EHF Human

Ets Homologous Factor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

EHF regulates epithelial differentiation, morphogenesis, and response to environmental stimuli. Key roles include:

  • Gene Regulation: Directly activates or represses targets like c-Met to influence branching morphogenesis .

  • Epithelial Homeostasis: Maintains epidermal and intestinal integrity; Ehf knockout mice develop skin papillomas, colonic inflammation, and increased tumor susceptibility .

  • Immune Modulation: In airway epithelia, EHF depletion alters 1,145 genes linked to immune response and cell adhesion .

Cancer Biology

EHF exhibits context-dependent roles:

Cancer TypeRole of EHFMechanismReference
Prostate CancerTumor suppressorDownregulated in DU145 cells; promotes apoptosis via caspase-3 activation
Non-Small Cell Lung Cancer (NSCLC)Oncogenic driverOverexpression correlates with poor prognosis; activates AKT/MAPK pathways
Colorectal Cancer (CRC)Suppresses tumorigenesisLoss accelerates Apc-initiated adenomas; interacts with CDX1 to restore differentiation

Inflammatory Pathways

  • Colitis: Ehf knockout mice show heightened susceptibility to DSS-induced colitis and epithelial hyperproliferation .

  • Lung Pathology: EHF depletion in bronchial epithelia dysregulates genes involved in mucus production and immune response .

Mechanistic Insights

  • Transcriptional Networks: EHF intersects with pathways like G2/M checkpoint regulation and epithelial-mesenchymal transition (EMT) .

  • Epigenetic Interactions: Cooperates with CDX1 to reactivate differentiation programs in dedifferentiated CRC cells .

  • Cell-Specific Effects: In pancreatic and prostate cancers, EHF loss correlates with promoter hypermethylation and oncogenic progression .

Applications in Research

  • Recombinant Protein Use: EHF Human (PRO-1436) is utilized for in vitro studies of DNA-binding activity, transcriptional regulation, and signaling pathways .

  • Therapeutic Targeting: Emerging evidence supports EHF as a biomarker for personalized oncology, particularly in CRC and NSCLC .

Ongoing Research and Challenges

  • Pangenome Studies: EHF variants are included in the human pangenome reference to improve genomic diversity representation .

  • Unresolved Questions: Dual oncogenic/tumor-suppressive roles require tissue-specific mechanistic studies.

Product Specs

Introduction
ETS homologous factor (EHF) is a member of the ESE subfamily of Ets transcription factors. Ets factors are a crucial group of transcriptional regulators involved in various biological processes, including hematopoiesis, angiogenesis, organogenesis, oncogenesis, and the establishment of neuronal connections. EHF expression is restricted to a specific subset of epithelial cells, with the highest levels observed in the glandular epithelium of the prostate, pancreas, salivary gland, and trachea. Notably, EHF can activate the c-Met promoter through three high-affinity binding sites, suggesting its role in branching morphogenesis.
Description
Recombinant human EHF, expressed in E. coli, is a single polypeptide chain consisting of 323 amino acids (residues 1-300) and possessing a molecular weight of 37.3 kDa. The protein includes a 23-amino acid His-tag fused to its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The EHF solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 90% as assessed by SDS-PAGE analysis.
Synonyms
ESE3, ESE3B, ESEJ, ETS homologous factor, hEHF, ETS domain-containing transcription factor, Epithelium-specific Ets transcription factor 3, EHF.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMILEGGG VMNLNPGNNL LHQPPAWTDS YSTCNVSSGF FGGQWHEIHP QYWTKYQVWE WLQHLLDTNQ LDANCIPFQE FDINGEHLCS MSLQEFTRAA GTAGQLLYSN LQHLKWNGQC SSDLFQSTHN VIVKTEQTEP SIMNTWKDEN YLYDTNYGST VDLLDSKTFC RAQISMTTTS HLPVAESPDM KKEQDPPAKC HTKKHNPRGT HLWEFIRDIL LNPDKNPGLI KWEDRSEGVF RFLKSEAVAQ LWGKKKNNSS MTYEKLSRAM RYYYKREILE RVDGRRLVYK FGKNARGWRE NEN.

Q&A

What does EHF refer to in human research contexts?

EHF in human research contexts has two primary meanings, both with significant implications for human health and wellbeing:

Alternatively, EHF also refers to ETS Homologous Factor, a transcription factor belonging to the epithelium-specific subfamily of the E26 transformation-specific (ETS) transcription factor family . In this biological context, EHF plays critical roles in epithelial tissue function, gene regulation, and has significant implications for human disease, particularly in cancer research . EHF can function as both a tumor activator and tumor suppressor, making it an important focus for cancer biology research .

What are the foundational research approaches in Ergonomics & Human Factors (EHF)?

Ergonomics & Human Factors research employs several foundational research approaches aimed at understanding and optimizing human-system interactions:

The multidisciplinary approach is central to EHF research, integrating expertise from engineering, psychology, medicine, nursing, environmental health, and design to comprehensively address complex human-system interactions . This approach recognizes that human factors challenges rarely exist within the boundaries of a single discipline.

EHF research typically employs a systems-oriented methodology that views humans as integral components of larger systems rather than isolated elements . This perspective examines how humans interact with technology, environments, and organizational structures within integrated systems.

Research in this field incorporates both field and laboratory studies, with workplace simulations offering controlled environments to study human performance under various conditions while maintaining ecological validity . These simulations allow researchers to manipulate variables and observe responses without the ethical concerns of real-world interventions.

Risk assessment methodologies form another cornerstone of EHF research, identifying, analyzing, and evaluating potential hazards in human-system interactions, particularly in areas like physical ergonomics and occupational safety . These assessments often utilize standardized tools and protocols developed specifically for ergonomic evaluation.

Human performance measurement through various psychophysical, cognitive, and biomechanical metrics allows researchers to quantify how design changes or interventions affect human wellbeing and system outcomes . These measurements may include response times, error rates, musculoskeletal strain, cognitive workload, and user satisfaction.

How does ETS Homologous Factor (EHF) function in human cellular processes?

ETS Homologous Factor (EHF) functions as a transcription factor with complex and context-dependent roles in human cellular processes:

EHF regulates gene expression in epithelial tissues by binding to specific DNA sequences and influencing transcription . This regulatory function is particularly important in maintaining proper epithelial cell identity and function. Research using ChIP-seq has identified 11,326 genomic binding sites for EHF in human bronchial epithelial cells, demonstrating its broad influence on the transcriptome .

In terms of its molecular structure, the EHF gene produces two distinct transcript variants: a long form that includes exon 1 (EHF-LF) and a short form that excludes exon 1 (EHF-SF) . These different variants appear to have distinct functional properties, with the short form specifically capable of abrogating ETS1-mediated activation of the ZEB1 promoter by promoting degradation of ETS1 proteins .

EHF plays a critical role in epithelial-mesenchymal transition (EMT) regulation, acting as an anti-EMT factor by inhibiting the expression of ZEB family proteins . This function has significant implications for cancer progression, as EMT is a key process in metastasis. RNA-seq analysis following EHF depletion identified 1,145 genes that are differentially regulated, with 625 increasing and 520 decreasing in expression, demonstrating EHF's broad regulatory impact .

In cellular signaling pathways, EHF can function as both an activator and repressor of gene expression . This dual functionality allows it to fine-tune complex cellular processes, including differentiation, proliferation, and inflammatory responses. As an activator, EHF overexpression can promote cellular differentiation and proliferation mechanisms, while as a suppressor, it can promote the expression of several miRNAs and block key oncogenic transformation pathways .

What experimental designs are most effective for Ergonomics & Human Factors research?

Effective experimental designs in Ergonomics & Human Factors research must balance scientific rigor with ecological validity. The following approaches have proven particularly valuable:

Mixed-methods designs that combine quantitative measurements with qualitative assessments provide comprehensive insights into human-system interactions . These designs allow researchers to capture both objective performance metrics and subjective experiences, creating a more complete understanding of ergonomic factors. For example, combining biomechanical measurements with participant interviews can reveal both physical strain and perceived comfort in workplace evaluations.

Longitudinal studies are essential for understanding the long-term impacts of ergonomic interventions or occupational exposures . Unlike cross-sectional approaches, longitudinal designs can capture cumulative effects, adaptation processes, and the sustainability of interventions over time. This approach is particularly valuable when studying chronic musculoskeletal disorders or the effectiveness of workplace redesigns.

Simulation-based experimental designs allow researchers to recreate complex work environments under controlled conditions . These simulations can range from physical mockups of workstations to computer-based virtual environments that model complex systems. The key advantage is the ability to systematically manipulate variables while maintaining safety and research control.

Comparative experimental designs testing multiple ergonomic solutions against baseline conditions help identify optimal approaches . These designs typically involve factorial approaches where several factors are manipulated simultaneously to understand both main effects and interactions. This is particularly useful when evaluating alternative interfaces, tools, or workstation configurations.

Participatory research designs that actively involve end-users throughout the research process have shown significant benefits in EHF research . This approach not only improves the ecological validity of findings but also enhances the likelihood of successful implementation. Participatory ergonomics committees that include workers, management, and EHF researchers have demonstrated effectiveness in developing sustainable workplace interventions.

How are genomic and transcriptomic approaches applied to study ETS Homologous Factor (EHF)?

Modern genomic and transcriptomic methodologies have revolutionized the study of ETS Homologous Factor (EHF), providing unprecedented insights into its functions and regulatory networks:

ChIP sequencing (ChIP-seq) has emerged as a powerful technique for identifying EHF binding sites across the genome . This approach involves immunoprecipitation of DNA fragments bound by EHF, followed by high-throughput sequencing. In human bronchial epithelial cells, ChIP-seq revealed 11,326 genomic binding sites for EHF with an irreproducible discovery rate (IDR) < 0.05, demonstrating its extensive regulatory potential . The binding sites can be annotated based on genomic location using tools such as the Cis-regulatory Element Annotation System (CEAS) software to understand their distribution relative to promoters, enhancers, and other functional elements .

RNA sequencing (RNA-seq) following EHF depletion through siRNA techniques allows researchers to identify genes directly and indirectly regulated by EHF . In human bronchial epithelial cells, this approach revealed 1,145 differentially expressed genes (DEGs) after EHF knockdown, with 625 increasing and 520 decreasing in expression . This demonstrates EHF's capacity to both activate and repress gene expression depending on context.

The integration of ChIP-seq and RNA-seq data through computational approaches such as Binding and Expression Target Analysis (BETA) enables identification of direct regulatory targets of EHF . This integrative analysis revealed 425 putative direct targets repressed by EHF and 434 targets activated by EHF, providing a comprehensive map of its regulatory network .

For functional validation studies, targeted gene knockdown using siRNA followed by phenotypic assays has proven effective for understanding EHF's role in cellular processes . For example, wound scratch assays performed after EHF depletion in primary human bronchial epithelial cells demonstrated its impact on wound repair processes, with measurements taken at 0, 3, and 6 hours post-injury to quantify healing rates .

Mutation analysis studies have identified functional variants of EHF and their differential effects on cellular processes . Research has discovered point mutations within the conserved ETS domain of EHF that abolish its original function and potentially cause it to act as a dominant negative, enhancing metastasis in vivo . These findings demonstrate the importance of structural integrity for EHF's tumor-suppressive functions.

What approaches help resolve data contradictions in EHF research?

Resolving data contradictions is a critical challenge in EHF research, particularly given its multidisciplinary nature and complex contexts. Researchers employ several methodological approaches to address these contradictions:

Triangulation of multiple data sources and methodologies helps researchers validate findings and identify the source of contradictions . By examining a research question using different approaches (e.g., laboratory experiments, field observations, computational modeling), researchers can determine whether contradictions reflect methodological artifacts or genuine complexity in the phenomenon under study. For example, apparent contradictions in EHF transcription factor function can often be resolved by considering cell-type specificity and microenvironmental conditions .

Context-specific analysis recognizes that EHF findings may be contradictory because they are highly dependent on the specific context being studied . For the ETS Homologous Factor, its dual role as both tumor activator and suppressor illustrates this principle—the same factor can have opposite effects depending on cellular context, cancer type, or disease stage . Similarly, in Ergonomics & Human Factors research, interventions that work in one workplace setting may fail in another due to organizational or environmental differences .

Multidisciplinary collaborative analysis brings together experts from different fields to interpret contradictory findings through diverse theoretical lenses . Given that EHF research spans disciplines including molecular biology, oncology, ergonomics, psychology, and engineering, contradictions sometimes arise from disciplinary differences in assumptions, methods, or terminology. Collaborative analysis helps reconcile these perspectives and develop more integrated understanding.

Longitudinal and repeated-measures designs can resolve apparent contradictions by revealing temporal dynamics or individual differences that cross-sectional studies miss . For example, the wound healing effects of EHF observed at different time points (0, 3, and 6 hours) demonstrate the importance of temporal resolution in understanding biological processes .

How do mutations in the ETS Homologous Factor (EHF) gene affect its function in human disease progression?

Mutations in the ETS Homologous Factor (EHF) gene have profound and context-dependent effects on its function in human disease progression, particularly in cancer:

Point mutations within the conserved ETS domain of EHF can fundamentally alter its functionality, abolishing its original tumor-suppressive properties while potentially causing it to act as a dominant negative factor . These mutations appear to enhance metastatic potential in vivo, suggesting that the structural integrity of the ETS domain is critical for EHF's ability to regulate epithelial-mesenchymal transition (EMT) and suppress cancer progression . The molecular mechanism underlying this transformation involves disruption of EHF's ability to inhibit ETS1-mediated activation of the ZEB1 promoter .

The differential expression of EHF transcript variants (EHF-LF and EHF-SF) also significantly impacts disease progression . Research indicates that only the short form variant (EHF-SF) that excludes exon 1 is capable of abrogating ETS1-mediated activation of the ZEB1 promoter by promoting degradation of ETS1 proteins, thereby inhibiting EMT phenotypes in cancer cells . This suggests that alternative splicing of EHF transcripts represents an important regulatory mechanism that can influence disease outcomes.

In prostate cancer, significant downregulation of EHF has been observed in DU145 human prostate cancer cells compared to human primary prostate epithelial cells (HPEC) . Ingenuity Pathway Analysis (IPA) revealed that EHF showed the highest level of down-regulation (expression fold change) among all genes in the dataset, highlighting its potential importance as a tumor suppressor in this context . Upstream regulator analysis demonstrated that EHF leads to the predicted activation of CCBE1 and the predicted inhibition of FOXN1 and other proteins believed to produce anti-tumor effects .

Beyond cancer, EHF plays critical roles in epithelial dysfunction associated with respiratory diseases, including cystic fibrosis . EHF is positioned as a key member of the transcription factor network that regulates gene expression in the airway epithelium in response to endogenous and exogenous stimuli . Its location adjacent to an intergenic region containing enhancer-like features suggests it may function as a modifier of lung disease in cystic fibrosis patients .

What are the methodological challenges in quantifying human factors in complex systems?

Quantifying human factors in complex systems presents several methodological challenges that researchers must navigate:

The multidimensional nature of human-system interactions requires comprehensive measurement approaches that capture physical, cognitive, psychological, and social dimensions simultaneously . Traditional metrics often focus on isolated aspects (e.g., physical ergonomics or cognitive workload), failing to capture the integrated nature of human performance in complex systems. Development of composite measures that meaningfully combine these dimensions remains challenging, as relationships between dimensions are often non-linear and context-dependent.

Ecological validity versus experimental control represents a fundamental tension in human factors research . Laboratory studies offer precise control of variables but may not generalize to real-world settings, while field studies provide realism but introduce numerous confounding variables. Researchers increasingly employ simulation-based approaches that attempt to balance these concerns, but determining the appropriate level of fidelity remains challenging and is often contingent on the specific research questions being addressed.

Individual differences in human capabilities, preferences, and behaviors introduce significant variability in human factors data . These differences can manifest as large standard deviations in performance metrics, making it difficult to detect meaningful effects of system interventions or designs. Advanced statistical approaches including mixed-effects modeling, which accounts for both fixed effects (experimental manipulations) and random effects (individual differences), have proven valuable in addressing this challenge.

The dynamic and adaptive nature of human behavior complicates measurement and analysis . Humans naturally adapt to system changes over time, potentially masking initial effects or creating unexpected compensatory behaviors that traditional before-after measurements fail to capture. Longitudinal designs with appropriate time-series analysis techniques are necessary to understand these adaptation processes.

Integration of qualitative and quantitative data presents analytical challenges when seeking to develop comprehensive understanding of human factors . While mixed-methods approaches are increasingly recognized as valuable, standard procedures for integrating these different data types remain underdeveloped. Methods such as qualitative comparative analysis and mixed-methods synthesis frameworks offer promising approaches but require further refinement for complex systems applications.

How can researchers assess the impact of ETS Homologous Factor (EHF) on epithelial cell differentiation?

Assessing the impact of ETS Homologous Factor (EHF) on epithelial cell differentiation requires sophisticated experimental approaches that span molecular, cellular, and tissue-level analyses:

Gene expression profiling through RNA-seq following EHF manipulation represents a fundamental approach to understanding its role in differentiation . By comparing the transcriptome of cells with normal, depleted, or overexpressed EHF, researchers can identify differentiation-associated genes under EHF regulation. In human bronchial epithelial cells, RNA-seq after EHF depletion identified numerous differentially expressed genes including key transcription factors involved in epithelial differentiation such as SPDEF (SAM pointed domain-containing ETS transcription factor) .

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) enables identification of genomic regions directly bound by EHF, providing insights into its immediate regulatory targets in differentiation pathways . By correlating binding patterns with differentiation states, researchers can map the direct regulatory network through which EHF influences epithelial development. Analysis of binding motifs enriched in EHF-bound regions can also reveal co-regulatory relationships with other transcription factors involved in differentiation.

Three-dimensional organoid culture systems derived from primary epithelial cells offer valuable models for studying EHF's role in tissue-level differentiation processes . These systems recapitulate aspects of epithelial architecture and differentiation not observable in standard two-dimensional cultures. By manipulating EHF levels in these organoids through genetic approaches (CRISPR-Cas9, shRNA, overexpression), researchers can assess effects on cellular organization, polarization, and specialized function acquisition.

Time-course analyses of differentiation following EHF manipulation help establish causality and elucidate the temporal dynamics of its effects . This approach involves monitoring cellular changes at multiple timepoints after modifying EHF expression, using markers of different differentiation stages to determine whether EHF acts early as a lineage determination factor or later as a maturation regulator. Techniques like single-cell RNA-seq are particularly valuable for capturing heterogeneity in differentiation trajectories.

Comparative studies across different epithelial tissues can reveal context-dependent functions of EHF in differentiation . By studying EHF's role in multiple epithelial contexts (e.g., airway, intestinal, prostate), researchers can identify both shared and tissue-specific aspects of its function. This approach has revealed that while EHF generally promotes epithelial identity, its specific regulatory targets and phenotypic effects vary substantially across tissues.

What systems thinking frameworks are advancing Ergonomics & Human Factors research?

Systems thinking frameworks are transforming Ergonomics & Human Factors research by providing more holistic and dynamic perspectives on human-system interactions:

Sociotechnical systems approaches recognize that human work occurs within complex networks of social and technical elements that continuously interact and co-evolve . This framework moves beyond traditional linear cause-effect models to understand how system properties emerge from these interactions. In EHF research, this perspective helps explain why interventions that address isolated components often fail to produce expected improvements when implemented in real-world contexts.

Resilience engineering frameworks focus on how systems maintain performance under varying conditions, shifting emphasis from error prevention to capability enhancement . Rather than viewing human error as a problem to be eliminated, this approach examines how humans contribute to system resilience through adaptation, learning, and flexibility. EHF researchers applying this framework study how systems succeed under normal and challenging conditions, rather than focusing exclusively on failure analysis.

Complex adaptive systems theory provides mathematical and conceptual tools for understanding non-linear dynamics, emergence, and self-organization in human-system interactions . This framework helps EHF researchers move beyond reductionist approaches to address complexity directly. Methodologies derived from this perspective include agent-based modeling, network analysis, and dynamical systems approaches that can capture patterns in system behavior that traditional statistical methods might miss.

Human-centered design thinking integrates user needs, technological possibilities, and organizational requirements throughout the design process . This iterative approach emphasizes empathy, ideation, prototyping, and testing with actual users. When applied within EHF research, design thinking helps ensure that technical solutions address genuine human needs and constraints rather than creating new ergonomic challenges.

Ecological interface design frameworks focus on making system constraints and relationships perceptually evident to users, supporting effective decision-making in complex environments . This approach, derived from cognitive systems engineering, aims to create interfaces that reveal the fundamental structure of the work domain rather than prescribing specific procedures. EHF researchers have applied this framework to domains ranging from healthcare to process control, developing interfaces that support flexible human performance under varying conditions.

What new methodologies are advancing understanding of ETS Homologous Factor (EHF) in human disease?

Cutting-edge methodologies are rapidly advancing our understanding of ETS Homologous Factor (EHF) in human disease contexts:

CRISPR-Cas9 genome editing technologies enable precise modification of EHF and its regulatory elements in relevant cell types and model organisms . This approach allows researchers to create isogenic cell lines that differ only in specific EHF mutations or expression levels, providing cleaner experimental systems than traditional knockdown approaches. CRISPR screening methods can also identify synthetic lethal interactions with EHF alterations, revealing potential therapeutic vulnerabilities in cancers with aberrant EHF function.

Single-cell transcriptomics and epigenomics techniques reveal heterogeneity in EHF expression and function across individual cells within tissues . These approaches have demonstrated that EHF's regulatory impact varies substantially between cell subpopulations, even within the same tissue, helping explain seemingly contradictory findings from bulk tissue analyses. By correlating single-cell EHF expression patterns with cell states and phenotypes, researchers can map its role in maintaining epithelial heterogeneity and differentiation hierarchies.

Multi-omics integration approaches combine data from genomics, transcriptomics, proteomics, and metabolomics to develop comprehensive models of EHF's role in cellular networks . These integrative analyses have revealed that EHF functions as a hub connecting multiple regulatory pathways rather than operating in isolation. For example, in prostate cancer, Ingenuity Pathway Analysis identified connections between EHF downregulation and multiple downstream effectors including CCBE1 and FOXN1 .

Patient-derived organoids and xenografts provide clinically relevant models for studying EHF function in human disease contexts . These models retain the genetic and phenotypic heterogeneity of patient tumors, enabling more translational studies than traditional cell lines. By manipulating EHF in these systems, researchers can directly assess its impact on disease progression and treatment response in patient-specific contexts, supporting precision medicine approaches.

Structural biology and protein interaction mapping techniques are revealing the molecular mechanisms underlying EHF's context-dependent functions . X-ray crystallography and cryo-electron microscopy studies of EHF in complex with DNA and protein partners provide atomic-level insights into how mutations disrupt its function. Techniques like BioID and RIME (Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins) are identifying the extended protein interaction network of EHF, revealing how it recruits different cofactors in different cellular contexts.

What interdisciplinary approaches are enhancing EHF research outcomes?

Interdisciplinary approaches are increasingly vital for addressing complex questions in EHF research, yielding insights that would be inaccessible through single-discipline perspectives:

Integration of computational biology with experimental approaches has dramatically accelerated EHF research . Machine learning algorithms applied to large genomic datasets can identify patterns in EHF binding and regulatory activity that might be missed by traditional analysis methods. For example, analysis of EHF ChIP-seq data using computational approaches has revealed previously unrecognized binding motifs and co-regulatory relationships . These computational predictions then guide focused experimental validation, creating an efficient research cycle.

Combining molecular biology with systems physiology provides a more complete understanding of how EHF influences human health and disease . While molecular studies reveal mechanisms of EHF function at the cellular level, physiological approaches examine how these mechanisms translate to tissue, organ, and organism-level phenotypes. In respiratory research, this integration has shown how EHF's regulation of epithelial gene expression affects broader airway function and response to environmental challenges .

Ergonomics and biomedical engineering collaborations are yielding improved tools and interfaces for researchers studying both forms of EHF . Human-centered design principles from ergonomics inform the development of laboratory equipment and software interfaces for biomedical researchers, while engineering approaches to data visualization help make complex EHF datasets more interpretable. These collaborations enhance research efficiency and reduce technical barriers to advanced studies.

Social science methodologies combined with biological approaches provide context for understanding EHF research implications . Qualitative research methods including interviews, focus groups, and ethnographic observation help identify how EHF research findings might translate to real-world settings and what barriers might exist to implementation. This integration is particularly valuable for translating basic EHF findings into clinical or occupational interventions.

Cross-species comparative approaches reveal evolutionary conservation and divergence in EHF function . By studying EHF homologs across model organisms from fruit flies to mice, researchers can identify core conserved functions while understanding human-specific aspects. These comparative studies have shown that while the DNA-binding domain of EHF is highly conserved, regulatory mechanisms controlling its expression and activity show significant species-specific variation, with implications for translating findings from model organisms to humans.

How can ETS Homologous Factor (EHF) research inform cancer treatment strategies?

ETS Homologous Factor (EHF) research has significant potential to inform innovative cancer treatment strategies through several mechanistic pathways:

Targeted therapeutic approaches could exploit EHF's tumor suppressor function in epithelial cancers where it is downregulated . For prostate cancer, where EHF shows significant downregulation in cancer cells compared to normal epithelial cells, strategies to restore EHF expression or activity might inhibit tumor growth and metastasis . Potential approaches include epigenetic modifiers that reverse silencing of the EHF gene or small molecules that mimic EHF's inhibitory effect on oncogenic pathways.

Personalized treatment strategies could be developed based on EHF mutation status . Since point mutations within the conserved ETS domain of EHF can transform it from a tumor suppressor into a potential dominant negative that enhances metastasis, identifying these mutations in patient samples could help stratify patients for different treatment approaches . Patients with intact EHF might benefit from therapies that enhance its activity, while those with dominant negative mutations might require approaches that bypass or counteract these mutant proteins.

Inhibition of the epithelial-mesenchymal transition (EMT) represents a promising strategy informed by EHF research . Since EHF acts as an anti-EMT factor by inhibiting the expression of ZEB family proteins, therapies that either restore EHF function or directly target the downstream EMT pathways it regulates could reduce cancer invasiveness and metastasis . This approach might be particularly valuable in cancers where EMT drives progression and treatment resistance.

Combination therapies targeting EHF-regulated pathways could enhance treatment efficacy . Research has identified multiple genes and pathways regulated by EHF, including CCBE1, FOXN1, and various miRNAs involved in oncogenic transformation . By targeting these downstream effectors in combination, it may be possible to synthetically replicate the tumor-suppressive effects of EHF even in contexts where directly restoring EHF function is challenging.

Biomarker development based on EHF expression, mutation status, or activity signatures could improve cancer diagnosis and prognosis . The distinct regulatory signatures associated with normal versus aberrant EHF function might serve as molecular fingerprints to identify cancer subtypes, predict disease progression, or monitor treatment response. For example, the strong downregulation of EHF in DU145 prostate cancer cells suggests it could serve as a valuable diagnostic marker for certain prostate cancer subtypes .

What are effective strategies for implementing Ergonomics & Human Factors research findings?

Implementing Ergonomics & Human Factors research findings effectively requires strategic approaches that bridge the gap between research and practice:

Participatory implementation models that engage stakeholders throughout the process significantly improve adoption and sustainability . These approaches recognize that successful implementation depends not only on technical merit but also on organizational acceptance and user buy-in. By involving end-users, management, and other stakeholders from problem identification through solution development and implementation, EHF researchers can ensure interventions address actual needs and fit within existing work systems.

Staged implementation with iterative refinement allows for adaptation to specific contexts . Rather than attempting comprehensive changes all at once, this approach starts with pilot implementations that can be evaluated and refined before broader rollout. This strategy recognizes that EHF solutions often require tailoring to specific organizational contexts and provides opportunities for learning and adjustment throughout the implementation process.

Economic analysis frameworks help demonstrate the business case for EHF interventions . These approaches quantify both direct costs (equipment, training, etc.) and benefits (productivity improvements, reduced injuries, lower absenteeism) to calculate return on investment. By speaking the language of organizational decision-makers, these analyses can help secure resources and commitment for implementing EHF research findings.

Knowledge translation tools that convert research findings into practical guidance facilitate implementation by non-specialists . These tools might include decision support systems, implementation checklists, or visual design guides that distill complex research into actionable recommendations. Effective knowledge translation recognizes the different information needs and expertise levels of various stakeholders and provides appropriate resources for each.

Systems integration approaches ensure that EHF interventions work within existing organizational structures and processes . This strategy recognizes that even technically excellent solutions will fail if they conflict with other system elements or create unintended consequences. By mapping system relationships and constraints before implementation, researchers can anticipate integration challenges and design solutions that complement rather than disrupt existing practices.

How can researchers effectively translate complex EHF findings to different stakeholders?

Translating complex EHF research findings to different stakeholders requires tailored communication strategies that address varying knowledge needs, interests, and decision contexts:

Visual communication methods effectively convey complex relationships in EHF data to non-specialists . Interactive data visualizations, infographics, and conceptual models can make abstract relationships concrete and accessible. For the ETS Homologous Factor, pathway diagrams showing its regulatory relationships help clinicians understand its role in disease without requiring detailed molecular knowledge . For Ergonomics & Human Factors, visual representations of system relationships help management visualize how interventions might impact organizational performance .

Narrative case studies that contextualize findings within real-world scenarios increase relevance for practitioners . These narratives connect abstract research to concrete situations that stakeholders recognize from their own experience. For example, describing how specific EHF interventions improved safety outcomes in similar organizations makes research more compelling for safety managers than statistical summaries alone.

Tiered information structures accommodate different stakeholder needs and expertise levels . This approach provides executive summaries for decision-makers, practical guidelines for implementers, and detailed methodological information for specialists. Each level contains appropriate information without overwhelming or under-informing the intended audience. For complex molecular findings about ETS Homologous Factor, this might mean providing clinical implications for physicians while offering mechanistic details for researchers .

Collaborative knowledge co-creation involves stakeholders in interpreting and applying research findings . Rather than one-way knowledge transfer from researchers to users, this approach engages stakeholders in making sense of findings within their specific contexts. Workshops where researchers and practitioners jointly explore implications of EHF research for specific settings have proven particularly effective for translating complex findings into actionable insights.

Quantitative impact metrics tailored to stakeholder priorities demonstrate research relevance . For clinical stakeholders interested in ETS Homologous Factor research, metrics might include potential improvements in diagnostic accuracy or treatment response rates . For organizational stakeholders considering Ergonomics & Human Factors interventions, metrics might include expected reductions in workplace injuries or improvements in productivity . These quantitative translations help stakeholders evaluate the practical significance of research findings.

Product Science Overview

Structure and Function

EHF, like other members of the ETS family, contains a highly conserved DNA-binding domain known as the ETS domain. This domain is characterized by a winged helix-turn-helix structure that binds to DNA sequences with a central GGA(A/T) motif . The ETS domain is not only involved in DNA binding but also in protein-protein interactions, which are essential for the regulation of various cellular processes .

The primary function of EHF is to regulate the expression of genes involved in epithelial cell differentiation, proliferation, and homeostasis. It is particularly important in maintaining the integrity and function of epithelial tissues, such as the skin and the lining of the gastrointestinal tract .

Role in Development and Disease

EHF is essential for the post-natal homeostasis of the epidermis and colonic epithelium. Studies have shown that EHF functions as a tumor suppressor in the colon, where its deletion can enhance the development of adenomas, a type of benign tumor . In addition, EHF is highly expressed in the colonic epithelium, and its absence increases susceptibility to colitis, an inflammatory condition of the colon .

In the liver, EHF is expressed in cholangiocytes and hepatocytes, where it plays a role in liver fibrosis, a condition characterized by the excessive accumulation of extracellular matrix proteins . The expression of EHF in other cell types is relatively low, indicating its specific role in epithelial tissues .

Recombinant EHF

Recombinant EHF is produced using recombinant DNA technology, which involves inserting the EHF gene into a suitable expression system, such as bacteria or mammalian cells. This allows for the production of large quantities of EHF protein, which can be used for various research and therapeutic purposes.

Recombinant EHF is valuable in studying the molecular mechanisms underlying epithelial cell function and the role of EHF in disease. It can also be used in drug development to identify potential therapeutic targets for conditions such as cancer and inflammatory diseases.

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.