For optimal storage, keep at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 85.0%.
Synonyms
immediate early response 3, DIF-2, DIF2, GLY96, IEX-1, IEX-1L, IEX1, PRG1, Protein DIF-2, Immediate early protein GLY96, PACAP-responsive gene 1 protein, Differentiation-dependent gene 2 protein, IER3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMCHSRSC HPTMTILQAP TPAPSTIPGP RRGSGPEIFT FDPLPEPAAA PAGRPSASRG HRKRSRRVLY PRVVRRQLPV EEPNP.

Q&A

What is IER3 and what are its primary functions in human cells?

IER3 (Immediate Early Response 3), also known as IEX-1, is a stress-inducible gene belonging to the immediate early response gene family, which includes other well-characterized members like c-fos, c-jun, and c-myc . It is rapidly and transiently induced in response to diverse stimuli including growth factors, cytokines, ionizing radiation, viral infections, and other forms of cellular stress . IER3 plays pivotal roles in regulating cell apoptosis, proliferation, differentiation, and DNA repair . Mechanistically, it functions through involvement in several critical signaling pathways, including NF-κB, MAPK/ERK, and PI3K/Akt, affecting cell survival decisions under various stress conditions . This gene's expression and functionality are highly context-dependent, which explains its diverse and sometimes contradictory effects across different cell types and cancer models.

How does IER3 expression typically respond to cellular stress?

IER3 demonstrates a characteristic rapid and transient induction pattern in response to cellular stressors. As a stress-inducible gene, IER3 is quickly activated following exposure to stimuli such as growth factors, inflammatory cytokines, ionizing radiation, viral infections, and chemical stressors . The gene's expression is typically low under normal physiological conditions but shows significant upregulation within minutes to hours after stress exposure . This rapid response classifies it as an immediate early gene, enabling cells to quickly adapt to environmental challenges . The transient nature of IER3 expression is crucial for its function, as prolonged activation could lead to dysregulated cellular responses. In cancer research models, various treatments that induce cellular stress, such as chemotherapeutic agents or radiation, have been shown to alter IER3 expression levels, making it an important marker for cellular stress responses and potential therapeutic interventions .

What transcription factors and regulatory elements control IER3 expression?

IER3 expression is regulated by multiple transcription factors, with p53 being a particularly significant regulator in certain cellular contexts. In neuroblastoma cell lines with wild-type p53 (such as SH-SY5Y), p53 knockdown significantly reduces IER3 mRNA and protein levels, confirming IER3 as a p53-responsive gene . Other transcription factors known to regulate IER3 include NF-κB, AP-1, Sp1, and c-Myc, which bind to specific regulatory elements in the IER3 promoter region . The gene also contains several response elements, including p53 response elements, NF-κB binding sites, and growth factor response elements . Additionally, epigenetic mechanisms play a role in IER3 regulation, as evidenced by the differential chromatin modification patterns (H3K4me3 and H3K27me3) observed at the promoter regions of IER3 target genes across different cancer cell types . The complex interplay of these regulatory mechanisms contributes to the context-dependent expression and function of IER3 in different cellular environments.

How does IER3 expression vary across different cancer types?

IER3 expression demonstrates remarkable variability across different cancer types, which directly correlates with its functional impact as either an oncogene or tumor suppressor. In cervical carcinoma, lung adenocarcinoma, and breast cancer, IER3 exhibits oncogenic properties and is often upregulated . Contrastingly, in neuroblastoma, higher IER3 expression correlates with favorable prognosis, suggesting a tumor suppressor role in this context . This cancer type-specific expression pattern is further evidenced by clinical data showing that low IER3 expression combined with high MYCN levels is associated with poor survival in neuroblastoma patients .

In bladder cancer, increased IER3 expression has been observed and appears to be clinically significant . The differential expression across cancer types appears to be regulated by both genetic and epigenetic mechanisms. Cancer-specific transcription factor activity, epigenetic modifications, and interactions with other genes (such as IER3-AS1) all contribute to this variable expression pattern . Research using ChIP-seq analysis has revealed that IER3 occupancy on target genes also varies between cancer types, providing a molecular basis for its dual functionality .

What is the significance of IER3 and IER3-AS1 co-expression patterns in different cancers?

The relationship between IER3 and its antisense RNA partner, IER3-AS1, represents a critical aspect of IER3's cancer biology. In cancers where IER3 exhibits oncogenic functions (such as cervical, lung, and breast carcinomas), there is typically a strong positive expression correlation between IER3 and IER3-AS1 . This high correlation is associated with poor clinical outcomes, suggesting a cooperative oncogenic effect . Conversely, in cancers where IER3 acts as a tumor suppressor (such as glioblastoma and neuroblastoma), the expression correlation between these transcripts is remarkably poor .

How do researchers quantify IER3 expression in clinical samples?

Researchers employ multiple complementary techniques to quantify IER3 expression in clinical samples, each offering distinct advantages. Immunohistochemistry (IHC) is widely used to examine the subcellular localization and expression levels of IER3 protein in tissue specimens . This technique allows visualization of IER3 protein distribution within tissue architecture and enables scoring of expression intensity. For mRNA quantification, quantitative reverse transcription PCR (RT-qPCR) provides a precise method to measure IER3 transcript levels, as demonstrated in studies validating IER3 target genes in neuroblastoma cell lines .

RNA sequencing (RNA-seq) offers a more comprehensive approach, allowing genome-wide expression analysis and identification of IER3-regulated pathways, as shown in functional enrichment studies of IER3 knockdown cells . For protein-level detection, Western blotting is commonly used to quantify IER3 protein expression and validate its impact on downstream targets . More sophisticated approaches include chromatin immunoprecipitation (ChIP) analysis to examine IER3 occupancy on target gene promoters . For clinical prognostic evaluation, researchers often correlate IER3 expression levels with patient survival data using Kaplan-Meier analyses, as demonstrated in studies comparing IER3 expression patterns between neuroblastoma and cervical cancer datasets . The integration of these multiple techniques provides a robust assessment of IER3 expression and its clinical significance in human cancers.

Which signaling pathways does IER3 primarily interact with and how?

IER3 interacts with several critical signaling pathways that regulate cell survival, proliferation, and stress responses. The protein is deeply involved in modulating the NF-κB pathway, where it can either enhance or suppress NF-κB transcriptional activity depending on cellular context and stress conditions . Under conditions causing DNA damage, IER3 can suppress NF-κB activity in the nucleus, promoting apoptosis rather than survival .

The MAPK/ERK pathway represents another major signaling axis modulated by IER3. The protein affects cell differentiation and proliferation by regulating MAPK/ERK signaling components, with consequent effects on downstream target gene expression . Research has demonstrated that IER3-dependent oncogenic functions, including increased cell proliferation and S-phase prolongation, are mediated through the activation of immediate early response pathways comprising JUN and FOS genes, which are downstream of the EGR2 oncogene .

Additionally, IER3 influences the PI3K/Akt signaling pathway, affecting cell survival decisions . The protein also impacts ubiquitin-proteasome activity, providing another mechanism by which it can regulate protein stability and cellular processes . These interactions collectively explain how IER3 can exert diverse effects on cellular phenotypes across different cancer types, with the specific outcome depending on which pathways predominate in a given cellular context.

What is the relationship between IER3 and p53 in cancer progression?

The relationship between IER3 and p53 represents a significant axis in cancer biology, particularly in neuroblastoma. Research has established IER3 as a p53-responsive gene in neuroblastoma cell lines, where p53 activation leads to increased IER3 expression . In SH-SY5Y cells, which contain wild-type p53, siRNA-mediated knockdown of p53 significantly reduces both IER3 mRNA and protein levels . In contrast, in SK-N-BE(2) cells with mutated p53, IER3 expression remains unchanged following p53 knockdown, confirming the specificity of this regulatory relationship .

This p53-IER3 axis has important implications for cancer progression. In neuroblastoma, where IER3 functions as a tumor suppressor, the activation of IER3 by p53 may represent a mechanism through which p53 exerts its tumor-suppressive effects . The good prognosis associated with higher IER3 expression in neuroblastoma patients aligns with this model . Furthermore, treatments that activate p53, such as Nutlin and Selinexor, increase IER3 expression in neuroblastoma cell lines, suggesting potential therapeutic strategies targeting this pathway .

The p53-IER3 relationship may differ in cancers where IER3 functions as an oncogene, highlighting the context-dependent nature of these interactions. Understanding these nuanced relationships is crucial for developing targeted therapeutic approaches that consider the dual nature of IER3 in different cancer types.

How does IER3 differentially regulate EGR2 and ADAM19 in various cancer types?

In HeLa cells, where IER3 functions as an oncogene, IER3 binding leads to upregulation of EGR2, which subsequently activates the downstream immediate early response pathway comprising JUN and FOS genes . This activation promotes oncogenic functions such as increased cell proliferation, inhibition of apoptosis, and S-phase prolongation . Conversely, in neuroblastoma cells where IER3 acts as a tumor suppressor, IER3 binding preferentially upregulates ADAM19, which inhibits cell invasion and migration, contributing to tumor suppression .

The mechanism behind this differential regulation involves epigenetic factors. Analysis reveals distinct patterns of H3K4me3 (active) and H3K27me3 (repressive) chromatin modifications at the promoter regions of EGR2 and ADAM19 in different cell types . These epigenetic differences likely influence how IER3 binding affects gene expression in a context-dependent manner. This differential regulation explains how the same protein can exert opposite effects in different cancer types, highlighting the importance of cellular context in determining IER3 function .

What are the most effective methods for studying IER3 function in cancer cell lines?

For comprehensive investigation of IER3 function in cancer cell lines, researchers employ multiple complementary approaches. Gene knockdown using shRNA or siRNA technology represents a fundamental approach, as demonstrated in studies generating IER3 loss-of-function stable cell lines in neuroblastoma . This technique allows direct assessment of IER3's impact on cellular phenotypes such as proliferation, apoptosis, and migration.

RNA sequencing following IER3 manipulation provides valuable insights into global gene expression changes and affected pathways, as shown in functional enrichment analyses revealing cancer type-specific pathways regulated by IER3 . Chromatin immunoprecipitation sequencing (ChIP-seq) using IER3 antibodies enables genome-wide analysis of IER3 protein occupancy on target gene promoters, revealing direct regulatory interactions . This can be validated through ChIP-qPCR assays for specific target genes such as EGR2 and ADAM19 .

Functional assays including proliferation assays, cell cycle analysis, apoptosis assays, and migration/invasion assays are essential for characterizing IER3-dependent phenotypes . Subcellular localization studies using techniques like RNA fluorescence in situ hybridization help elucidate the spatial distribution of IER3 and its antisense partner IER3-AS1, which influences their functional interaction . Protein-protein interaction studies identify IER3's binding partners in different cellular contexts. Finally, correlation analyses between in vitro findings and patient data from resources like the R2 Genomics Platform strengthen the clinical relevance of experimental observations .

How can researchers effectively model the dual oncogenic/tumor suppressor roles of IER3?

Modeling the dual nature of IER3 requires careful selection of experimental systems and analytical approaches. Based on current research, an effective modeling strategy involves parallel investigation in multiple cancer cell lines representing both contexts where IER3 functions as an oncogene (e.g., HeLa cells for cervical carcinoma) and as a tumor suppressor (e.g., SH-SY5Y, SK-N-BE(2), and KELLY for neuroblastoma) . This comparative approach allows direct observation of context-dependent functions.

Genetic manipulation through both knockdown and overexpression systems in these model cell lines enables comprehensive functional characterization . Importantly, researchers should conduct identical functional assays across all cell line models, including proliferation, apoptosis, cell cycle, migration, and invasion assays, to directly compare IER3's impact . RNA-seq analysis following IER3 manipulation across different cell types reveals cancer-specific gene expression patterns and pathway alterations, illuminating the molecular basis of its dual function .

ChIP-seq and ChIP-qPCR analyses across different cancer cell types provide insights into how the same protein can differentially regulate target genes in a context-dependent manner . Investigation of epigenetic modifications (such as H3K4me3 and H3K27me3) at IER3 target gene promoters helps explain the mechanistic basis for differential gene regulation . Subcellular localization studies examining the spatial distribution of IER3 and IER3-AS1 in different cancer contexts reveal how compartmentalization influences function . Finally, correlation with clinical data from patient cohorts validates the biological relevance of experimental findings and strengthens translational implications .

What technical challenges arise when analyzing IER3 expression and function, and how can they be addressed?

Analyzing IER3 expression and function presents several technical challenges that require specific methodological considerations. The rapid and transient nature of IER3 induction as an immediate early response gene makes timing critical in experimental design . Researchers should implement time-course experiments with frequent sampling intervals to capture the dynamic expression patterns following stimulation. The context-dependent function of IER3 necessitates parallel analysis in multiple cell types representing different cancer contexts to avoid misinterpretation of results .

The functional interplay between IER3 and its antisense RNA partner IER3-AS1 adds complexity to expression analysis . This interaction requires simultaneous analysis of both transcripts, preferably with spatial resolution to determine their subcellular localization patterns. For protein-level analysis, IER3's relatively low basal expression in some cell types may require sensitive detection methods . Additionally, the protein's involvement in multiple signaling pathways creates interpretive challenges, as observed effects could result from alterations in various pathways .

To address these challenges, researchers should employ multiple complementary techniques for expression analysis (RT-qPCR, RNA-seq, Western blotting, IHC), with appropriate controls and normalization strategies . Subcellular fractionation combined with quantitative PCR helps determine the spatial distribution of IER3 and IER3-AS1 . Pathway-specific inhibitors or activators can help dissect which signaling pathways mediate observed IER3 effects . For functional analysis, conditional or inducible expression/knockdown systems provide temporal control, while CRISPR-based approaches offer more complete gene inactivation than RNA interference methods . Finally, integration of multiple datasets, including in vitro experiments, patient-derived samples, and clinical data, strengthens the interpretation of IER3's complex biology .

How does IER3 expression correlate with patient prognosis across different cancer types?

IER3 expression demonstrates striking cancer type-specific correlations with patient prognosis, reflecting its dual role as either an oncogene or tumor suppressor. In neuroblastoma, higher IER3 expression strongly correlates with favorable prognosis, while low expression is associated with worse outcomes . This relationship is further impacted by other factors, as evidenced by the observation that low IER3 levels combined with high MYCN expression correlate with particularly poor survival in neuroblastoma patients .

Conversely, in cancers where IER3 functions as an oncogene, such as cervical carcinoma, higher expression correlates with worse clinical outcomes . Kaplan-Meier survival analyses comparing neuroblastoma and cervical cancer datasets confirm these opposing prognostic patterns . The expression and prognostic significance of IER3 target genes also follow cancer-specific patterns. For instance, EGR2 (which mediates IER3's oncogenic functions) shows positive correlation with poor prognosis in cervical cancer but not in neuroblastoma . Meanwhile, ADAM19 (which mediates IER3's tumor suppressor functions) correlates with better prognosis in neuroblastoma but not in cervical cancer .

In bladder cancer, increased IER3 expression has been observed, with potential clinical significance that requires further investigation . These diverse prognostic correlations highlight the importance of cancer type-specific evaluation of IER3 expression for accurate prognostic assessment, underscoring the danger of generalizing findings across different cancer types.

What is the potential of IER3 as a biomarker for cancer diagnosis or prognosis?

IER3 shows considerable promise as a biomarker for cancer diagnosis and prognosis, particularly due to its cancer type-specific expression patterns and clinical correlations. Its differential expression across various human cancers correlates with either poor or favorable prognosis depending on the cancer type, making it a potentially valuable prognostic indicator when evaluated in the appropriate context . In neuroblastoma, higher IER3 expression predicts favorable outcomes, suggesting utility as a positive prognostic marker in this pediatric cancer .

The combination of IER3 with other markers enhances its biomarker potential. For instance, the inverse relationship between IER3 and MYCN expression in neuroblastoma provides a stronger prognostic indicator than either marker alone . Similarly, evaluation of IER3 target genes such as EGR2 and ADAM19 alongside IER3 itself could offer more comprehensive prognostic information .

Beyond expression level analysis, examination of IER3's antisense partner IER3-AS1 and their expression correlation adds another dimension to its biomarker potential . In cancers where these transcripts show high correlation, this pattern itself may serve as a prognostic indicator . For clinical application, immunohistochemistry protocols for IER3 detection in tissue specimens have been established and validated, facilitating potential translation to diagnostic use . As research advances, multi-marker panels incorporating IER3 alongside its target genes and interacting partners could provide more nuanced prognostic information across diverse cancer types .

How might understanding IER3's dual role inform cancer therapy development?

The dual role of IER3 as both an oncogene and tumor suppressor offers unique opportunities for cancer therapy development, but necessitates carefully tailored approaches based on cancer type. In cancers where IER3 functions as an oncogene (such as cervical and breast cancer), therapeutic strategies could focus on inhibiting IER3 expression or activity . This might involve developing small molecule inhibitors targeting IER3 itself or disrupting its interaction with oncogenic partners .

Conversely, in cancers where IER3 acts as a tumor suppressor (such as neuroblastoma), therapeutic approaches might aim to enhance IER3 expression or activity . Since IER3 is a p53-responsive gene in neuroblastoma, treatments that activate p53 (such as Nutlin and Selinexor) could indirectly increase IER3 expression, potentially enhancing its tumor suppressor functions . This strategy might be particularly relevant for the significant number of low-risk neuroblastoma patients who experience spontaneous tumor regression, a phenomenon that could potentially involve IER3-mediated mechanisms .

The identification of cancer-specific IER3 target genes provides additional therapeutic possibilities. In HeLa cells, targeting the IER3-EGR2-JUN/FOS axis might inhibit oncogenic functions, while in neuroblastoma, enhancing the IER3-ADAM19 pathway could strengthen tumor suppression . The differential spatial organization of IER3 and IER3-AS1 across cancer types suggests that targeting their interaction or localization might offer another therapeutic avenue . Additionally, epigenetic modifiers affecting the chromatin state of IER3 target genes could potentially switch IER3 function from oncogenic to tumor-suppressive or vice versa in specific contexts . These diverse approaches highlight how detailed understanding of IER3's context-dependent functions can inform precision medicine strategies for different cancer types.

How do epigenetic modifications influence IER3 function across different cancer contexts?

Epigenetic modifications play a crucial role in determining IER3's functional output across different cancer contexts, primarily by influencing the accessibility and activity of IER3 target genes. Research has revealed distinct patterns of histone modifications at the promoter regions of IER3 target genes in different cancer cell types . Specifically, the promoters of genes like EGR2 and ADAM19 show differential enrichment of activating (H3K4me3) and repressive (H3K27me3) histone marks in HeLa versus neuroblastoma cell lines .

These epigenetic differences likely explain how IER3, despite binding to the same target gene promoters in different cancer types, can drive opposite expression outcomes . For instance, in HeLa cells, the EGR2 promoter may feature a more permissive chromatin state that allows IER3 binding to enhance expression, while in neuroblastoma cells, a more repressive chromatin environment at the same promoter might prevent activation despite IER3 binding .

This epigenetic regulation creates a sophisticated mechanism by which the same protein can exert opposite functions in different cellular contexts. The cancer type-specific epigenetic landscape effectively determines which target genes respond to IER3 binding, thereby channeling IER3 activity toward either oncogenic or tumor-suppressive outcomes . Further research examining genome-wide chromatin accessibility (through techniques like ATAC-seq) in conjunction with IER3 ChIP-seq across different cancer types would provide deeper insights into how the epigenetic environment shapes IER3's functional diversity . Understanding these epigenetic mechanisms could potentially enable therapeutic strategies aimed at reprogramming IER3 function through epigenetic modifiers in specific cancer contexts.

What accounts for the contradictory research findings regarding IER3's role in cell proliferation and apoptosis?

The contradictory findings regarding IER3's impact on cell proliferation and apoptosis stem from several key factors related to its context-dependent function. Primarily, the cellular context plays a decisive role in determining IER3's functional output . In HeLa cells, IER3 promotes proliferation and inhibits apoptosis, while in neuroblastoma cells, it may have opposing effects . This context dependence is mediated through differential regulation of target genes—in HeLa cells, IER3 activates the oncogenic EGR2-JUN/FOS pathway, while in neuroblastoma, it preferentially activates the tumor-suppressive ADAM19 pathway .

The subcellular localization pattern of IER3 and its antisense partner IER3-AS1 further explains these contradictions . Their differential compartmentalization across cancer types enables interaction with distinct protein complexes, potentially leading to different functional outcomes . Cancer-specific epigenetic landscapes also contribute significantly to these contradictions by determining which target genes respond to IER3 binding .

Methodological differences across studies add another layer of complexity. Variations in cell culture conditions, knockdown/overexpression efficiency, timing of analyses (given IER3's dynamic expression pattern), and assay sensitivities can all influence experimental outcomes . Additionally, the activation state of key signaling pathways that interact with IER3 (such as NF-κB, MAPK/ERK, and PI3K/Akt) varies across cell types and experimental conditions, potentially altering IER3's effects .

To reconcile these contradictions, researchers should conduct parallel experiments across multiple cell types using standardized methodologies, perform time-course analyses to capture the dynamic nature of IER3 responses, and comprehensively assess pathway activation states alongside IER3 manipulation . Such approaches would provide a more nuanced understanding of how IER3 exerts seemingly opposite effects across different experimental systems.

What emerging technologies or methodological approaches might advance our understanding of IER3 biology?

Emerging technologies offer promising avenues to deepen our understanding of IER3's complex biology. Single-cell omics approaches, including single-cell RNA-seq and single-cell ATAC-seq, would reveal cell-to-cell heterogeneity in IER3 expression, chromatin accessibility, and target gene regulation within tumors . This could help identify specific cellular subpopulations where IER3 exerts its strongest effects and potentially explain contradictory findings from bulk analyses.

CRISPR-based technologies beyond simple knockouts, such as CRISPR activation (CRISPRa) and CRISPR interference (CRISPRi), would enable more precise temporal and spatial control over IER3 expression . CRISPR-based epigenome editing could directly test how modifying specific histone marks at IER3 target gene promoters affects their responsiveness to IER3 regulation . Proximity labeling techniques like BioID or APEX could identify the cancer type-specific protein interactome of IER3, providing insights into how its binding partners differ between contexts where it functions as an oncogene versus a tumor suppressor .

Advanced imaging techniques, including super-resolution microscopy combined with single-molecule RNA FISH, would offer detailed visualization of the spatial organization and potential interaction between IER3 and IER3-AS1 in different cellular compartments . Patient-derived organoids and xenografts representing diverse cancer types would provide more physiologically relevant models to study IER3's context-dependent functions compared to traditional cell lines .

Multi-omics integration approaches combining transcriptomics, proteomics, epigenomics, and metabolomics data would offer a more comprehensive view of how IER3 impacts cellular physiology across different cancer contexts . Computational approaches leveraging machine learning algorithms could help identify patterns in large datasets that predict IER3's functional impact based on cellular context . Together, these advanced methodologies would significantly enhance our understanding of the molecular mechanisms underlying IER3's dual role in cancer biology.

What are the critical unanswered questions about IER3 function in human cancers?

Despite significant advances in IER3 research, several critical questions remain unanswered. Perhaps most fundamentally, the precise molecular switch that determines whether IER3 functions as an oncogene or tumor suppressor in a given cellular context remains elusive . While current research has identified differential target gene regulation and epigenetic factors as contributors, the master regulatory mechanism that controls this functional switch requires further investigation .

The potential role of post-translational modifications in regulating IER3 function represents another significant knowledge gap. Whether IER3 undergoes cancer type-specific phosphorylation, ubiquitination, or other modifications that alter its activity or binding preferences remains largely unexplored . Additionally, the comprehensive protein interactome of IER3 across different cancer types has not been fully characterized, limiting our understanding of how protein-protein interactions influence its context-dependent functions .

The regulatory relationship between IER3 and its antisense partner IER3-AS1 warrants deeper investigation, particularly regarding how their interaction is controlled and how cellular compartmentalization affects their functional interplay . The role of IER3 in cancer stem cells and tumor heterogeneity remains an open question, as does its potential involvement in therapy resistance mechanisms .

From a clinical perspective, the potential of IER3 as a therapeutic target has not been thoroughly evaluated, nor have specific inhibitors or activators been developed . The prognostic value of IER3 expression in comprehensive patient cohorts across multiple cancer types requires further validation to establish its clinical utility . Addressing these questions would significantly advance our understanding of IER3 biology and potentially open new avenues for cancer diagnosis and treatment.

How might IER3 research inform our understanding of spontaneous tumor regression in neuroblastoma?

IER3 research offers intriguing insights into the perplexing phenomenon of spontaneous tumor regression in neuroblastoma, which occurs in a significant number of low-risk patients . The finding that higher IER3 expression correlates with favorable prognosis in neuroblastoma suggests it may play a role in this regression process . As a tumor suppressor in neuroblastoma, IER3 could potentially contribute to spontaneous regression through several mechanisms that warrant further investigation.

The tumor suppressor functions of IER3 in neuroblastoma, particularly its regulation of the ADAM19 pathway which inhibits cell invasion and migration, may directly contribute to tumor regression by limiting aggressive tumor behavior . Furthermore, IER3's role in DNA replication and repair pathways in neuroblastoma suggests it might influence genomic stability, potentially pushing genetically damaged neuroblastoma cells toward apoptosis rather than continued proliferation .

As a p53-responsive gene in neuroblastoma, IER3 may mediate p53-dependent apoptotic responses to oncogenic stress or DNA damage, contributing to elimination of tumor cells . The observed inverse relationship between IER3 and MYCN expression in neuroblastoma patients is particularly relevant, as MYCN amplification is a major negative prognostic factor . This suggests that in non-MYCN-amplified tumors, higher IER3 expression might counteract residual oncogenic signaling, facilitating regression.

Future research directions should include detailed temporal analysis of IER3 expression during documented cases of spontaneous regression, investigation of IER3's role in neuroblastoma differentiation (as regression often involves differentiation of neuroblasts), and exploration of potential therapeutic strategies to enhance IER3 expression or activity specifically in high-risk neuroblastoma that typically does not undergo spontaneous regression . Such studies could provide valuable insights into not only neuroblastoma biology but also the general mechanisms of tumor regression that might be applicable to other cancer types.

What potential exists for developing IER3-targeted therapeutics with cancer type-specific effects?

The dual nature of IER3 as both an oncogene and tumor suppressor creates unique opportunities for developing cancer type-specific therapeutics. For cancers where IER3 functions as an oncogene (such as cervical and breast cancer), potential therapeutic strategies include small molecule inhibitors directly targeting IER3 protein, antisense oligonucleotides or siRNA-based approaches to reduce IER3 expression, and compounds disrupting the interaction between IER3 and its oncogenic binding partners .

Conversely, for cancers where IER3 acts as a tumor suppressor (such as neuroblastoma), therapeutic development might focus on small molecule activators enhancing IER3 activity, epigenetic modifiers that increase IER3 expression, or drugs that stabilize IER3 protein by inhibiting its degradation . Given IER3's responsiveness to p53 in neuroblastoma, p53-activating compounds like Nutlin and Selinexor represent an indirect approach to enhance IER3 expression in tumors with wild-type p53 .

The identification of cancer-specific IER3 target genes offers another avenue for therapeutic development. In cervical cancer, inhibitors targeting the EGR2-JUN/FOS pathway might counteract IER3's oncogenic effects, while in neuroblastoma, drugs enhancing ADAM19 expression or activity could augment IER3's tumor suppressor functions . The differential subcellular localization of IER3 and IER3-AS1 across cancer types suggests potential for developing drugs that alter their compartmentalization or interaction in a cancer-specific manner .

Development of these targeted approaches would require comprehensive preclinical validation across multiple cancer models to ensure cancer type-specific efficacy and avoid inadvertent promotion of cancer in tissues where IER3 has the opposite function . Companion diagnostics determining IER3's functional state in individual tumors would be essential for patient stratification . With careful development and validation, IER3-targeted therapeutics could represent a sophisticated approach to cancer treatment that exploits the context-dependent nature of this fascinating gene.

Product Science Overview

Gene and Protein Structure

IER3 is located on chromosome 6 in humans and encodes a protein that is approximately 16.9 kDa in size . The protein is primarily expressed in the cytoplasm of cells and has been identified in various tissues, including the liver, kidney, and heart .

Function and Mechanism

The IER3 protein exerts diverse effects on cellular processes. It can function as both an oncogene and a tumor suppressor, depending on the type of cancer . In some cancers, such as bladder cancer, increased expression of IER3 is associated with aggressive progression and poor prognosis . The protein is involved in regulating apoptosis by interacting with key signaling pathways, including the NF-κB and MAPK pathways .

Clinical Significance

IER3 has been studied extensively for its role in cancer biology. In bladder cancer, high expression of IER3 is significantly associated with advanced disease stages and shorter overall survival . This makes IER3 a potential prognostic marker for bladder cancer patients . Additionally, IER3 is being explored as a therapeutic target, with research focusing on modulating its expression to control cancer progression .

Research and Applications

IER3 is widely used in research to study its role in various diseases and cellular processes. Recombinant IER3 protein, expressed in systems like HEK293 cells, is available for research purposes . This recombinant protein is used in various assays, including ELISA, to quantify IER3 levels in different samples .

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 2025 TheBiotek. All Rights Reserved.