ENSA Human

Endosulfine Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Insulin Secretion Modulation

ENSA binds sulfonylurea receptors (ABCC8/SUR1) on pancreatic β-cells, inhibiting K(ATP) channel currents and stimulating insulin release . This positions it as a candidate gene for type 2 diabetes research .

Cell Cycle Regulation

  • S-Phase Control: ENSA depletion extends S-phase duration by reducing replication fork density. This occurs via PP2A-B55–mediated dephosphorylation of Treslin, a replication origin activator .

  • Mitotic Entry: The MASTL-ENSA-PP2A/B55 axis ensures proper mitotic substrate phosphorylation. Dysregulation links ENSA to cancer progression and chemotherapy resistance .

Key Studies

Study FocusMethodology & FindingsImplicationsSource
Cisplatin ResistanceENSA depletion in SCC38 cells increased DNA damage (γ-H2AX), CHK2 phosphorylation, and apoptosis. MASTL-ENSA pathway inhibition sensitizes cancer cells to cisplatin .Therapeutic targeting in oral squamous-cell carcinoma .
Retinal DegenerationBiallelic ENSA LoF variants caused retinitis pigmentosa (RP). Ensa knockout mice showed reduced rod/cone responses .ENSA as a novel RP gene .
S-Phase RegulationENSA knockdown reduced replication fork density by 40% in DNA combing assays, delaying S-phase progression .Mechanistic insight into replication stress .

Clinical and Therapeutic Implications

  • Cancer: MASTL/ENSA overexpression correlates with poor survival in HPV- oral cancer patients. Targeting this axis enhances cisplatin efficacy .

  • Diabetes: ENSA’s interaction with K(ATP) channels makes it a potential biomarker for insulin dysregulation .

  • Retinal Diseases: ENSA mutations disrupt photoreceptor function, implicating it in inherited blindness .

Regulatory Pathways and Inhibitors

  • PP2A/B55 Interaction: Phosphorylated ENSA (pS67) inhibits PP2A-B55, promoting mitotic progression. This pathway is conserved in human platelets and cancer cells .

  • MASTL Inhibitors: Compounds like GKI-1 suppress MASTL kinase activity, indirectly modulating ENSA phosphorylation and PP2A regulation .

Comparative Roles of ENSA and ARPP19

FeatureENSAARPP19
S-Phase RoleRegulates replication fork density No direct S-phase involvement
Mitotic FunctionBinds PP2A-B55 to control mitotic exit Binds PP2A-B55 but lacks S-phase role
Tissue ExpressionUbiquitous (muscle, brain, endocrine) Predominantly neuronal

Product Specs

Introduction
ENSA, an endogenous sulfonylurea receptor ligand, diminishes K(ATP) channel currents by impeding sulfonylurea binding, consequently stimulating insulin secretion. As a member of the conserved cAMP-regulated phosphoprotein (ARPP) family, ENSA, or Endosulfine Alpha, acts as an endogenous ligand for the sulfonylurea receptor, ABCC8/SUR1. This function establishes it as an endogenous regulator of KATP channels, influencing insulin secretion through KATP channel interaction. ENSA's role in type 2 diabetes is currently under investigation. Its expression spans various tissues, including muscle, brain, and endocrine tissues.
Description
Recombinant human ENSA, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 141 amino acids (with residues 1-121 forming the ENSA sequence). It possesses a molecular weight of 15.5 kDa. The ENSA sequence is fused at the N-terminus to a 20 amino acid His-Tag. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The Endosulfine Alpha protein solution is buffered in 20mM Tris (pH 8.0) and supplemented with 1mM DTT and 10% glycerol.
Purity
Purity exceeds 90.0%, as determined by SDS-PAGE analysis.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended periods, storage at -20°C is recommended. To ensure optimal stability during long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA. Minimize freeze-thaw cycles to maintain product integrity.
Synonyms
ARPP-19e, Alpha endosulfine isoform 3, Alpha-endosulfine, ENSA, MGC4319, MGC8394, MGC78563.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSQKQEEENP AEETGEEKQD TQEKEGILPE RAEEAKLKAK YPSLGQKPGG SDFLMKRLQK GQKYFDSGDY NMAKAKMKNK QLPSAGPDKN LVTGDHIPTP QDLPQRKSSL VTSKLAGGQV E.

Q&A

What is ENSA and what is its primary function in human cells?

ENSA functions as a regulatory protein involved in cell cycle control in human cells. Research has identified ENSA's ability to control the length of the S phase of the cell cycle . To investigate ENSA's function in cellular processes, researchers typically employ gene knockdown experiments using shRNA followed by phenotypic assessments.

When studying ENSA's role in cell cycle regulation, recommended methodologies include:

  • Flow cytometry analysis after propidium iodide staining to evaluate cell cycle distribution

  • Immunoblotting for cyclins and CDKs to establish ENSA's position in the cell cycle regulatory network

  • BrdU incorporation assays to specifically measure S-phase progression

  • Time-lapse microscopy to directly observe cell cycle dynamics following ENSA manipulation

How does ENSA expression vary across normal human tissues versus cancer tissues?

ENSA expression follows tissue-specific patterns that differ significantly between normal and cancerous tissues. In TNBC specifically, IHC analysis reveals that ENSA protein levels are much higher in tumor specimens compared to adjacent normal tissues .

To properly assess these differences, researchers should:

  • Perform immunohistochemistry (IHC) on paired tumor and normal tissue samples using validated antibodies

  • Quantify staining intensity using established scoring systems

  • Complement IHC with qRT-PCR and Western blotting for quantitative assessment

  • Use tissue microarrays for high-throughput analysis across multiple samples

  • Compare findings with public databases like TCGA and METABRIC to validate observations

What experimental models are suitable for studying ENSA in human cancer research?

Selecting appropriate experimental models requires careful consideration of research objectives and available resources. Based on published approaches in ENSA research, recommended models include:

Model TypeExamplesAdvantagesLimitations
Cell LinesBT549, MDA-MB-231Easy manipulation, cost-effectiveLimited heterogeneity
Patient-Derived Xenografts (PDX)Mini-PDX modelsMaintains tumor heterogeneityResource intensive, lack immune components
3D OrganoidsPatient-derived organoidsPhysiologically relevant, manipulableComplex establishment
Animal ModelsMouse xenograft modelsIn vivo context, metastasis studiesSpecies differences
Clinical SamplesTissue microarraysDirect clinical relevanceLimited experimental manipulation

For meaningful results, researchers should authenticate cell lines through STR profiling, regularly test for mycoplasma contamination, and validate findings across multiple model systems .

How does ENSA amplification contribute to cancer progression mechanisms?

ENSA amplification promotes cancer progression through multiple interconnected mechanisms, with cholesterol biosynthesis playing a central role. Research reveals that ENSA promotes tumor growth by activating the cholesterol biosynthesis program in TNBC cells .

The mechanistic pathway operates as follows:

  • ENSA amplification leads to increased expression

  • Elevated ENSA promotes STAT3 phosphorylation (p-STAT3)

  • Phosphorylated STAT3 activates SREBP2 transcription

  • SREBP2 upregulates cholesterol biosynthesis genes

  • Enhanced cholesterol metabolism supports tumor growth

To investigate this pathway experimentally:

  • Perform integrated analysis of copy number and gene expression data

  • Use ChIP-seq to confirm STAT3 binding to the SREBP2 promoter

  • Measure cholesterol metabolites using mass spectrometry

  • Conduct rescue experiments with STAT3 inhibitors like Stattic

  • Validate findings in vivo using xenograft models with ENSA knockdown and overexpression

What approaches can resolve contradictory findings in ENSA research literature?

When confronting contradictory findings in research literature, a systematic approach is essential to resolve discrepancies. The WikiContradict benchmark highlights the importance of addressing real-world inter-context conflicts in scientific data .

Methodological approach to resolving contradictions:

  • Systematic comparative analysis:

    • Create a structured comparison table of contradictory studies

    • Identify key differences in methodologies and experimental contexts

    • Assess sample sizes and statistical approaches

  • Critical technical evaluation:

    • Examine antibody specificity for ENSA detection

    • Determine if studies distinguished between ENSA isoforms

    • Evaluate potential off-target effects in genetic manipulation approaches

  • Validation experiments:

    • Design head-to-head comparisons using identical methodologies

    • Perform side-by-side testing of different antibodies

    • Use multiple genetic manipulation approaches (siRNA, shRNA, CRISPR)

  • Meta-analysis:

    • Apply formal meta-analytic techniques to integrate results across studies

    • Weight evidence based on study quality and sample size

    • Test for publication bias that might contribute to apparent contradictions

How does ENSA expression correlate with clinical outcomes, and what statistical approaches should be used to analyze this relationship?

For robust statistical analysis of ENSA as a prognostic marker:

Statistical ApproachApplicationAdvantagesConsiderations
Kaplan-Meier MethodInitial survival analysisVisual representation of differencesRequires defined cutpoint for ENSA expression
Log-rank TestTesting significance of survival differencesRobust non-parametric approachDoes not adjust for covariates
Cox Proportional HazardsMultivariate analysisAdjusts for confounding factorsAssumes proportional hazards
ROC Curve AnalysisDetermining optimal cutpointsObjective determination of thresholdsMust validate in independent cohorts
Machine Learning ModelsIntegrating ENSA with other markersCan capture complex relationshipsRequires large sample sizes

When reporting results:

  • Include both univariate and multivariate analyses

  • Report hazard ratios with confidence intervals

  • Validate findings in multiple independent cohorts

  • Test robustness across different ENSA expression cutpoints

How should researchers design experiments to effectively study ENSA function?

Designing rigorous experiments for ENSA functional studies requires careful planning and adherence to principles of good experimental design. The Experimental Design Assistant (EDA) from NC3Rs can help ensure experimental rigor and reduce animal use while increasing scientific validity .

Key experimental design principles:

  • Include appropriate controls:

    • Negative controls (non-targeting siRNA/shRNA)

    • Positive controls (known regulators of processes being studied)

    • Rescue experiments (re-expressing ENSA to confirm specificity of effects)

  • Apply rigorous randomization and blinding:

    • Randomize allocation of experimental units

    • Blind analysis where possible to prevent bias

    • Document randomization procedures in protocols

  • Ensure adequate sample sizes:

    • Perform power calculations before beginning experiments

    • Consider variability in ENSA expression across samples

    • Plan for sufficient replicates (biological and technical)

  • Account for sex as a biological variable:

    • Include both male and female samples where relevant

    • Consider sex-specific effects in the analysis

    • Report data disaggregated by sex

  • Use validated reagents and protocols:

    • Validate antibody specificity with appropriate controls

    • Authenticate cell lines through STR profiling

    • Use consistent protocols across experiments for comparability

What are the optimal approaches for genetic manipulation of ENSA in research models?

Selecting appropriate genetic manipulation techniques for ENSA studies is crucial for obtaining reliable results. Based on current research practices, the following approaches are recommended:

TechniqueApplicationAdvantagesLimitations
siRNAShort-term knockdownSimple, inexpensive, rapidTransient effect, off-target potential
shRNAStable knockdownLong-term studies, selectableClonal variation, off-target effects
CRISPR/Cas9Complete knockoutClean deletion, stablePotential compensation, difficult rescue
Inducible systemsTemporal controlControlled expression timingLeakiness, complex setup
OverexpressionGain-of-functionMimics amplificationNon-physiological levels

For ENSA manipulation in TNBC models, researchers have successfully employed shRNA knockdown followed by rescue with the most common transcript to demonstrate specificity of effects . This approach has revealed that ENSA downregulation markedly impairs cell growth and colony formation while inducing apoptosis in TNBC cells.

Verification strategies should include:

  • qRT-PCR and Western blotting to confirm knockdown/overexpression

  • Multiple independent shRNA/sgRNA sequences to rule out off-target effects

  • Rescue experiments with wild-type or mutant ENSA

  • Comparison of effects across multiple cell lines with different baseline ENSA expression

How can researchers optimize the translational potential of ENSA findings from bench to bedside?

Maximizing the translational relevance of ENSA research requires strategic planning from initial experimental design through clinical application. Based on successful translational research approaches, consider:

  • Use clinically relevant experimental models:

    • Patient-derived xenografts (PDXs) that maintain tumor heterogeneity

    • Mini-PDX models for higher throughput drug testing

    • Organoids derived from patient samples with varying ENSA expression levels

  • Develop robust biomarker strategies:

    • Standardize IHC protocols for ENSA detection in clinical samples

    • Establish clear scoring criteria with inter-observer validation

    • Define clinically meaningful cutpoints for ENSA positivity

  • Design rationally informed clinical trials:

    • Stratify patients based on ENSA expression/amplification status

    • Include pharmacodynamic markers (e.g., cholesterol metabolites, p-STAT3)

    • Collect pre- and post-treatment biopsies to evaluate mechanism of action

    • Consider window-of-opportunity trials to assess biological effects

  • Explore combination therapies:

    • Test STAT3 inhibitors in ENSA-high vs. ENSA-low models

    • Evaluate synergy between cholesterol pathway inhibitors and ENSA-targeted approaches

    • Investigate immunotherapy combinations based on microenvironment effects

  • Address resistance mechanisms preemptively:

    • Identify potential bypass pathways through unbiased screens

    • Develop strategies to monitor resistance emergence

    • Test rational combinations to prevent resistance development

What bioinformatic approaches are most effective for analyzing ENSA in multi-omics datasets?

Integrating ENSA data across multiple omics platforms requires sophisticated bioinformatic approaches. For comprehensive analysis:

  • Copy number analysis:

    • Use GISTIC or similar algorithms to identify significant copy number alterations

    • Determine frequency of ENSA amplification across cancer types

    • Correlate copy number status with expression levels

  • Transcriptomic integration:

    • Apply differential expression analysis comparing ENSA-high vs. ENSA-low samples

    • Perform gene set enrichment analysis to identify associated pathways

    • Use weighted gene co-expression network analysis (WGCNA) to identify co-regulated gene modules

  • Proteomic correlation:

    • Correlate ENSA protein levels with phosphoproteomic data

    • Identify post-translational modifications affecting ENSA function

    • Map protein-protein interaction networks centered on ENSA

  • Integrated pathway analysis:

    • Apply pathway enrichment analysis to identify biological processes associated with ENSA

    • Use tools like GSEA, Ingenuity Pathway Analysis, or Metascape to reveal cholesterol metabolism and other pathways co-regulated with ENSA

    • Perform causal network analysis to establish directionality in regulatory relationships

  • Visualization techniques:

    • Develop multi-dimensional visualizations (e.g., oncoprints, heatmaps)

    • Create network diagrams showing ENSA interactions

    • Use dimensionality reduction techniques (PCA, t-SNE, UMAP) for data exploration

How should researchers validate putative ENSA-regulated pathways identified through high-throughput analyses?

Validation of high-throughput findings is essential to establish causality in ENSA-regulated pathways. Based on successful approaches in the literature:

  • Prioritize validation targets:

    • Focus on consistent findings across multiple datasets

    • Prioritize genes with known biological relevance to cancer

    • Select targetable pathways with therapeutic potential

  • Apply orthogonal validation techniques:

    • Confirm RNA-seq findings with qRT-PCR

    • Validate protein changes with Western blotting or immunohistochemistry

    • Verify pathway activation with reporter assays

  • Establish causality through perturbation experiments:

    • Manipulate ENSA expression and measure effects on pathway components

    • Perform rescue experiments by modulating downstream effectors

    • Use pharmacological inhibitors to target key nodes in the pathway

  • Confirm mechanistic links:

    • Use ChIP-seq to verify transcription factor binding

    • Perform co-immunoprecipitation to confirm protein-protein interactions

    • Apply CRISPR screening to identify functional dependencies

  • Validate in diverse models:

    • Test across multiple cell lines with varying ENSA status

    • Confirm in animal models and patient-derived samples

    • Analyze correlations in independent patient cohorts

For ENSA-regulated cholesterol biosynthesis, researchers validated this pathway by demonstrating that ENSA knockdown decreased expression of cholesterol biosynthesis enzymes and SREBP2 at both mRNA and protein levels .

What approaches help identify potential therapeutic vulnerabilities in ENSA-amplified cancers?

Identifying therapeutic opportunities in ENSA-amplified cancers requires systematic approaches to discover vulnerabilities. Based on successful strategies:

  • Synthetic lethality screening:

    • Perform genome-wide CRISPR screens in ENSA-high vs. ENSA-low cells

    • Identify genes selectively required in ENSA-amplified contexts

    • Validate top hits with orthogonal approaches (siRNA, small molecules)

  • Drug sensitivity profiling:

    • Test compound libraries across cell line panels with varying ENSA status

    • Focus on FDA-approved drugs for repurposing opportunities

    • Identify synergistic drug combinations through matrix screening

  • Pathway-informed targeting:

    • Exploit ENSA-mediated cholesterol dependency using statins or other inhibitors

    • Target STAT3 signaling with inhibitors like Stattic in ENSA-high tumors

    • Evaluate combination approaches targeting multiple nodes in the pathway

  • Biomarker development:

    • Correlate drug sensitivity with ENSA expression levels

    • Develop assays to identify patients likely to respond to targeted therapies

    • Create companion diagnostics for clinical trial stratification

  • Resistance mechanism anticipation:

    • Study acquired resistance to ENSA pathway inhibitors

    • Identify bypass mechanisms through sequential biopsies or model systems

    • Develop rational combinations to prevent or overcome resistance

Research has demonstrated that ENSA-high TNBC tumors show higher sensitivity to STAT3 inhibitors than those with relatively low ENSA expression, suggesting ENSA expression could serve as a biomarker for effective treatment with STAT3 inhibitors .

What emerging technologies will advance our understanding of ENSA function in human disease?

Emerging technologies promise to deepen our understanding of ENSA biology and its role in disease. Key technological frontiers include:

  • Single-cell multi-omics:

    • Single-cell RNA sequencing to map ENSA expression heterogeneity

    • Spatial transcriptomics to understand ENSA expression in tissue context

    • Single-cell proteomics to capture protein-level regulation

    • Integration of multiple single-cell modalities for comprehensive profiling

  • Advanced genome editing:

    • Base editing for precise ENSA modification without double-strand breaks

    • Prime editing for introducing specific mutations

    • CRISPRi/CRISPRa for reversible modulation of ENSA expression

    • CRISPR screens targeting non-coding regulatory regions of ENSA

  • Protein structure and interaction:

    • AlphaFold-based structural prediction of ENSA protein

    • Cryo-EM to visualize ENSA protein complexes

    • Proximity labeling techniques to map ENSA protein interaction networks

    • Targeted protein degradation approaches as both research tools and potential therapeutics

  • Advanced in vivo models:

    • Humanized mouse models incorporating patient-derived immune components

    • CRISPR-engineered models with endogenous ENSA alterations

    • Patient-derived xenografts with preserved tumor microenvironment

    • Organoid biobanks representing diverse ENSA expression patterns

  • Clinical research tools:

    • Digital pathology with AI analysis of ENSA expression patterns

    • Liquid biopsy approaches to monitor ENSA copy number in circulation

    • Molecular imaging probes for non-invasive assessment of ENSA-related pathways

    • Real-world data integration for clinical outcome correlation

How might ENSA research inform precision medicine approaches across different cancer types?

ENSA research has significant implications for precision oncology beyond TNBC. To maximize translational impact:

  • Expand cancer type profiling:

    • Perform pan-cancer analysis of ENSA amplification and expression

    • Identify additional cancer types with ENSA-driven biology

    • Determine cancer-specific mechanisms of ENSA function

  • Develop predictive biomarkers:

    • Create comprehensive biomarker panels including ENSA status

    • Establish ENSA testing in routine clinical diagnostics where relevant

    • Correlate ENSA with response to specific therapeutic approaches

  • Design ENSA-informed clinical trials:

    • Stratify patients based on ENSA expression/amplification

    • Test targeted therapies in ENSA-defined populations

    • Develop combined biomarker approaches integrating ENSA with other markers

  • Explore rational combinations:

    • Pair STAT3 inhibitors with existing standard-of-care therapies

    • Combine cholesterol pathway inhibitors with immunotherapies

    • Test synthetic lethal approaches in ENSA-amplified contexts

  • Address resistance mechanisms:

    • Monitor for changes in ENSA status during treatment

    • Identify and target bypass pathways

    • Develop sequential or alternating treatment strategies

The sensitivity of ENSA-high tumors to STAT3 inhibitors like Stattic demonstrates the potential for ENSA expression to serve as a predictive biomarker for targeted therapies .

What considerations are important when designing ENSA-focused funding proposals?

Developing successful funding proposals for ENSA research requires careful attention to experimental design, methodology, and broader impact. Based on funding guidelines and advice from grant panels:

  • Address experimental design rigorously:

    • Clearly state research questions and hypotheses

    • Provide detailed methodology with appropriate controls

    • Consider using the Experimental Design Assistant (EDA) to strengthen experimental plans

    • Include power calculations to justify sample sizes

  • Incorporate sex as a biological variable:

    • Plan to use both male and female samples/models where relevant

    • Consider potential sex differences in ENSA biology

    • Include sex as a factor in statistical analyses

    • Justify single-sex studies when appropriate (e.g., breast cancer focus)

  • Demonstrate translational potential:

    • Articulate clear path from basic discoveries to clinical application

    • Include preliminary data supporting clinical relevance

    • Collaborate with clinicians to strengthen translational aspects

    • Consider patient involvement in study design where appropriate

  • Apply the 3Rs principles:

    • Describe strategies to replace, reduce, and refine animal use

    • Justify animal models when alternatives are insufficient

    • Explain how experimental design minimizes animal numbers while maintaining statistical power

    • Consider using the EDA to optimize animal experiments

  • Plan for data sharing and reproducibility:

    • Detail data management and sharing plans

    • Describe validation strategies for key findings

    • Include resources for reproduction/replication studies

    • Outline plans for pre-registration of study protocols where appropriate

Successful proposals will demonstrate rigorous methodology while highlighting the potential impact of ENSA research on both scientific understanding and clinical practice.

Product Science Overview

Gene and Protein Structure

The ENSA gene is located on chromosome 1q21.3 . The protein encoded by this gene plays a crucial role in various cellular processes. It is known to be an endogenous regulator of ATP-sensitive potassium (KATP) channels through its interaction with the beta-cell sulfonylurea receptor (SUR1) . The KATP channel is an octameric complex composed of two structurally unrelated types of subunits: Kir6.2 (KCNJ11) and the sulfonylurea receptor (SUR) .

Function and Mechanism

Endosulfine Alpha is involved in the regulation of insulin secretion. It modulates insulin release by inhibiting the binding of sulfonylureas to their receptor in vitro . This protein also inhibits cloned KATP channel currents, thereby stimulating insulin secretion . Additionally, it has been proposed as a candidate gene for type 2 diabetes due to its role in insulin regulation .

During mitosis, Endosulfine Alpha acts as a protein phosphatase inhibitor, specifically inhibiting protein phosphatase 2A (PP2A) . When phosphorylated at Ser-67, it interacts with PPP2R2D (PR55-delta) and inhibits its activity, which is essential for maintaining high cyclin-B1-CDK1 activity during the M phase .

Clinical Significance

Mutations or dysregulation of the ENSA gene have been associated with various diseases, including Charcot-Marie-Tooth Disease, Axonal, Type 2F, and Primary Ciliary Dyskinesia . The protein’s role in insulin secretion also links it to diabetes research and potential therapeutic targets .

Research and Applications

Recombinant human Endosulfine Alpha has been expressed in various systems for research purposes. It is used to study its role in insulin secretion, cell cycle regulation, and its potential as a therapeutic target for diabetes and other related disorders .

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.