DSTN Human

Destrin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to DSTN Human

DSTN (Destrin), also known as Actin Depolymerizing Factor (ADF), is a 19.5 kDa protein encoded by the DSTN gene in humans . Recombinant DSTN Human (PRO-1137) is produced in E. coli and widely used in laboratory research to study actin dynamics, cytoskeletal remodeling, and cellular processes such as migration and proliferation . DSTN belongs to the ADF/Cofilin family and plays a critical role in regulating actin filament turnover by severing filamentous actin (F-actin) and promoting monomeric actin (G-actin) recycling .

2.1. Molecular Composition

  • Amino Acid Sequence: 173 residues (1-165) with an 8-amino-acid His-tag at the C-terminus .

  • Tertiary Structure: Comprises a central β-sheet flanked by α-helices, resembling gelsolin-family proteins .

2.2. Production and Formulation

  • Expression System: E. coli .

  • Buffer Composition: 20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 1 mM DTT, 10% glycerol .

  • Stability: Stable at 4°C for 2–4 weeks; long-term storage at -20°C with carrier proteins (e.g., 0.1% HSA/BSA) .

Biological Functions and Mechanisms

DSTN regulates actin dynamics through pH-independent severing of F-actin, facilitating cytoskeletal plasticity . Key mechanisms include:

  1. RhoA/SRF Signaling: DSTN expression is upregulated by RhoA and TGF-β pathways, forming a negative feedback loop to modulate serum response factor (SRF)-dependent gene expression .

  2. Smooth Muscle Cell (SMC) Phenotypic Modulation: Depletion of DSTN enhances SMC differentiation markers (e.g., SM α-actin) while inhibiting migration and proliferation .

  3. Phosphorylation Regulation: Activity is suppressed by phosphorylation, which is reversed by slingshot phosphatases .

Table 2: Functional Roles of DSTN in Cellular Processes

ProcessRole of DSTNClinical Relevance
Actin SeveringDepolymerizes F-actin to maintain G-actin poolsAtherosclerosis, restenosis
SMC DifferentiationInhibits SRF/MRTF-A nuclear localizationVascular remodeling
Immune RegulationModulates TLR7/8 signaling in autoimmune diseasesLupus erythematosus

4.1. Disease Associations

  • Atherosclerosis: DSTN downregulation post-vascular injury exacerbates neointima formation .

  • Alzheimer’s Disease (AD): DSTN is implicated in actin dysregulation linked to synaptic dysfunction .

  • Autoimmune Disorders: Dual TLR7/8 inhibition by DSTN-related pathways suppresses IFN-α and proinflammatory cytokines .

4.2. Key Research Findings

  • In Vitro Studies: siRNA-mediated DSTN depletion reduces SMC proliferation by 40% and migration by 60% .

  • In Vivo Models: Carotid artery injury in mice decreases DSTN expression by >50% within 7 days .

Applications in Research

  1. Actin Dynamics Studies: Used to investigate F-actin/G-actin equilibrium in cancer metastasis .

  2. Therapeutic Development: Target for TLR7/8 inhibitors in autoimmune diseases .

  3. Diagnostic Tools: IGS scoring in lupus trials and AD biomarker panels .

Comparative Analysis with Cofilin (CFL1)

FeatureDSTNCFL1
Tissue SpecificityEnriched in SMCs Ubiquitous
Actin BindingPrefers G-actin Binds both G-/F-actin
Disease LinkAtherosclerosis, AD Cancer, neurodegeneration

Future Directions

  • Therapeutic Targeting: Small-molecule inhibitors of DSTN for vascular diseases .

  • Genomic Studies: CRISPR screens to identify DSTN interactors in AD models .

  • Clinical Trials: Phase II trials for E6742 (TLR7/8 inhibitor) in lupus .

Product Specs

Introduction
Actin depolymerizing factor (Destrin/DSTN) is a protein that belongs to the ADF/Cofilin/destrin family. This family is known for its ability to rapidly depolymerize filamentous actin (F-Actin) in a stoichiometric manner. The ADF family plays a crucial role in regulating the turnover rate of actin within living cells. Destrin, specifically, is a small actin-binding protein sensitive to phosphoinositides and capable of depolymerizing actin filaments in laboratory settings. Its function is independent of pH. While present in various epithelial and endothelial cells, Destrin is largely absent in adult mouse heart and skeletal muscle cells. Although sharing a 71% sequence similarity with Cofilin, Destrin exhibits differences in its interaction with Actin.
Description
Recombinant human DSTN, produced in E. coli, is a single polypeptide chain consisting of 173 amino acids (residues 1-165) with a molecular weight of 19.5 kDa. It features an 8 amino acid His-tag fused at the C-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The DSTN solution is provided at a concentration of 0.5mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 0.1M NaCl, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 95% by SDS-PAGE analysis.
Synonyms
Destrin (actin depolymerizing factor), ACTDP, ADF, bA462D18.2 (destrin (actin depolymerizing factor ADF) (ACTDP)), destrin, DSN.
Source
E.coli.
Amino Acid Sequence
MASGVQVADE VCRIFYDMKV RKCSTPEEIK KRKKAVIFCL SADKKCIIVE EGKEILVGDV GVTITDPFKH FVGMLPEKDC RYALYDASFE TKESRKEELM FFLWAPELAP LKSKMIYASS KDAIKKKFQG IKHECQANGP EDLNRACIAE KLGGSLIVAF EGCPVLEHHH HHH

Q&A

What is the molecular function of DSTN in human cellular systems?

DSTN (Destrin) functions primarily as an actin-binding protein responsible for increasing the turnover rate of actin in vivo. It plays a crucial role in precise regulation of cytoskeleton remodeling and actin filament dynamics. As part of the actin-binding protein family, DSTN is involved in numerous cellular processes including cell division, proliferation, and membrane transport .

The methodological approach to studying DSTN's molecular function typically involves:

  • Protein-protein interaction studies to identify binding partners

  • Actin polymerization/depolymerization assays

  • Live-cell imaging of cytoskeletal dynamics

  • Biochemical assays measuring actin turnover rates

What experimental methods are recommended for studying DSTN expression in human tissues?

For comprehensive analysis of DSTN expression, researchers should employ multiple complementary techniques:

  • Transcriptional analysis: qRT-PCR remains the gold standard for quantifying DSTN mRNA expression, as demonstrated in studies of colorectal cancer cell lines where DSTN expression was compared between cancer cells (HT29, HCT116) and normal cells (FHC) .

  • Protein expression analysis: Western blotting and immunohistochemistry (IHC) are effective methods for detecting DSTN protein levels. IHC has been successfully used to correlate DSTN expression with radiation resistance in rectal cancer tissues .

  • Epigenetic regulation: DNA methylation analysis using techniques such as Agena MassARRAY Methylation can reveal epigenetic regulation of DSTN, which has been linked to radiation resistance in cancer .

  • Expression manipulation: siRNA knockdown and overexpression studies help elucidate DSTN function, as demonstrated in experiments with HT29 and HCT116 cell lines .

How should researchers design experiments to study the relationship between DSTN and disease pathology?

A methodologically sound experimental design should include:

  • Hypothesis formulation: Establish clear null (H₀) and alternative (H𝐴) hypotheses. For example, H₀: "There is no difference in DSTN expression between normal and diseased tissues" .

  • Variable identification: Clearly define independent variables (IVs) such as disease state, treatment conditions, or genetic background, and dependent variables (DVs) such as DSTN expression levels, cell behavior, or patient outcomes .

  • Control selection: Include appropriate controls (positive, negative, and experimental) to validate findings and rule out confounding factors.

  • Statistical planning: Determine sample size, statistical tests, and significance thresholds before conducting experiments.

  • Translational approach: Combine in vitro cell culture experiments, in vivo animal models, and when possible, human patient samples to establish clinical relevance.

What are the best practices for purifying DSTN protein for functional studies?

For purification of DSTN protein:

  • Recombinant expression systems: Escherichia coli expression systems have been successfully used to produce recombinant human DSTN protein with >95% purity, suitable for SDS-PAGE and mass spectrometry analysis .

  • Protein sequence considerations: The full-length human DSTN protein (165 amino acids) can be expressed and purified for functional studies .

  • Quality control: Verify protein integrity through SDS-PAGE, Western blot, and activity assays before proceeding with functional studies.

  • Storage conditions: Optimize buffer composition and storage conditions to maintain protein stability and activity.

How does DSTN hypomethylation contribute to radiotherapy resistance, and what methodologies can assess this relationship?

DSTN hypomethylation has been associated with radiotherapy resistance in rectal cancer. To investigate this relationship, researchers should employ a multi-faceted methodology:

  • Methylation analysis: Use Agena MassARRAY Methylation to analyze the methylation status of DSTN in radiation-resistant versus radiation-sensitive tissues .

  • DNA methyltransferase inhibition: Treat cells with decitabine (a DNA methylation inhibitor) to demonstrate the causal relationship between DSTN methylation and expression. This approach has revealed that reducing methylation levels leads to increased DSTN expression in colorectal cancer cells .

  • Functional validation: Perform radiation sensitivity assays (such as CCK-8 assay and colony formation assay) after manipulating DSTN expression to establish causality:

    • siRNA knockdown of DSTN in radiation-resistant cells (e.g., HT29) increases radiation sensitivity

    • Overexpression of DSTN in radiation-sensitive cells (e.g., HCT116) confers radiation resistance

  • In vivo confirmation: Xenograft models can validate in vitro findings, as demonstrated by studies showing DSTN-overexpressing HCT116 cells forming xenografts with worse responses to radiation therapy .

What mechanisms underlie DSTN interaction with cellular signaling pathways, and how can these be experimentally validated?

DSTN has been found to interact with important signaling pathways, particularly the Wnt/β-Catenin pathway. Methodologies to study these interactions include:

  • Protein-protein interaction studies:

    • Co-immunoprecipitation to detect direct binding between DSTN and pathway components (e.g., β-Catenin)

    • Proximity ligation assays to visualize protein interactions in situ

    • FRET/BRET analysis for real-time monitoring of interactions

  • Pathway activity assessment:

    • Reporter gene assays (e.g., TOPFlash) to measure Wnt/β-Catenin pathway activation

    • Western blotting for phosphorylated pathway components

    • Nuclear localization studies of transcription factors

  • Genetic manipulation approaches:

    • CRISPR/Cas9-mediated gene editing to modify binding domains

    • Domain mapping through truncation mutants

    • Point mutations to disrupt specific interactions

  • Computational modeling:

    • Protein-protein docking simulations

    • Molecular dynamics studies of interaction stability

    • Systems biology approaches to model pathway perturbations

How can researchers reconcile contradictory data on DSTN function across different experimental systems?

When faced with contradictory data on DSTN function, researchers should apply these methodological approaches:

  • Systematic comparison of experimental conditions:

    • Cell type differences (epithelial vs. mesenchymal, normal vs. cancer)

    • Culture conditions (2D vs. 3D, media composition)

    • Analytical methods (antibody specificity, detection thresholds)

  • Heterogeneity analysis:

    • Single-cell techniques to identify subpopulations with differential DSTN activity

    • Spatial analysis of DSTN function within tissues

    • Temporal dynamics of DSTN activity during cellular processes

  • Context-dependent function assessment:

    • Stress conditions (radiation, hypoxia, nutrient deprivation)

    • Cell cycle phase-specific analysis

    • Interaction with tissue-specific factors

  • Integration of multi-omics data:

    • Combine transcriptomics, proteomics, and functional data

    • Network analysis to identify condition-specific interaction partners

    • Meta-analysis of published datasets

What are the optimal experimental designs for studying DSTN-mediated radiation resistance in vivo?

For robust in vivo studies of DSTN-mediated radiation resistance:

  • Animal model selection:

    • Xenograft models using cell lines with modified DSTN expression

    • Patient-derived xenografts to maintain tumor heterogeneity

    • Genetically engineered mouse models for systemic effects

  • Radiation protocol optimization:

    • Determine optimal radiation dose based on preliminary cell culture experiments (e.g., 8Gy for HCT116, 10Gy for HT29)

    • Fractionated vs. single-dose radiation to mimic clinical protocols

    • Local vs. whole-body radiation depending on research question

  • Comprehensive outcome measures:

    • Tumor volume measurements over time

    • Survival analysis

    • Histopathological assessment of tumor response

    • Molecular analysis of excised tumors (IHC, RNA-seq, methylation analysis)

  • Translational validation:

    • Correlation with patient samples and clinical outcomes

    • Therapeutic intervention studies targeting DSTN or its regulators

    • Combination therapy approaches (radiation + epigenetic modifiers)

What advanced imaging techniques are most effective for visualizing DSTN-actin interactions?

Several advanced imaging techniques can effectively visualize DSTN-actin interactions:

  • Super-resolution microscopy:

    • STORM/PALM for nanoscale resolution of DSTN-actin structures

    • SIM for improved resolution of dynamic cytoskeletal changes

    • Expansion microscopy to physically enlarge samples for better visualization

  • Live-cell imaging approaches:

    • Fluorescent protein fusions (e.g., DSTN-GFP, RFP-actin)

    • FRAP (Fluorescence Recovery After Photobleaching) to measure turnover kinetics

    • Optogenetic tools for spatial and temporal control of DSTN activity

  • Correlative microscopy:

    • CLEM (Correlative Light and Electron Microscopy) to combine functional and ultrastructural information

    • AFM (Atomic Force Microscopy) combined with fluorescence for structural-functional analysis

  • Quantitative image analysis:

    • Automated tracking of actin filament dynamics

    • Machine learning approaches for pattern recognition in complex cytoskeletal networks

    • 3D reconstruction and time-lapse analysis

How can researchers effectively modulate DSTN expression to study its functional impact?

Multiple approaches can be employed to modulate DSTN expression:

  • Genetic knockdown/knockout techniques:

    • siRNA for transient DSTN knockdown, as successfully used in HT29 cells to study radiation sensitivity

    • shRNA for stable knockdown studies

    • CRISPR/Cas9 for complete knockout or precise genetic modifications

  • Overexpression strategies:

    • Plasmid-based overexpression systems, as used in HCT116 cells to induce radiation resistance

    • Inducible expression systems (e.g., Tet-On/Off) for temporal control

    • Viral vectors for high-efficiency transduction

  • Epigenetic modulation:

    • DNA methyltransferase inhibitors (e.g., decitabine) to reverse DSTN methylation and increase expression

    • Histone deacetylase inhibitors to potentially alter DSTN expression

    • CRISPR-based epigenetic editors for locus-specific modulation

  • Post-translational regulation:

    • Inhibitors or activators of DSTN-modifying enzymes

    • Peptide mimetics to compete for binding sites

    • Targeted protein degradation approaches (PROTACs)

What assays can quantitatively measure DSTN-dependent actin dynamics?

To quantitatively assess DSTN-dependent actin dynamics:

  • Biochemical actin assays:

    • Pyrene-actin polymerization/depolymerization kinetics

    • Sedimentation assays to measure F-actin/G-actin ratios

    • Actin critical concentration determination

  • Cellular actin dynamics:

    • G-actin/F-actin fractionation followed by Western blotting

    • FRAP analysis of fluorescently labeled actin

    • LifeAct or SiR-actin visualization of actin dynamics in living cells

  • Functional consequence assessment:

    • Cell migration assays (wound healing, transwell)

    • Cell division analysis (time to complete mitosis, cytokinesis failures)

    • Membrane trafficking quantification

  • Mechanical property measurements:

    • Atomic force microscopy to measure cell stiffness

    • Optical tweezers for single-filament manipulation

    • Traction force microscopy to quantify cellular forces

How should researchers interpret changes in DSTN methylation patterns in relation to gene expression?

When analyzing DSTN methylation and expression data:

  • Correlation analysis:

    • Directly compare DNA methylation levels (e.g., from Agena MassARRAY Methylation) with mRNA expression (qRT-PCR) and protein levels (Western blot/IHC)

    • Calculate correlation coefficients and statistical significance

    • Perform multivariate analysis to account for confounding factors

  • Causal relationship establishment:

    • Use DNA methyltransferase inhibitors (e.g., decitabine) to demonstrate that demethylation increases DSTN expression

    • Employ methylation-specific recombinant constructs to directly test promoter activity

    • Perform chromatin immunoprecipitation to examine transcription factor binding at methylated vs. unmethylated sites

  • Context-specific analysis:

    • Compare methylation-expression relationships across different cell types and conditions

    • Identify CpG sites with the strongest correlation to expression changes

    • Examine the effect of environmental factors on methylation-expression relationships

  • Clinical relevance assessment:

    • Correlate methylation patterns with patient outcomes such as disease-free survival (DFS)

    • Stratify patient samples by methylation status and compare treatment responses

    • Develop predictive models incorporating methylation data

What statistical approaches are appropriate for analyzing DSTN expression in relation to patient outcomes?

For robust statistical analysis of DSTN expression and patient outcomes:

  • Survival analysis methods:

    • Kaplan-Meier curves to visualize survival differences between high and low DSTN expression groups

    • Cox proportional hazards regression for multivariate analysis

    • Competing risk analysis when multiple outcome events are possible

  • Expression threshold determination:

    • ROC curve analysis to identify optimal cutoff values for "high" vs. "low" expression

    • Quantile-based categorization (tertiles, quartiles)

    • Continuous variable analysis to avoid arbitrary categorization

  • Multivariate modeling:

    • Include relevant clinical covariates (age, stage, treatment)

    • Test for interaction effects between DSTN and treatment modalities

    • Develop and validate predictive nomograms

  • Meta-analytical approaches:

    • Forest plots to visualize effect sizes across studies

    • Random-effects models to account for between-study heterogeneity

    • Funnel plots to assess publication bias

How can researchers effectively analyze contradictory results from different experimental models of DSTN function?

When confronted with contradictory results across experimental models:

  • Systematic comparison framework:

    • Create a comprehensive table documenting experimental conditions, cell types, analytical methods, and outcomes

    • Identify patterns in contradictions (e.g., cell type-specific effects)

    • Weight evidence based on methodological rigor and reproducibility

  • Heterogeneity exploration:

    • Investigate if contradictions reflect true biological heterogeneity

    • Test hypotheses in multiple cell lines simultaneously under identical conditions

    • Examine effects of experimental timing, dosage, and microenvironment

  • Reconciliation strategies:

    • Develop integrated models that accommodate seemingly contradictory results

    • Design experiments specifically to test competing hypotheses

    • Consider context-dependent functions as explanations for contradictions

  • Translational relevance assessment:

    • Determine which model systems best recapitulate human disease

    • Compare with clinical data where available

    • Weigh contradictory findings based on relevance to research question

What experimental design is optimal for studying DSTN's role in radiation resistance?

The optimal experimental design should include:

Table 1: Comprehensive Experimental Design for DSTN Radiation Resistance Studies

Experimental ApproachCell/Tissue ModelsKey AssaysControlsOutcome Measures
DSTN knockdownRadiation-resistant cell lines (e.g., HT29)siRNA transfectionScrambled siRNACell viability (CCK-8), Colony formation, Apoptosis (flow cytometry)
DSTN overexpressionRadiation-sensitive cell lines (e.g., HCT116)Plasmid transfectionEmpty vectorIC50 values, Colony formation, Apoptosis rates
Methylation analysisPatient tissues (radiation-resistant vs. sensitive)Agena MassARRAY MethylationNormal tissuesMethylation levels at specific CpG sites
Protein expressionPatient samples, cell linesIHC, Western blotAntibody controlsExpression levels, subcellular localization
In vivo validationXenograft modelsTumor growth after radiationNon-irradiated controlsTumor volume, histopathology
Pathway analysisCell lines with DSTN modificationWestern blot, Reporter assaysPathway inhibitorsWnt/β-Catenin activity markers

How do DSTN expression levels correlate with radiation sensitivity in cancer models?

Based on available research data:

Table 2: Relationship Between DSTN Status and Radiation Response

DSTN StatusCell LineIC50 ValueColony FormationApoptosis RateTumor Response
Normal expressionHT29 (control)HighHighLowPoor response
KnockdownHT29 + siDSTNDecreasedDecreasedIncreasedImproved response
Normal expressionHCT116 (control)LowLowHighGood response
OverexpressionHCT116 + DSTNIncreasedEnhancedDecreasedWorse response
HypomethylatedRadiation-resistant RC tissues---Poor clinical outcome
HypermethylatedRadiation-sensitive RC tissues---Better clinical outcome

This table summarizes findings that DSTN knockdown in radiation-resistant HT29 cells led to decreased IC50 values, reduced colony formation capacity, and increased apoptosis after radiation. Conversely, DSTN overexpression in radiation-sensitive HCT116 cells resulted in increased radiation tolerance with higher IC50 values, enhanced colony formation, and reduced radiation-induced apoptosis .

What methodologies can detect changes in DSTN-dependent cytoskeletal dynamics?

Table 3: Methodologies for Assessing DSTN-Dependent Cytoskeletal Changes

MethodologyApplicationMeasurementsAdvantagesLimitations
Live-cell imagingActin dynamics visualizationFilament turnover rates, Cytoskeletal reorganizationReal-time analysis, Spatial informationPhototoxicity, Requires specialized equipment
FRAPActin turnover kineticsRecovery half-time, Mobile fractionQuantitative, Single-cell resolutionLimited to fluorescently tagged proteins
G/F-actin fractionationGlobal actin statusG-actin:F-actin ratioBiochemical quantification, Population-level dataNo spatial information, Requires cell lysis
ImmunofluorescenceCytoskeletal architectureStress fiber density, Cortical actin integrityPreserves cellular structure, Compatible with fixed samplesStatic analysis only
Single-filament assaysDirect DSTN-actin interactionSevering rate, Binding affinityMechanistic insights, Controlled conditionsIn vitro system, May not reflect cellular environment
Traction force microscopyFunctional consequencesCell-generated forcesFunctional readout, Links molecular to mechanicalIndirect measure of DSTN activity
Atomic force microscopyCell mechanical propertiesCortical stiffness, ViscoelasticityDirect mechanical measurementsLow throughput, Technical complexity

Product Science Overview

Structure and Function

Destrin is a small, phosphoinositide-sensitive actin-binding protein. It is composed of 165 amino acids and has a molecular weight of approximately 19.5 kDa . The protein is capable of depolymerizing actin filaments in vitro, which is crucial for various cellular processes such as cell motility, division, and maintenance of cell shape .

Expression and Localization

Destrin is found in various epithelial and endothelial cells but is practically absent from adult mouse heart and skeletal muscle cells . It shares 71% sequence homology with cofilin, another member of the same superfamily, but the two proteins differ in their interaction with actin .

Recombinant Production

Recombinant human destrin is typically produced in E. coli and purified using conventional chromatography techniques . The recombinant protein often includes a C-terminal His-tag to facilitate purification and detection . The protein is stored in a buffer containing Tris-HCl, NaCl, glycerol, and DTT to maintain its stability .

Applications

Recombinant destrin is used in various research applications, including studies on actin dynamics, cell motility, and cytoskeletal organization. It is also used to investigate the molecular mechanisms underlying actin filament depolymerization and the role of actin-binding proteins in cellular processes .

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.