STRAP Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
MAP activator with WD repeats antibody; MAWD antibody; PTWD antibody; Serine-threonine kinase receptor-associated protein antibody; serine/threonine kinase receptor associated protein antibody; strap antibody; STRAP_HUMAN antibody; UNR-interacting protein antibody; UNRIP antibody; WD 40 repeat protein PT WD antibody; WD-40 repeat protein PT-WD antibody
Target Names
STRAP
Uniprot No.

Target Background

Function
The SMN complex plays a critical role in pre-mRNA splicing by catalyzing the assembly of small nuclear ribonucleoproteins (snRNPs), which are the building blocks of the spliceosome. Most spliceosomal snRNPs contain a common set of Sm proteins (SNRPB, SNRPD1, SNRPD2, SNRPD3, SNRPE, SNRPF, and SNRPG) that assemble into a heptameric protein ring on the Sm site of the small nuclear RNA, forming the core snRNP (Sm core). In the cytosol, the Sm proteins SNRPD1, SNRPD2, SNRPE, SNRPF, and SNRPG are held in an inactive 6S pICln-Sm complex by the chaperone CLNS1A, which controls the assembly of the core snRNP. To assemble core snRNPs, the SMN complex accepts the trapped 5Sm proteins from CLNS1A, forming an intermediate complex. Binding of snRNA within 5Sm triggers the removal of the SMN complex, allowing SNRPD3 and SNRPB to bind and complete the assembly of the core snRNP. STRAP plays a role in the cellular distribution of the SMN complex. It negatively regulates TGF-beta signaling but positively regulates the PDPK1 kinase activity by enhancing its autophosphorylation and significantly reducing the association of PDPK1 with the 14-3-3 protein.
Gene References Into Functions
  • These findings suggest that STRAP promotes invasion and metastasis of colorectal cancer, partly through inhibiting ubiquitin-dependent degradation of beta-catenin and promoting Wnt/beta-catenin signaling. PMID: 26910283
  • The research revealed a novel STRAP-NOTCH1-HES1 molecular axis as a CSC regulator in colorectal cancer. PMID: 28827371
  • MAWD and MAWBP were downregulated and associated with the differentiation grade in GC tissues. PMID: 26373288
  • STRAP inhibits the transactivation function of Sp1 either by directly blocking its DNA binding domain or by destabilizing Sp1 protein through the ubiquitin-proteasome pathway in the cell cycle G1 phase. PMID: 25483064
  • Strap augments the apoptotic effects of mitochondrial p53. PMID: 25168243
  • Coexpression of MAWBP and MAWD inhibited epithelial mesenchymal transformation, and EMT-aided malignant cell progression was suppressed in gastric cancer. PMID: 23687415
  • Data indicate serine-threonine kinase receptor-associated protein (STRAP) as a novel regulator of the coordinated translation of collagen alpha1(I) and alpha2(I) mRNAs. PMID: 23918805
  • Both the TPR and OB-fold domains localize to the chromatin of p53 target genes and exhibit intrinsic regulatory activity necessary for the Strap-dependent p53 response. PMID: 22362889
  • STRAP protein decreases ubiquitination of the intracellular domain of Notch3 and may help to stabilize it. PMID: 21502811
  • B-MYB acts as a positive regulator of STRAP. PMID: 21148321
  • Nucleotide pulldown identified STRAP (serine/threonine kinase receptor-associated protein) as a further protein recruited to the MMP28 promoter and acting functionally with Sp1. PMID: 20144149
  • STRAP can act as a negative regulator of ASK1. PMID: 19880523
  • Unrip functions in the pathway of small nuclear ribonucleoproteins biogenesis. PMID: 15848170
  • Biochemical evidence for the existence of another, yet atypical, SMN complex component, termed unr-interacting protein (unrip). PMID: 16159890
  • STRAP acts as an intermediate signaling molecule linking between the phosphatidylinositol 3-kinase/PDK1 and the TGF-beta signaling pathways. PMID: 16251192
  • Gemin8 has an essential role in the proper structural organization of the SMN complex and the involvement of the heteromeric subunit containing Gemin6, Gemin7, Gemin8, and Unrip in the recruitment of Sm proteins to the snRNP assembly pathway. PMID: 17023415
  • The direct interaction of NM23-H1 and STRAP is important for the regulation of TGF-beta-dependent biological activity as well as NM23-H1 activity. PMID: 17314099
  • These results indicate that upregulation of STRAP might play a role in tumor development as an early event for colorectal cancers. PMID: 17316412
  • An interplay between PPM1G and unrip determines compartment-specific phosphorylation patterns, localization, and function of the SMN complex. PMID: 17984321
  • Strap regulation reflects the coordinated interplay between different DNA damage-activated protein kinases, ATM and Chk2 (Checkpoint kinase 2), where phosphorylation by each kinase provides a distinct functional consequence on the activity of Strap. PMID: 18833288

Show More

Hide All

Database Links

HGNC: 30796

OMIM: 605986

KEGG: hsa:11171

STRING: 9606.ENSP00000392270

UniGene: Hs.743971

Protein Families
WD repeat STRAP family
Subcellular Location
Cytoplasm. Nucleus. Note=Localized predominantly in the cytoplasm but also found in the nucleus.

Q&A

What is STRAP protein and why is it significant in research?

STRAP (Serine/threonine Kinase Receptor Associated Protein) is a biologically significant protein involved in multiple cellular pathways and signaling mechanisms. The protein has gained research attention due to its involvement in various cellular processes including signal transduction, protein-protein interactions, and its potential role in disease mechanisms. The full-length STRAP protein (covering amino acids 1-350) is commonly used as an immunogen for antibody production, with various epitopes from different regions (N-terminal, C-terminal, and internal sequences) targeted for specific research applications . Understanding STRAP's functional role requires reliable antibodies that can specifically detect this protein in various experimental contexts, making STRAP antibodies essential tools in molecular and cellular biology research.

What are the primary applications of STRAP antibodies in research?

STRAP antibodies are employed in multiple research applications, with the most common being Western Blotting (WB), Immunohistochemistry (IHC), and Flow Cytometry (FACS) . Each application requires specific antibody characteristics and validation steps to ensure reliable results. For Western blotting, STRAP antibodies can detect the native protein in cell or tissue lysates, allowing researchers to quantify expression levels and study post-translational modifications. In immunohistochemistry, these antibodies enable visualization of STRAP protein localization within tissues and cells, providing insights into its spatial distribution. Flow cytometry applications allow for the analysis of STRAP protein expression at the single-cell level, particularly valuable for heterogeneous cell populations. Some STRAP antibodies are also suitable for ELISA applications, though this varies by specific antibody clone .

How do I select the most appropriate STRAP antibody for my research?

Selecting the appropriate STRAP antibody requires consideration of several factors:

  • Research application: Determine whether the antibody has been validated for your specific application (WB, IHC, FACS, etc.) as antibodies may perform differently across applications .

  • Species reactivity: Verify that the antibody reacts with your species of interest. Some STRAP antibodies show broad cross-reactivity across species (Human, Mouse, Rat, Dog, Cow, Guinea Pig, Horse, Rabbit, Zebrafish, Bat, Monkey), while others are more limited in their reactivity .

  • Epitope recognition: Consider which region of the STRAP protein you need to target. Antibodies targeting different regions (N-terminal, C-terminal, or specific amino acid ranges) may provide different information, especially if you are studying protein isoforms or truncated variants .

  • Clonality: Polyclonal antibodies often provide higher sensitivity due to recognition of multiple epitopes, while monoclonal antibodies offer higher specificity to a single epitope .

  • Validation data: Examine available validation data, including Western blot images showing the expected band size, positive and negative controls, and cross-reactivity assessments .

The combination of these factors should guide your selection to ensure optimal experimental outcomes.

What are the recommended validation methods for STRAP antibodies in Western blotting?

Validating STRAP antibodies for Western blotting requires a systematic approach to ensure specificity, reproducibility, and reliability of results. The following methods are recommended based on current best practices:

These validation steps should be documented and included in research publications to enhance reproducibility across the scientific community.

How can I determine if multiple bands in my Western blot indicate non-specific binding or biologically relevant variants?

Multiple bands in Western blot analysis using STRAP antibodies do not necessarily indicate non-specific binding. Several biological explanations should be considered:

  • Post-translational modifications: STRAP protein may undergo modifications such as phosphorylation, glycosylation, or ubiquitination, resulting in mobility shifts .

  • Splice variants: Different isoforms of STRAP protein may be present in your sample, each with slightly different molecular weights .

  • Protein degradation: Sample preparation conditions might cause protein degradation, resulting in fragments detected by the antibody .

  • Cross-reactivity with related proteins: The antibody might recognize epitopes present in proteins with structural similarity to STRAP .

To determine the nature of additional bands:

  • Compare with literature data: Review published Western blots using STRAP antibodies to identify commonly observed patterns.

  • Use different antibodies: Test multiple antibodies targeting different epitopes of STRAP. If the same pattern appears, it supports biological relevance rather than non-specificity .

  • Conduct domain-specific analysis: Use antibodies targeting specific domains (N-terminal, C-terminal) to identify which portion of the protein is present in each band .

  • Perform protein knockdown: Reduce STRAP expression through siRNA or CRISPR-Cas9 and observe which bands diminish, confirming their relationship to STRAP .

  • Mass spectrometry validation: Excise the bands of interest and analyze by mass spectrometry to definitively identify their protein content .

Through systematic analysis, you can differentiate between non-specific binding and biologically significant protein variants.

What controls should I include when using STRAP antibodies in immunohistochemistry?

When using STRAP antibodies for immunohistochemistry (IHC), the following controls should be implemented to ensure valid and interpretable results:

Control TypePurposeImplementationPriority
Known source tissuePositive controlInclude tissue known to express STRAP at detectable levelsHigh
Tissue from null animalNegative controlUse tissue from STRAP knockout modelsHigh
No primary antibodyNegative controlOmit STRAP antibody but include all other reagentsHigh
Peptide competitionSpecificity controlPre-incubate antibody with purified STRAP antigenMedium
Nonimmune serumNegative controlUse serum from same species as primary antibodyLow
Isotype controlSpecificity controlUse non-targeting antibody of same isotypeMedium

For IHC applications, it is particularly important to:

  • Optimize antigen retrieval: Different fixation methods may affect epitope accessibility, requiring optimization of retrieval methods to ensure consistent staining .

  • Titrate antibody concentration: Determine the optimal antibody dilution that maximizes specific signal while minimizing background .

  • Include positive and negative tissue controls in each experiment: This allows for direct comparison between experiments and helps identify technical variations .

  • Document staining patterns: Thoroughly document subcellular localization of staining, as this provides information about the biological relevance of the results .

These controls help distinguish true STRAP protein detection from technical artifacts, enhancing the reliability of IHC results.

How can I design experiments to investigate STRAP protein interactions using antibody-based techniques?

Investigating STRAP protein interactions requires careful experimental design using various antibody-based techniques. Here are methodological approaches:

  • Co-immunoprecipitation (Co-IP):

    • Use STRAP antibodies to pull down STRAP protein complexes from cell lysates

    • Identify interacting partners through Western blotting or mass spectrometry

    • Include appropriate controls: IgG control, input sample, and reciprocal IP (using antibodies against suspected interacting partners)

    • Consider cross-linking to stabilize transient interactions

  • Proximity Ligation Assay (PLA):

    • Detect protein-protein interactions in situ with high specificity

    • Use STRAP antibody in combination with antibodies against potential interacting partners

    • Fluorescent signals appear only when proteins are in close proximity (<40 nm)

    • Quantify interaction frequency and subcellular localization

  • Chromatin Immunoprecipitation (ChIP):

    • If investigating STRAP's role in transcriptional regulation

    • Optimize fixation conditions and sonication parameters

    • Use highly specific STRAP antibodies validated for ChIP applications

    • Include input controls and IgG negative controls

  • Förster Resonance Energy Transfer (FRET):

    • For live-cell analysis of protein interactions

    • Label STRAP and potential partners with appropriate fluorophores

    • Measure energy transfer when proteins interact closely

    • Complement with antibody validation in fixed cells

When designing these experiments, consider the dynamic nature of protein interactions. STRAP, like many signaling proteins, may engage in transient or context-dependent interactions that require careful timing and experimental conditions to detect . Document all experimental conditions thoroughly to ensure reproducibility.

How should I troubleshoot inconsistent STRAP antibody results between different experimental platforms?

Inconsistent results with STRAP antibodies across different experimental platforms (e.g., Western blot vs. IHC vs. FACS) can stem from multiple factors. Here's a systematic troubleshooting approach:

  • Epitope accessibility differences:

    • Different applications expose different protein epitopes

    • For native conditions (like FACS), ensure the antibody recognizes surface-exposed epitopes

    • For denatured conditions (like Western blot), internal epitopes may become accessible

    • Solution: Try antibodies targeting different regions of STRAP

  • Sample preparation variations:

    • Fixation methods can affect epitope recognition in IHC and ICC

    • Lysis buffers influence protein extraction and epitope preservation in Western blot

    • Solution: Optimize sample preparation for each technique separately

  • Antibody concentration optimization:

    • Each technique requires different antibody concentrations

    • Perform titration experiments for each application

    • Solution: Document optimal concentrations for each technique in your hands

  • Cross-platform validation strategies:

    • Use orthogonal methods to confirm results

    • Combine genetic approaches (knockdown/knockout) with antibody detection

    • Compare results with published literature on STRAP localization and expression

  • Protocol standardization:

    • Document detailed protocols for each application

    • Control for batch effects in antibodies and reagents

    • Implement consistent positive and negative controls

Remember that antibodies may be validated for certain applications but not others. Always check the recommended applications for your specific STRAP antibody and consult validation data before concluding that inconsistent results indicate experimental problems rather than technical limitations .

What are the considerations for using STRAP antibodies in multi-color flow cytometry experiments?

Using STRAP antibodies in multi-color flow cytometry experiments requires careful planning and optimization:

  • Antibody fluorophore selection:

    • Choose fluorophores with minimal spectral overlap

    • Consider brightness requirements based on expected STRAP expression levels

    • For intracellular STRAP staining, select fluorophores resistant to fixation/permeabilization

  • Panel design considerations:

    • Place STRAP antibody on a channel with appropriate sensitivity

    • Consider whether STRAP detection is a primary or secondary readout

    • Allocate brighter fluorophores to lower-expressed targets and dimmer fluorophores to abundant proteins

  • Controls for intracellular staining:

    • Include isotype controls conjugated to the same fluorophore

    • Use STRAP-knockout or knockdown samples as negative controls

    • Include single-color controls for compensation setup

  • Fixation and permeabilization optimization:

    • Test multiple fixation/permeabilization protocols as they affect epitope accessibility

    • Balance preservation of cellular structure with antibody penetration

    • Document optimal conditions that maintain both surface marker detection and intracellular STRAP staining

  • Antibody titration:

    • Perform titration experiments to determine optimal signal-to-noise ratio

    • Plot staining index against antibody concentration to identify optimal dilution

    • Remember that optimal concentration may differ from that used in Western blot or IHC

  • Data interpretation considerations:

    • Establish clear positive/negative boundaries using controls

    • Consider using fluorescence-minus-one (FMO) controls

    • For quantitative analysis, include calibration beads to standardize measurements across experiments

Since STRAP is primarily an intracellular protein, the permeabilization step is critical. Different permeabilization reagents (saponin, methanol, commercial kits) may yield different staining patterns and should be systematically tested .

How can I ensure reproducibility when using different lots of STRAP antibodies in long-term studies?

Ensuring reproducibility when using different antibody lots over long-term studies is critical for research integrity. Follow these methodological approaches:

  • Initial lot-to-lot comparison:

    • When receiving a new lot, perform side-by-side comparison with the previous lot

    • Test both lots on identical samples under identical conditions

    • Quantify signal intensity, background levels, and band/staining patterns

    • Document any differences in sensitivity or specificity

  • Reference sample banking:

    • Create and maintain frozen aliquots of reference samples (cell lysates, tissue sections)

    • Use these banked samples to validate each new antibody lot

    • Include positive controls (high STRAP expression) and negative controls (low/no expression)

  • Standardized validation protocol:

    • Develop a standard operating procedure (SOP) for antibody validation

    • Include dilution series, exposure times, and quantification parameters

    • Apply this SOP to each new lot before use in experiments

  • Batch processing strategy:

    • When possible, purchase sufficient antibody from a single lot for all critical experiments

    • For unavoidable lot changes, perform overlap experiments with both lots

    • Consider including a "bridging sample" run with both old and new lots in publications

  • Documentation practices:

    • Record lot numbers, validation dates, and performance metrics

    • Include lot information in methods sections of publications

    • Maintain a laboratory database of antibody performance characteristics

  • Quantitative acceptance criteria:

    • Establish quantitative criteria for lot acceptance (e.g., signal within 20% of previous lot)

    • Define specific parameters that must be met before a new lot is used in experiments

    • Document the decision-making process for accepting or rejecting new lots

By implementing these practices, you can minimize variation introduced by antibody lot changes and maintain data consistency throughout long-term studies.

What statistical approaches are recommended for quantifying Western blot data using STRAP antibodies?

Quantitative analysis of Western blot data using STRAP antibodies requires rigorous statistical approaches to ensure accuracy and reproducibility:

  • Sample size determination:

    • Conduct power analysis to determine appropriate biological replicate numbers

    • Typically, a minimum of 3-5 independent biological replicates is recommended

    • For subtle changes in STRAP expression, more replicates may be necessary

  • Normalization strategies:

    • Always normalize STRAP signals to appropriate loading controls (GAPDH, β-actin, tubulin)

    • Verify that loading control expression is stable across experimental conditions

    • Consider using total protein normalization (stain-free gels or Ponceau staining) as an alternative

  • Linear dynamic range verification:

    • Establish the linear dynamic range of detection for both STRAP and loading control antibodies

    • Run serial dilutions of samples to create standard curves

    • Ensure quantification occurs within the linear range to avoid saturation effects

  • Quantification methodology:

    • Use densitometry software that allows background subtraction

    • Define regions of interest consistently across all blots

    • For complex patterns (multiple bands), clearly define which bands are included in analysis

  • Statistical testing approaches:

    • For two-group comparisons: paired or unpaired t-tests (depending on experimental design)

    • For multiple group comparisons: ANOVA with appropriate post-hoc tests

    • For non-normally distributed data: non-parametric alternatives (Mann-Whitney, Kruskal-Wallis)

    • Include effect sizes alongside p-values for more complete reporting

  • Reporting standards:

    • Present both representative blot images and quantification graphs

    • Include error bars representing standard deviation or standard error

    • Report exact p-values rather than thresholds (p<0.05)

    • Include information about sample sizes and replication

By adhering to these statistical approaches, researchers can generate more reliable and reproducible quantitative data from Western blots using STRAP antibodies.

How should discrepancies between protein detection (STRAP antibody) and mRNA expression data be interpreted?

Discrepancies between STRAP protein levels (detected by antibodies) and mRNA expression data are common and require careful interpretation:

  • Biological mechanisms explaining discrepancies:

    • Post-transcriptional regulation: mRNA stability, microRNA targeting

    • Translational regulation: ribosome occupancy, translation efficiency

    • Post-translational modifications affecting protein stability

    • Protein trafficking and compartmentalization

    • Different half-lives of mRNA versus protein

  • Technical considerations:

    • Antibody specificity: confirm that the antibody detects all relevant STRAP isoforms

    • mRNA detection limitations: primer design may miss certain splice variants

    • Temporal dynamics: protein levels may lag behind mRNA changes

    • Detection sensitivity differences between protein and mRNA methods

  • Validation approaches:

    • Time-course experiments to detect temporal relationships between mRNA and protein changes

    • Use multiple antibodies targeting different STRAP epitopes

    • Apply orthogonal protein detection methods (mass spectrometry)

    • Employ genetic approaches (overexpression, knockdown) to confirm relationships

  • Interpretation framework:

    • Consider the biology of STRAP in your specific context

    • Evaluate whether discrepancies are consistent across experimental conditions

    • Determine if the discrepancy is quantitative (magnitude) or qualitative (direction)

    • Research whether similar discrepancies have been reported for STRAP in literature

  • Integration strategies:

    • Use computational approaches to integrate protein and mRNA data

    • Apply mathematical modeling to understand regulatory dynamics

    • Consider proteome-wide studies that have examined mRNA-protein correlations

    • Report both datasets transparently, acknowledging limitations

Remember that direct comparisons between mRNA and protein levels are not always applicable due to the complex relationship between transcription and translation. Discrepancies often reveal important biological regulation mechanisms rather than technical errors .

How can STRAP antibodies be utilized in structural biology and rational vaccine design?

STRAP antibodies have emerging applications in structural biology and rational vaccine design, leveraging advanced methodological approaches:

  • Structural characterization of STRAP-antibody complexes:

    • X-ray crystallography of STRAP-antibody complexes provides atomic-level resolution of interaction sites

    • Cryo-electron microscopy (cryo-EM) enables visualization of STRAP-antibody complexes without crystallization

    • NMR spectroscopy characterizes the dynamic nature of STRAP-antibody interactions in solution

  • Epitope mapping applications:

    • Use multiple STRAP antibodies targeting different epitopes to create comprehensive conformational maps

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) identifies antibody binding regions

    • Alanine scanning mutagenesis determines critical residues for antibody recognition

  • Structure-guided antibody engineering:

    • Use structural data to modify STRAP antibodies for improved specificity or affinity

    • Engineer antibodies to recognize specific conformational states of STRAP

    • Develop bispecific antibodies that target STRAP and interacting partners simultaneously

  • Leveraging conformational dynamics:

    • Consider STRAP as a conformational ensemble rather than a static structure

    • Use antibodies to trap specific conformational states for functional studies

    • Develop antibodies sensitive to post-translational modifications that alter STRAP conformation

  • Applications in vaccine development:

    • If STRAP has relevance in pathogen interactions, antibodies can guide immunogen design

    • Structure-based immunogen design focuses immune responses on relevant epitopes

    • Sequential immunization strategies guided by structural understanding of antibody-antigen interactions

These advanced applications integrate structural biology with antibody technology to gain deeper insights into STRAP function and potential therapeutic applications.

What are the latest methodologies for developing and validating STRAP antibodies for super-resolution microscopy?

Super-resolution microscopy techniques require specialized considerations for STRAP antibody selection and validation:

  • Antibody selection criteria for super-resolution applications:

    • High specificity is critical as non-specific binding becomes more apparent at nanoscale resolution

    • Affinity considerations: higher affinity antibodies generally provide better signal-to-noise ratios

    • Small probes (Fab fragments, nanobodies) may provide better spatial resolution than full IgG antibodies

    • Photostability of conjugated fluorophores must match the imaging technique (STED, STORM, PALM)

  • Validation strategies specific to super-resolution microscopy:

    • Compare conventional and super-resolution imaging patterns using the same antibody

    • Use genetic approaches (STRAP-knockout controls) to confirm specificity at nanoscale resolution

    • Perform dual-color imaging with antibodies against different STRAP epitopes to confirm co-localization

    • Validate with orthogonal approaches (tagged STRAP expression) for localization confirmation

  • Sample preparation optimization:

    • Test multiple fixation protocols to preserve nanoscale structures

    • Optimize permeabilization to ensure antibody accessibility while maintaining ultrastructure

    • Evaluate clearing techniques to reduce background without affecting epitope accessibility

    • Consider the distance between epitope and fluorophore (linkage error)

  • Quantitative validation approaches:

    • Establish resolution measurements with known standards

    • Perform cluster analysis to characterize STRAP distribution patterns

    • Use nearest neighbor analysis to evaluate specificity of labeling

    • Implement colocalization analysis with known STRAP interaction partners

  • Controls specific to super-resolution techniques:

    • Include single-fluorophore controls for multi-color super-resolution imaging

    • Use spatially separated fiducial markers for drift correction

    • Implement density-based controls to assess labeling efficiency

    • Include negative controls processed identically to experimental samples

These methodologies ensure that STRAP antibodies provide reliable and reproducible results in cutting-edge super-resolution microscopy applications, enabling visualization of STRAP localization and interactions at nanoscale resolution.

How can machine learning approaches enhance STRAP antibody validation and experimental design?

Machine learning (ML) approaches are increasingly valuable for enhancing STRAP antibody validation and experimental design:

  • Automated image analysis for validation:

    • ML algorithms can analyze Western blot images to identify specific versus non-specific bands

    • Convolutional neural networks (CNNs) can assess staining patterns in IHC/ICC for consistency

    • Pattern recognition algorithms can detect subtle batch-to-batch antibody variations

    • Automated analysis reduces subjective interpretation and increases reproducibility

  • Epitope prediction improvements:

    • ML models predict antibody binding sites with increasing accuracy

    • Deep learning approaches integrate sequence and structural information

    • These predictions help select optimal epitopes for new STRAP antibody development

    • Models can predict cross-reactivity with related proteins, enhancing specificity assessment

  • Experimental design optimization:

    • ML algorithms can identify optimal combinations of experimental conditions

    • Bayesian optimization approaches efficiently explore parameter spaces

    • Active learning strategies direct experiments toward the most informative next steps

    • Reduce experimental iterations required for antibody validation and protocol optimization

  • Data integration and pattern discovery:

    • ML methods integrate data across multiple validation approaches

    • Identify subtle patterns in antibody performance across different applications

    • Detect relationships between antibody characteristics and experimental outcomes

    • Generate hypotheses about factors affecting antibody reliability

  • Predictive models for antibody performance:

    • Train models on existing validation data to predict performance in new applications

    • Forecast lot-to-lot variation effects based on historical data

    • Predict optimal antibody dilutions for new experimental conditions

    • Identify high-risk experiments where antibody performance might be compromised

  • Implementation considerations:

    • Require sufficient training data from standardized validation experiments

    • Validate ML model predictions with experimental verification

    • Develop user-friendly interfaces for researchers without ML expertise

    • Document ML methodologies thoroughly for reproducibility

By incorporating these ML approaches, researchers can enhance the rigor of STRAP antibody validation, optimize experimental design, and increase the reliability and reproducibility of results across different research applications.

What are the current consensus best practices for STRAP antibody usage in multimodal research programs?

The current consensus best practices for STRAP antibody usage in multimodal research programs integrate validation, documentation, and methodological considerations:

  • Comprehensive validation across applications:

    • Validate each STRAP antibody independently for each application (WB, IHC, FACS, etc.)

    • Implement application-specific controls and acceptance criteria

    • Document validation results systematically for reference and reporting

    • Consider the dynamic range and sensitivity requirements for each application

  • Integrated experimental approach:

    • Combine multiple techniques to build a coherent understanding of STRAP biology

    • Use orthogonal methods to confirm key findings (antibody-based and non-antibody methods)

    • Implement genetic approaches (knockout/knockdown) alongside antibody detection

    • Consider temporal and spatial dimensions in experimental design

  • Standardized reporting practices:

    • Document detailed antibody information (catalog number, lot, dilution, incubation conditions)

    • Include validation data in publications or supplementary materials

    • Show representative images of control experiments

    • Clearly describe quantification methodologies and statistical approaches

  • Cross-laboratory standardization:

    • Establish common validation protocols within research collaborations

    • Share reference samples between laboratories

    • Implement standard operating procedures for key techniques

    • Use consistent data analysis approaches across the research program

  • Continuous validation strategy:

    • Regularly reassess antibody performance, especially with new lots

    • Update protocols based on new validation findings

    • Maintain a central database of antibody performance characteristics

    • Document changes in antibody behavior over time

By adhering to these consensus best practices, researchers can ensure reliable, reproducible results when using STRAP antibodies across diverse experimental modalities, ultimately enhancing the quality and impact of their research programs.

What future developments are anticipated in STRAP antibody technology and applications?

Future developments in STRAP antibody technology and applications are likely to advance along several promising trajectories:

  • Enhanced antibody engineering:

    • Development of recombinant antibodies with defined sequences for improved reproducibility

    • Creation of smaller binding fragments (nanobodies, affimers) for improved tissue penetration and spatial resolution

    • Site-specific conjugation technologies for precise control of fluorophore or enzyme attachment

    • Bispecific antibodies targeting STRAP and interacting partners simultaneously

  • Advanced imaging applications:

    • Integration with emerging super-resolution techniques beyond current limits

    • Live-cell compatible antibody formats for dynamic STRAP visualization

    • Multiplexed imaging approaches allowing simultaneous detection of dozens of targets alongside STRAP

    • Expansion microscopy compatible antibodies for enhanced spatial resolution

  • Single-cell analysis integration:

    • Antibody-based methods for spatial transcriptomics combined with STRAP protein detection

    • Mass cytometry (CyTOF) panel development incorporating STRAP detection

    • Single-cell proteomics approaches using STRAP antibodies

    • Microfluidic applications for high-throughput single-cell analysis

  • Computational and AI integration:

    • Advanced algorithms for antibody design targeting specific STRAP epitopes

    • Machine learning approaches for predicting cross-reactivity and performance

    • Automated validation pipelines for high-throughput antibody assessment

    • Digital pathology integration with STRAP antibody staining patterns

  • Therapeutic and diagnostic applications:

    • Development of STRAP-targeting antibodies for potential therapeutic applications

    • Diagnostic applications if STRAP emerges as a biomarker in disease states

    • Antibody-drug conjugates targeting STRAP in relevant disease contexts

    • Theranostic applications combining imaging and therapeutic functions

  • Reproducibility technologies:

    • Blockchain-based antibody validation records for enhanced transparency

    • Community-based validation platforms for sharing performance data

    • Standardized reference materials for validation across laboratories

    • Automated validation robots for consistent antibody assessment

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.