SCRL17 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
SCRL17 antibody; At2g25685 antibody; F3N11Putative defensin-like protein 239 antibody; Putative S locus cysteine-rich-like protein 17 antibody; Protein SCRL17 antibody; SCR-like protein 17 antibody
Target Names
SCRL17
Uniprot No.

Target Background

Database Links
Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What are the optimal storage conditions for SCRL17 Antibody?

For optimal longevity and activity preservation of SCRL17 Antibody, follow these evidence-based storage guidelines:

  • Store at -20 to -70°C for up to 12 months from receipt date in original packaging

  • After reconstitution, store at 2-8°C under sterile conditions for up to 1 month

  • For longer storage after reconstitution, store at -20 to -70°C for up to 6 months

It is critical to use a manual defrost freezer and avoid repeated freeze-thaw cycles as each cycle can reduce antibody activity by approximately 10-15%. For maximum stability, aliquot the reconstituted antibody into single-use volumes before freezing.

What reconstitution protocol is recommended for SCRL17 Antibody?

Proper reconstitution ensures optimal antibody performance. Follow this methodological approach:

  • Allow the lyophilized antibody to reach room temperature (15-25°C)

  • Reconstitute in sterile, ultrapure water or recommended buffer (typically PBS)

  • Gently rotate or invert the vial—avoid vortexing to prevent protein denaturation

  • Allow solution to sit for 15-20 minutes at room temperature for complete dissolution

  • If not using immediately, prepare single-use aliquots to minimize freeze-thaw cycles

A reconstitution calculator can help determine the appropriate buffer volume based on your desired final concentration . Document the reconstitution date and concentration for experimental reproducibility.

How should SCRL17 Antibody specificity be validated before experimental use?

Rigorous validation is essential before incorporating SCRL17 Antibody into research protocols. Implement these complementary approaches:

  • Positive and negative controls:

    • Test against tissues/cells known to express or lack the target

    • Include genetic knockouts or knockdowns as gold-standard controls

  • Epitope blocking:

    • Pre-incubate antibody with immunizing peptide

    • Compare staining patterns between blocked and unblocked conditions

  • Orthogonal validation:

    • Correlate protein detection with mRNA expression data

    • Compare results with alternative antibodies targeting different epitopes

  • Specificity confirmation:

    • Western blot analysis to verify molecular weight specificity

    • Immunoprecipitation followed by mass spectrometry identification

  • Genetic manipulation:

    • Test in CRISPR/Cas9-edited cell lines

    • Verify signal reduction or elimination in knockout models

A comprehensive validation strategy employs at least three independent approaches to ensure research reproducibility and reliability .

What approaches are recommended for determining optimal working dilutions?

Systematic titration is critical for determining the optimal working concentration of SCRL17 Antibody across applications:

  • Perform serial dilution experiments starting from manufacturer's recommended range

  • For immunohistochemistry/immunofluorescence:

    • Test dilutions ranging from 1:50 to 1:1000

    • Evaluate signal-to-noise ratio at each concentration

    • Document staining pattern, intensity, and background

  • For Western blotting:

    • Test dilutions from 1:200 to 1:5000

    • Assess band specificity, intensity, and background

    • Determine minimum concentration yielding detectable specific signal

  • For flow cytometry:

    • Test concentrations from 0.1-10 μg/mL

    • Calculate staining index (mean positive - mean negative/2× SD of negative)

    • Select concentration with highest staining index

ApplicationRecommended Initial Dilution RangeOptimization MetricQuality Control Parameter
IHC-P1:100-1:500Signal:background ratioPositive tissue controls
IF/ICC1:100-1:500Signal intensity/specificitySubcellular localization
WB1:500-1:2000Band specificityMolecular weight verification
Flow1:50-1:200Staining indexFluorescence-minus-one controls
ELISA1:1000-1:10000Detection limitStandard curve linearity

Document optimal dilutions for each application and lot number to ensure experimental reproducibility .

What strategies can address weak or absent signal when using SCRL17 Antibody?

When encountering weak or absent signal, implement this systematic troubleshooting approach:

  • Epitope accessibility issues:

    • Optimize antigen retrieval methods (heat-induced vs. enzymatic)

    • Test multiple retrieval buffers (citrate pH 6.0, EDTA pH 8.0, Tris pH 9.0)

    • Extend retrieval time (10-30 minutes)

    • For formaldehyde-fixed samples, consider longer retrieval times

  • Antibody concentration optimization:

    • Increase antibody concentration incrementally

    • Extend primary antibody incubation time (overnight at 4°C)

    • Test different detection systems with higher sensitivity

  • Sample preparation assessment:

    • Verify proper fixation protocols (duration, penetration)

    • Assess tissue integrity and antigen preservation

    • Consider testing fresh samples to rule out antigen degradation

  • Detection system enhancement:

    • Switch to more sensitive detection methods (TSA, polymer-based)

    • For fluorescence applications, use brighter fluorophores

    • Consider signal amplification approaches (avidin-biotin)

  • Technical verification:

    • Test antibody on known positive controls

    • Verify integrity of detection reagents

    • Assess antibody functionality using simple ELISA

The systematic modification of individual parameters will help identify and address the specific cause of signal issues .

How can non-specific binding and high background be reduced when using SCRL17 Antibody?

High background can obscure specific signals. Implement these methodological approaches to improve signal-to-noise ratio:

  • Blocking optimization:

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Extend blocking time (1-2 hours at room temperature)

    • Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions

    • Consider dual blocking with protein and serum

  • Washing protocol enhancement:

    • Increase number of wash steps (minimum 3×5 minutes)

    • Use gentle agitation during washing

    • Add 0.05-0.1% Tween-20 to wash buffers

    • Ensure complete removal of wash buffer between steps

  • Antibody dilution adjustment:

    • Increase dilution of primary antibody

    • Optimize secondary antibody concentration

    • Pre-absorb antibodies with irrelevant tissues

  • Endogenous enzyme inhibition:

    • For peroxidase-based detection, block with 0.3-3% H₂O₂

    • For alkaline phosphatase, add levamisole

    • Quench endogenous biotin with avidin-biotin blocking kit

  • Tissue-specific considerations:

    • For tissues with high endogenous Fc receptors, add Fc block

    • For fatty tissues, include additional detergent in wash buffers

    • For highly autofluorescent samples, use Sudan Black or TrueBlack

Systematic optimization of these parameters will significantly improve signal specificity while reducing background interference .

How can SCRL17 Antibody be incorporated into multiplexed immunoassays?

Multiplexed detection with SCRL17 Antibody requires specialized approaches to maintain specificity while enabling simultaneous target detection:

  • Panel design considerations:

    • Select antibodies from different host species when possible

    • Use isotype-specific secondary antibodies to avoid cross-reactivity

    • Plan detection strategy based on target abundance and localization

    • Consider spectral overlap when selecting fluorophores

  • Sequential staining approaches:

    • Implement tyramide signal amplification for sequential detection

    • Use complete stripping or blocking between rounds

    • Validate absence of cross-reactivity between rounds

  • Spectral imaging optimization:

    • Implement appropriate spectral unmixing algorithms

    • Include single-stained controls for spectral fingerprinting

    • Utilize computational approaches to resolve overlapping signals

  • Mass cytometry integration:

    • Metal-tag conjugation of SCRL17 for CyTOF applications

    • Optimize panel design to minimize signal spillover

    • Implement barcoding strategies for batch consistency

  • Validation requirements:

    • Compare multiplex results with single-staining controls

    • Verify absence of steric hindrance between antibodies

    • Document potential epitope blocking between antibodies

Recent advances in multiplexed imaging have enabled simultaneous detection of 40+ targets, requiring meticulous panel design and validation .

What approaches can be used to study antibody-antigen interactions using cryoEM?

CryoEM offers powerful capabilities for structural characterization of antibody-antigen complexes:

  • Sample preparation for cryoEM:

    • Purify antibody-antigen complex to homogeneity

    • Optimize buffer conditions for particle distribution

    • Screen grid types and vitrification conditions

  • Data collection strategy:

    • Implement dose fractionation for motion correction

    • Collect tilt series for particles with preferred orientation

    • Use phase plates for enhanced contrast of smaller complexes

  • Structure determination workflow:

    • Implement 2D classification to identify homogeneous particles

    • Perform 3D reconstruction with imposed symmetry if appropriate

    • Refine to highest possible resolution

  • Epitope mapping applications:

    • Identify specific binding interfaces at near-atomic resolution

    • Characterize conformational epitopes difficult to study by other methods

    • Determine antibody approach angles and binding footprints

  • Integration with sequence information:

    • Combine cryoEM with next-generation sequencing data

    • Identify antibody sequences from structural information

    • Enable rapid monoclonal antibody discovery from polyclonal sera

CryoEM analysis of antibody-antigen complexes typically requires resolutions of 4Å or better for detailed epitope characterization, but even intermediate resolution (~6-8Å) can provide valuable binding orientation information .

How can neutralizing activity of SCRL17 Antibody be quantitatively assessed?

Neutralization assessment requires functional assays that quantify the antibody's ability to inhibit biological activity:

  • Receptor-ligand interaction assays:

    • ELISA-based competition assays

    • Surface plasmon resonance for real-time kinetics

    • Cell-based reporter systems for functional blockade

  • Virus neutralization approaches (for viral targets):

    • Plaque reduction neutralization tests (PRNT)

    • Pseudovirus neutralization assays

    • Cell-based viral entry inhibition assays

  • Quantitative metrics:

    • IC50/EC50 determination with 95% confidence intervals

    • Maximum inhibition percentage

    • Area under the neutralization curve

  • Mechanism of action studies:

    • Pre- vs. post-attachment neutralization

    • Antibody-dependent cellular cytotoxicity (ADCC) assessment

    • Complement-dependent cytotoxicity (CDC) evaluation

  • In vivo validation:

    • Passive transfer protection studies

    • Challenge studies with pathogens

    • Biomarker assessment in appropriate animal models

For SARS-CoV-2 related research, emerging approaches combine antibodies targeting different epitopes to enhance neutralization breadth and potency against emerging variants .

How can single B-cell sequencing enhance antibody discovery and characterization?

Single B-cell sequencing represents a powerful approach for discovering and characterizing antibodies with desired properties:

  • Isolation of antigen-specific B cells:

    • Fluorescence-activated cell sorting using labeled antigens

    • Magnetic enrichment of antigen-specific B cells

    • Microfluidic approaches for single-cell isolation

  • Sequencing methodologies:

    • 5' RACE PCR for paired heavy and light chain sequences

    • Next-generation sequencing of antibody repertoires

    • Single-cell RNA-seq for transcriptional profiling

  • Bioinformatic analysis:

    • Clonal family identification and clustering

    • Lineage analysis and somatic hypermutation mapping

    • Complementarity-determining region (CDR) characterization

  • Recombinant antibody production:

    • Cloning of variable regions into expression vectors

    • Transient expression in mammalian cells

    • Purification and functional validation

  • Integration with structural data:

    • Combining sequence information with structural studies

    • Structure-guided maturation and optimization

    • Epitope-specific antibody discovery

This approach allows for the isolation of naturally occurring antibodies with desired characteristics, bypassing traditional hybridoma technology limitations . The methodology captures the native heavy and light chain pairing, enabling more faithful recapitulation of the original antibody properties.

What computational approaches are emerging for antibody engineering and optimization?

Computational methods are revolutionizing antibody engineering and optimization:

  • Structure-based design approaches:

    • Homology modeling of antibody variable regions

    • Molecular dynamics simulations of antibody-antigen interactions

    • In silico alanine scanning to identify critical binding residues

  • Machine learning applications:

    • Prediction of antibody developability properties

    • Optimization of humanization strategies

    • De novo antibody design based on target epitopes

  • Library design methodologies:

    • Computational design of focused mutagenesis libraries

    • CDR optimization through in silico modeling

    • Framework optimization for stability enhancement

  • Affinity maturation strategies:

    • Energy-based optimization of binding interfaces

    • Electrostatic complementarity enhancement

    • Hydrophobic core redesign for stability

  • Developability assessment:

    • Aggregation propensity prediction

    • Identification of post-translational modification sites

    • Chemical stability prediction

These computational approaches can significantly accelerate antibody engineering while reducing experimental burden. Integration of artificial intelligence methods has further enhanced predictive capabilities for antibody optimization .

What statistical approaches are recommended for analyzing variability in antibody-based assays?

Robust statistical analysis is essential for interpreting antibody assay data:

  • Assessing technical variability:

    • Intra-assay coefficient of variation (CV) calculation

    • Inter-assay reproducibility assessment

    • Nested analysis of variance (ANOVA) for multi-level variation

  • Sample size determination:

    • Power analysis based on preliminary data

    • Sequential testing approaches with adaptive designs

    • Minimum detectable difference calculations

  • Statistical testing methodologies:

    • Paired vs. unpaired t-tests for simple comparisons

    • ANOVA with appropriate post-hoc corrections

    • Non-parametric alternatives for non-normal distributions

    • Mixed effects models for repeated measures designs

  • Advanced statistical considerations:

    • Bayesian approaches for small sample sizes

    • Multiple testing correction (Bonferroni, FDR)

    • Equivalence testing for biosimilarity assessment

  • Data visualization best practices:

    • Representation of individual data points

    • Clear indication of sample sizes

    • Appropriate error bars (SD vs. SEM vs. CI)

    • Consistent scaling and color schemes

For immunoassays, acceptable inter-assay CV is typically <15% and intra-assay CV <10%. Implementing appropriate statistical approaches ensures reliable interpretation of experimental results and facilitates reproducibility across laboratories .

How can machine learning enhance image analysis for antibody-based detection methods?

Machine learning approaches are transforming quantitative analysis of antibody-based imaging:

  • Cell segmentation advancements:

    • Deep learning for precise cell boundary detection

    • Instance segmentation for overlapping cells

    • Multi-channel integration for phenotypic classification

  • Quantification approaches:

    • Automated intensity measurement across cellular compartments

    • Object-based colocalization analysis

    • Morphological feature extraction and classification

  • Multiplexed analysis capabilities:

    • Cell type identification in heterogeneous populations

    • Spatial relationship mapping between cell types

    • Neighborhood analysis in tissue microenvironments

  • Implementation considerations:

    • Training data requirements and annotation approaches

    • Transfer learning for model adaptation

    • Validation against manual quantification

  • Software and workflow integration:

    • Open-source platforms (CellProfiler, QuPath, ImageJ)

    • Cloud-based analysis pipelines

    • Standardized data formats for reproducibility

Machine learning approaches have demonstrated superior performance compared to traditional threshold-based methods, particularly for complex tissue architectures and variable staining patterns. These methods can reduce human bias while increasing throughput and reproducibility .

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.