SRPX Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Fundamental Composition of Biotin-Conjugated Antibodies

Biotin-conjugated antibodies combine two critical biological components:

  • Primary antibody: Targets specific antigens through Fab regions

  • Biotin molecule: Covalently attached via lysine residues or carbohydrate chains using NHS ester chemistry

The biotin-spacer (Biotin-SP) configuration with a 6-atom spacer arm enhances binding accessibility, improving detection sensitivity by 40-60% compared to direct conjugation .

Key Technical Parameters

PropertyStreptavidin-AP Conjugate Streptavidin-HRP System RFP Antibody Biotin
Binding Affinity (Kd)10⁻¹⁵ M10⁻¹⁵ MN/A
Detection Limit0.1-1 ng/mL (ELISA)0.01 ng/mL (Enhanced) 5 ng/mL (WB)
Optimal Working Dilution1:5,0001:10,0001:1,000
Signal Amplification8-12x15-20x3-5x
Stability2 years at 4°C18 months at -20°C6 months at 4°C

Multiplex Detection Capabilities

Biotin conjugates enable parallel detection through:

  • Streptavidin-AP (Alkaline Phosphatase): NBT/BCIP substrate (blue)

  • Streptavidin-HRP (Horseradish Peroxidase): TMB substrate (yellow)

Quality Control Standards

Certified biotin conjugates must demonstrate:

  • ≥90% monomeric purity (SEC-HPLC)

  • <5% free biotin contamination

  • Defined biotin:antibody ratio (4-6 molecules per IgG)

Case Study: RFP Antibody Biotin Conjugate

  • Host Species: Rabbit IgG

  • Validation Data:

    • Western Blot: 5 ng detection limit

    • ELISA Linear Range: 1-100 ng/mL

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery time may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery times.
Synonyms
SRPX antibody; ETX1 antibody; Sushi repeat-containing protein SRPX antibody
Target Names
SRPX
Uniprot No.

Target Background

Function
SRPX may play a role in phagocytosis during disc shedding, cell adhesion to cells other than the pigment epithelium, or signal transduction.
Gene References Into Functions
  1. SRPX1 co-accumulated with Abeta deposits in cerebral blood vessels of all autopsied cases with severe cerebral amyloid angiopathy (CAA); no SRPX1 co-accumulated with Abeta deposits in senile plaques. Additionally, we demonstrated that both Abeta40 and Abeta42 bound to SRPX1 in vitro and enhanced SRPX1 expression in primary cultures of cerebrovascular smooth muscle cells. SRPX1 enhanced caspase activity induced by Abeta40. PMID: 28478503
  2. The SRPX protein contains the P-DUDES structural domain in its C-terminal region. This domain exhibits significant, albeit remote, sequence similarity to thioredoxin-like domains and is predicted to possess an oxidoreductase function. PMID: 209648
  3. PELO is subcellularly localized at the actin cytoskeleton, interacts with HAX1, EIF3G, and SRPX proteins, and this interaction occurs at the cytoskeleton. This interaction may facilitate PELO to detect and degrade aberrant mRNAs. PMID: 20406461
  4. An apoptosis-inducing protein localized in the endoplasmic reticulum which increases efficiency of apoptosis PMID: 15021917
  5. expression of drs mRNA in well-differentiated, moderately differentiated, and poorly differentiated lung adenocarcinoma tissues (drs; down-regulated by v-scr) PMID: 12152160
  6. down-regulation of drs mRNA is closely correlated with carcinomas which arise from adenomatous polyps in the course of the adenoma-carcinoma sequence, but most carcinomas arising de novo are independent of the tumor suppressor function of the drs gene PMID: 12716466
  7. Down-regulation of drs mRNA is closely correlated with development of prostate carcinoma, suggesting a tumor-suppressor function of the drs gene in this cancer. PMID: 12874760
  8. Downregulation of drs tumor suppressor gene in highly malignant human pulmonary neuroendocrine tumors. PMID: 19424611
  9. This is the first report demonstrating overexpression of ETX1 in glaucomatous trabecular meshwork. PMID: 19862339

Show More

Hide All

Database Links

HGNC: 11309

OMIM: 300187

KEGG: hsa:8406

STRING: 9606.ENSP00000367794

UniGene: Hs.15154

Subcellular Location
Cell surface. Note=Possibly surface of photoreceptor cell.
Tissue Specificity
Retina and heart; less in placenta, pancreas, lung, liver, skeletal muscle, kidney and brain.

Q&A

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

SRPX (Sushi repeat-containing protein SRPX) is a protein that may play important biological roles in phagocytosis during disk shedding, cell adhesion to cells other than the pigment epithelium, and signal transduction pathways . The significance of SRPX in research stems from its potential involvement in cellular processes that are fundamental to understanding both normal physiology and pathological conditions. Researchers often study SRPX in cell biology contexts to elucidate its functional roles in cellular communication and structural organization.

What are the key characteristics of SRPX Antibody, Biotin conjugated?

SRPX Antibody, Biotin conjugated (e.g., CSB-PA022689LD01HU) is a polyclonal antibody developed in rabbit hosts that specifically targets human SRPX protein. The antibody is generated using recombinant Human Sushi repeat-containing protein SRPX (amino acids 31-237) as the immunogen . The biotin conjugation enables signal amplification through high-affinity interaction with avidin or streptavidin molecules. This antibody typically has IgG isotype characteristics, is purified using Protein G chromatography (>95% purity), and is stored in a buffer containing preservative (0.03% Proclin 300), 50% glycerol, and 0.01M PBS at pH 7.4 .

How does biotin conjugation enhance antibody performance in immunoassays?

Biotin conjugation enhances antibody performance through several mechanisms:

  • Signal amplification: Biotin forms a high-affinity, non-covalent bond with avidin/streptavidin, enabling multiple detection molecules to bind to a single antibody, thereby significantly amplifying the detection signal .

  • Versatility in detection systems: Biotinylated antibodies can be detected using various streptavidin or avidin conjugates linked to fluorophores, enzymes, or other detection molecules, providing flexibility in experimental design .

  • Sensitivity improvement: When biotin includes a spacer arm (like Biotin-SP), it extends the biotin moiety away from the antibody surface, making it more accessible to binding sites on streptavidin and thereby increasing sensitivity, especially with alkaline phosphatase-conjugated streptavidin .

  • Application across multiple techniques: The biotin-streptavidin system enables consistent use across various techniques including ELISA, Western blotting, immunohistochemistry, immunocytochemistry, and flow cytometry .

What is the optimal protocol for using SRPX Antibody, Biotin conjugated in ELISA?

Optimal ELISA Protocol for SRPX Antibody, Biotin conjugated:

  • Coating: Coat 96-well plate with capture antibody (anti-SRPX) at 1-2 μg/ml in carbonate buffer (pH 9.6) overnight at 4°C.

  • Blocking: Block remaining protein-binding sites with 1-5% BSA or appropriate blocking solution for 1-2 hours at room temperature.

  • Sample addition: Add samples and standards in dilution buffer (typically PBS with 0.05% Tween-20 and 1% BSA) for 1-2 hours at room temperature.

  • Primary detection: Apply SRPX Antibody, Biotin conjugated at optimal dilution (typically 1:2000-1:10000 for ELISA applications) . Incubate for 1-2 hours at room temperature.

  • Secondary detection: Add streptavidin-HRP or streptavidin conjugated to appropriate enzyme at recommended dilution. Incubate for 30-60 minutes.

  • Substrate addition: Add appropriate substrate (TMB for HRP) and monitor color development.

  • Stopping reaction: Add stop solution (e.g., 2N H₂SO₄ for TMB).

  • Measurement: Read absorbance at appropriate wavelength (450nm for TMB).

Between each step, wash the plate 3-5 times with washing buffer (PBS with 0.05% Tween-20) to remove unbound reagents.

How should researchers optimize biotin-conjugated antibody concentration for signal amplification techniques?

Optimization Process for Biotin-Conjugated Antibody Concentration:

  • Titration experiment: Perform a checkerboard titration with serial dilutions of the biotinylated antibody (typically ranging from 1:100 to 1:10,000) against various concentrations of target protein.

  • Signal-to-noise assessment: For each dilution, calculate the signal-to-noise ratio by dividing specific signal by background signal. The optimal dilution typically provides the highest signal-to-noise ratio rather than the strongest absolute signal.

  • Streptavidin conjugate optimization: Once the optimal antibody dilution is established, optimize the streptavidin conjugate concentration through a similar titration procedure.

  • Incubation parameters: Test different incubation times (30 minutes to overnight) and temperatures (4°C, room temperature, 37°C) to determine conditions that maximize specific binding while minimizing background.

  • Buffer composition adjustment: Modify buffer components (salt concentration, pH, blocking agents) to improve specificity and reduce non-specific binding.

  • Positive and negative controls: Always include appropriate controls to validate results and ensure specificity of the detection system.

  • Endogenous biotin blocking: When working with tissues or cells that contain endogenous biotin, incorporate a biotin blocking step to prevent false positive signals .

How can researchers address non-specific binding issues when using SRPX Antibody, Biotin conjugated?

Strategies to Minimize Non-Specific Binding:

  • Optimize blocking conditions: Test different blocking agents (BSA, casein, normal serum, commercial blockers) at various concentrations (1-5%) and incubation times (1-3 hours) to identify the most effective combination for your specific sample type.

  • Adjust antibody dilution: Increase the dilution of the SRPX Antibody, Biotin conjugated to reduce non-specific interactions, while maintaining sufficient specific binding.

  • Modify wash protocols: Increase the number of washes (from 3 to 5-7) and/or the stringency of wash buffers (adding additional detergent or salt) to remove weakly bound antibodies.

  • Pre-adsorption: Consider pre-adsorbing the antibody with proteins from the species of your sample to remove cross-reactive antibodies.

  • Buffer optimization: Adjust buffer components by adding detergents (0.05-0.1% Tween-20), carrier proteins (0.1-1% BSA), or increasing salt concentration (150-500 mM NaCl) to reduce non-specific interactions.

  • Control for endogenous biotin: Use commercial endogenous biotin blocking kits before applying biotinylated antibodies, particularly important for tissues rich in endogenous biotin (liver, kidney, brain) .

  • Cross-reactivity testing: Verify antibody specificity using Western blot or immunoprecipitation to ensure it recognizes only the intended target.

  • Secondary detection optimization: Use highly cross-adsorbed streptavidin conjugates to minimize non-specific binding to endogenous biotin-containing proteins.

What are the critical storage conditions for maintaining SRPX Antibody, Biotin conjugated activity over time?

Optimal Storage and Handling Conditions:

  • Temperature: Store the antibody at -20°C or -80°C for long-term storage. Avoid repeated freeze-thaw cycles by creating small, single-use aliquots upon receipt .

  • Buffer composition: The antibody is typically stored in a buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative . This composition helps maintain stability during freeze-thaw cycles.

  • Aliquoting strategy: Divide the original antibody solution into small volumes (10-20 μl) sufficient for single experiments to prevent contamination and degradation from repeated handling.

  • Working dilution stability: Diluted antibody solutions should be prepared fresh for each experiment. If storage is necessary, keep at 4°C for no more than 1 week.

  • Light exposure: Minimize exposure to light, especially important for fluorophore-conjugated detection reagents used with biotinylated antibodies.

  • Contamination prevention: Use sterile techniques when handling the antibody to prevent microbial contamination, which can degrade antibody activity and introduce experimental artifacts.

  • Monitoring antibody performance: Periodically test antibody activity using positive controls to ensure continued functionality, especially for antibodies stored for extended periods.

How does the performance of SRPX Antibody, Biotin conjugated compare with FITC-conjugated alternatives in multiplexed imaging?

Comparative Analysis of Biotin vs. FITC Conjugation for Multiplexed Imaging:

ParameterBiotin ConjugationFITC Conjugation
Signal AmplificationHigh (through multiple streptavidin binding)Limited (direct fluorescence)
Detection FlexibilityHigh (can use various streptavidin conjugates)Fixed (FITC emission spectrum)
Sequential StainingCompatible with multi-round staining protocolsLess suitable for sequential staining
Photobleaching ResistanceDependent on streptavidin-fluorophore usedModerate to low (FITC bleaches relatively quickly)
Background ConcernsEndogenous biotin can cause backgroundAutofluorescence in similar spectrum can interfere
Multiplexing CapacityHigh (when used with spectrally distinct fluorophores)Limited by spectral overlap with other fluorophores
Procedure ComplexityMore steps (primary + streptavidin detection)Fewer steps (direct detection)
Cost ConsiderationsHigher (requires additional detection reagents)Lower (single reagent system)

For multiplexed imaging applications, the SRPX Antibody, Biotin conjugated (CSB-PA022689LD01HU) offers superior flexibility compared to the FITC-conjugated version (CSB-PA022689LC01HU) . The biotin-conjugated antibody enables signal amplification through multivalent streptavidin binding, which is particularly advantageous for detecting low-abundance targets. Additionally, researchers can select from various streptavidin conjugates with distinct fluorophores to optimize spectral separation in multiplexed experiments.

What methodological approaches can researchers use to validate SRPX antibody specificity in their experimental systems?

Comprehensive Validation Strategies for SRPX Antibody Specificity:

  • Western blot analysis:

    • Compare observed molecular weight with predicted SRPX protein size

    • Include positive and negative control samples

    • Perform peptide competition assays by pre-incubating antibody with immunizing peptide

  • Knockout/knockdown validation:

    • Test antibody on SRPX knockout/knockdown samples (CRISPR, siRNA, shRNA)

    • Compare signal intensity between wild-type and knockout/knockdown samples

    • Quantify signal reduction corresponding to knockdown efficiency

  • Orthogonal antibody comparison:

    • Compare staining patterns with alternate SRPX antibodies targeting different epitopes

    • Evaluate concordance in signal localization and intensity

    • Identify consensus binding patterns across multiple antibodies

  • Recombinant expression systems:

    • Overexpress tagged SRPX protein in cell lines

    • Confirm co-localization of anti-SRPX signal with tag-specific antibody

    • Test antibody on non-transfected versus transfected cells

  • Mass spectrometry validation:

    • Perform immunoprecipitation using the SRPX antibody

    • Analyze pulled-down proteins by mass spectrometry

    • Confirm presence of SRPX protein and identify potential cross-reactive proteins

  • Immunohistochemistry controls:

    • Include tissues known to express or lack SRPX

    • Perform absorption controls by pre-incubating with recombinant SRPX protein

    • Compare staining patterns with in situ hybridization data for SRPX mRNA

  • Cross-species reactivity assessment:

    • Test antibody on samples from multiple species where sequence homology is known

    • Confirm reactivity aligns with sequence conservation predictions

    • Document species-specific variations in binding efficiency

What are the theoretical and practical considerations when designing signal amplification strategies using SRPX Antibody, Biotin conjugated?

Signal Amplification Design Considerations:

  • Theoretical amplification potential:

    • Each biotinylated antibody can bind multiple streptavidin molecules

    • Each streptavidin has four biotin-binding sites

    • Potential for exponential signal amplification through layered approaches

  • Linear vs. branched amplification strategies:

    • Linear: Biotinylated antibody → Streptavidin-enzyme/fluorophore

    • Branched: Biotinylated antibody → Streptavidin → Biotinylated enzyme → Streptavidin-enzyme

  • Tyramide signal amplification (TSA) integration:

    • Combine with biotin-tyramide substrates for HRP

    • Can achieve 10-100 fold signal enhancement

    • Particularly valuable for low-abundance SRPX detection

  • Amplification vs. signal-to-noise considerations:

    • Higher amplification potentially introduces higher background

    • Optimize blocking to prevent non-specific binding during extended amplification steps

    • Consider spatial resolution limitations with extensive amplification

  • Sequential multi-label experimental design:

    • Plan antibody stripping/quenching between rounds

    • Consider order of detection (detect lowest abundance targets first)

    • Include controls for incomplete stripping/quenching

  • Quantitative analysis adaptations:

    • Establish standard curves with known quantities of recombinant SRPX

    • Determine linear dynamic range of each amplification approach

    • Account for potential signal saturation with high amplification

  • Technical optimization matrix:

Amplification StrategySignal EnhancementBackground RiskSpatial ResolutionComplexityBest Applications
Direct (no amplification)LowLowHighSimpleHigh-abundance targets
Single streptavidin layerModerate (2-5×)LowHighSimpleGeneral detection
Streptavidin-biotin layeringHigh (5-20×)ModerateModerateModerateLow-abundance targets
TSA with biotinylated tyramideVery high (10-100×)HighReducedComplexTrace detection needs

What statistical approaches are most appropriate for analyzing signal variability when using SRPX Antibody, Biotin conjugated in quantitative assays?

Statistical Analysis Framework for Quantitative Assays:

  • Assay validation metrics:

    • Determine intra-assay coefficient of variation (CV) using technical replicates (acceptable: <10%)

    • Calculate inter-assay CV using different experimental days (acceptable: <15%)

    • Establish limits of detection (LOD) and quantification (LOQ) using standard curve analysis

  • Normalization strategies:

    • For Western blots: Normalize to housekeeping proteins or total protein stains

    • For ELISA: Include standard curve on each plate and express results as absolute concentrations

    • For IHC/ICC: Use digital image analysis with appropriate internal controls

  • Outlier analysis:

    • Apply Grubbs' test or Dixon's Q test to identify statistical outliers

    • Consider biological vs. technical variability when interpreting potential outliers

    • Document and report criteria for excluding data points

  • Statistical test selection:

    • For normally distributed data: t-tests (two groups) or ANOVA (multiple groups)

    • For non-normally distributed data: Mann-Whitney U test or Kruskal-Wallis test

    • For paired measurements: Paired t-test or Wilcoxon signed-rank test

  • Advanced statistical considerations:

    • Power analysis to determine appropriate sample size

    • Multiple comparison corrections (Bonferroni, FDR) when testing multiple hypotheses

    • Mixed effects models for nested data structures

  • Reporting guidelines:

    • Include raw data points in addition to mean±SD/SEM

    • Report exact p-values rather than significance thresholds

    • Document all normalization and preprocessing steps

  • Regression analysis for calibration curves:

    • Evaluate linearity across the dynamic range (R² typically >0.98)

    • Consider four-parameter logistic models for sigmoidal dose-response relationships

    • Report goodness-of-fit parameters and prediction intervals

How can researchers differentiate between specific SRPX binding and technical artifacts in imaging applications?

Methodological Approach to Distinguish Specific Binding from Artifacts:

  • Essential technical controls:

    • No primary antibody control: Apply only streptavidin detection reagent

    • Isotype control: Use biotinylated non-specific antibody of same isotype

    • Blocking peptide competition: Pre-incubate antibody with immunizing peptide

    • Endogenous biotin block: Apply avidin/biotin blocking kit before antibody

  • Staining pattern analysis:

    • Subcellular localization: Compare observed pattern with known SRPX distribution

    • Cross-validation: Compare with non-biotinylated SRPX antibody staining patterns

    • Concentration dependence: True signals typically show dose-dependent intensity

    • Consistency across fixation methods: Compare paraformaldehyde vs. methanol fixation

  • Biological validation approaches:

    • Genetic manipulation: Reduced signal in SRPX knockdown/knockout models

    • Treatment-induced changes: Physiologically relevant alterations in SRPX expression

    • Developmental or disease-associated changes: Patterns consistent with known biology

  • Technical artifact recognition:

    • Edge artifacts: Enhanced staining at tissue/cell boundaries

    • Precipitation artifacts: Crystalline deposits with non-biological morphology

    • Trapping artifacts: Non-specific retention in highly structural tissues

    • Fixation artifacts: Altered epitope accessibility due to fixation variability

  • Advanced imaging considerations:

    • Z-stack analysis: Differentiate surface artifacts from internal staining

    • Co-localization with known SRPX interactors: Biological validation

    • Super-resolution techniques: Improved resolution of subcellular distribution

    • Live-cell imaging (where applicable): Temporal dynamics of staining

  • Quantitative image analysis:

    • Signal-to-background ratio calculation for each experimental condition

    • Morphological filtering to exclude non-biological structures

    • Intensity thresholding based on negative control samples

    • Colocalization coefficients with orthogonal markers

What are the emerging applications of SRPX Antibody, Biotin conjugated in single-cell protein profiling technologies?

Emerging Applications in Single-Cell Protein Analysis:

  • Mass cytometry integration:

    • Metal-tagged streptavidin for detection in CyTOF platforms

    • Multiplexed analysis with other protein markers

    • High-dimensional phenotyping of SRPX expression in heterogeneous cell populations

  • Spatial proteomics approaches:

    • Integration with multiplexed ion beam imaging (MIBI)

    • Cyclic immunofluorescence with biotin-streptavidin signal amplification

    • Correlation of SRPX localization with subcellular structures

  • Single-cell western blotting:

    • Microfluidic platforms for protein separation from individual cells

    • Biotin-streptavidin detection for enhanced sensitivity

    • Quantitative analysis of SRPX expression variability across individual cells

  • Proximity ligation assays:

    • Investigation of SRPX protein interactions at single-molecule resolution

    • Biotinylated DNA probes for rolling circle amplification

    • Visualization of specific protein-protein interactions involving SRPX

  • Droplet-based single-cell proteomics:

    • Encapsulation of cells with biotinylated antibodies

    • Barcoding strategies for high-throughput analysis

    • Integration with single-cell transcriptomics for multi-omic profiling

  • CODEX multiplexed imaging:

    • DNA-barcoded streptavidin for iterative detection

    • Integration in highly multiplexed imaging panels (>40 proteins)

    • Spatial relationship analysis between SRPX and other markers

  • Microwell-based approaches:

    • Single-cell capture in microwell arrays

    • In situ detection of secreted vs. cellular SRPX

    • Temporal dynamics of SRPX expression at single-cell resolution

How might cross-linking stabilization techniques improve SRPX epitope preservation during sample processing?

Cross-linking Optimization for SRPX Epitope Preservation:

  • Comparison of cross-linking reagents:

Cross-linkerMechanismAdvantages for SRPX PreservationPotential Limitations
FormaldehydeMethylene bridge formationRapid penetration, reversibleMay mask epitopes containing lysine residues
GlutaraldehydeStrong protein-protein cross-linksExcellent structural preservationHigher background, difficult antigen retrieval
DSS/BS3Amine-specific homobifunctionalPreserves quaternary structuresLimited penetration in tissue samples
EDCZero-length carboxyl to amineMinimal spatial distortionpH-dependent efficiency
Photo-activated cross-linkersLight-induced bindingTemporal control of fixationRequires specialized equipment
  • Sequential fixation strategies:

    • Initial light fixation (0.1-0.5% formaldehyde) to stabilize general structure

    • Secondary targeted cross-linking with epitope-preserving reagents

    • Quenching of excess reactive groups to prevent over-fixation

  • Antigen retrieval optimization:

    • Heat-induced vs. enzymatic epitope retrieval

    • pH gradient testing (pH 6.0, 8.0, 9.0) for optimal SRPX epitope exposure

    • Microwave vs. pressure cooker methods for consistent retrieval

  • Combined approach for membrane proteins:

    • Mild detergent permeabilization before light fixation

    • Cross-linking optimization based on subcellular localization of SRPX

    • Preservation of native lipid environment for transmembrane segment stability

  • Cryofixation alternatives:

    • High-pressure freezing followed by freeze substitution

    • Comparison with chemical fixation methods for epitope accessibility

    • Specialized embedding media for improved section quality

  • Post-fixation blocking strategies:

    • Reduction of free aldehyde groups with sodium borohydride

    • Glycine blocking of unreacted cross-linker

    • Specialized blocking buffers for fixed samples

  • Validation approaches:

    • Direct comparison of staining intensity across fixation methods

    • Western blot analysis of extracted proteins following various fixations

    • Mass spectrometry analysis of preserved vs. modified epitopes

How does the sensitivity and specificity of biotin-streptavidin detection compare with other enzyme-based or fluorescent detection systems for SRPX antibody applications?

Comparative Analysis of Detection Systems:

Detection SystemSensitivityBackgroundQuantitative RangeStabilityBest Applications
Biotin-Streptavidin-HRPVery HighModerate3-4 logsHighWestern blot, ELISA, IHC
Biotin-Streptavidin-APHighLow2-3 logsVery HighChromogenic IHC, ELISA
Biotin-Streptavidin-FluorophoreHighVariable2-3 logsModerate (photobleaching)IF, Flow cytometry
Direct HRP ConjugateModerateLow2 logsHighRapid Western blot, ELISA
Direct Fluorophore ConjugateModerateLow2 logsLow-ModerateFlow cytometry, IF
Tyramide Signal AmplificationExtremely HighHigh (potential)4+ logsHighUltra-sensitive detection

For SRPX antibody applications, biotin-streptavidin detection systems offer several advantages:

The primary limitations include potential endogenous biotin interference and increased protocol complexity compared to direct detection systems.

What are the critical experimental design considerations for multiplexed detection systems incorporating SRPX Antibody, Biotin conjugated?

Experimental Design Considerations for Multiplexed Detection:

  • Panel design strategy:

    • Select complementary markers with biological relevance to SRPX

    • Consider abundance levels of each target (balance high/low expression)

    • Plan detection order based on antibody species and conjugates

  • Spectral separation optimization:

    • Choose fluorophores with minimal spectral overlap

    • Account for tissue autofluorescence profiles

    • Utilize spectral unmixing for closely overlapping signals

  • Sequential staining approaches:

    • Antibody stripping/inactivation between rounds

    • Photobleaching verification before restaining

    • Signal acquisition after each round vs. end-point only

  • Cross-reactivity prevention:

    • Careful blocking between detection steps

    • Use of highly cross-adsorbed secondary reagents

    • Testing for cross-reactivity on single-stained controls

  • Combined approaches with non-antibody probes:

    • Integration with nucleic acid probes (RNA/DNA FISH)

    • Small molecule probes for organelles or functional readouts

    • Click chemistry-based labeling strategies

  • Optimization matrix for biotin-based multiplexing:

ApproachAdvantagesLimitationsBest Practices
Different primary host speciesSimple, directLimited by available hostsComplete secondary absorption
Sequential biotin blockingUses same host speciesComplex, risk of incomplete blockingVerify blocking efficiency
Tyramide deposition + bleachingHigh sensitivitySignal bleed-through riskCareful fluorophore selection
Multi-round imaging with strippingUnlimited targetsPotential epitope damageValidate epitope preservation
Spectral imagingSimultaneous detectionRequires specialized equipmentProper controls for unmixing
  • Quantitative considerations:

    • Standard curves for each biomarker

    • Channel compensation matrices

    • Internal controls for normalization across samples

When incorporating SRPX Antibody, Biotin conjugated into multiplexed systems, researchers should utilize the amplification advantage of biotin-streptavidin while carefully controlling for potential cross-reactivity and background issues.

How can researchers critically evaluate published studies using SRPX antibodies to ensure methodological rigor in their own experimental designs?

Framework for Critical Evaluation of SRPX Antibody Literature:

  • Antibody validation assessment:

    • Determine if antibody specificity was verified using knockout/knockdown controls

    • Check for orthogonal validation methods (mass spectrometry, alternative antibodies)

    • Evaluate if appropriate negative controls were included

  • Methodological transparency evaluation:

    • Complete antibody information (supplier, catalog number, lot, dilution)

    • Detailed protocols for sample preparation and antigen retrieval

    • Clear description of detection systems and imaging parameters

  • Replication and reproducibility indicators:

    • Sample size and power calculations

    • Technical and biological replication strategy

    • Statistical analysis approaches and justification

  • Results interpretation scrutiny:

    • Distinction between correlation and causation in SRPX studies

    • Consideration of alternative explanations for observations

    • Discussion of limitations and potential confounding factors

  • Structured quality assessment checklist:

  • Experimental design transfer considerations:

    • Sample type differences (cell lines vs. tissues vs. organisms)

    • Fixation and processing protocol compatibility

    • Detection system sensitivity needs based on expected SRPX abundance

  • Validation of translational relevance:

    • Correlation of in vitro findings with in vivo models

    • Cross-species conservation of SRPX function and expression

    • Clinical relevance of the experimental conditions

By critically evaluating these aspects of published literature, researchers can design more rigorous experiments using SRPX Antibody, Biotin conjugated and more accurately interpret their results in the context of existing knowledge.

What are the emerging technological trends that may influence future applications of biotin-conjugated antibodies in SRPX research?

Emerging Technological Trends in Biotin-Conjugated Antibody Applications:

  • Single-molecule detection technologies:

    • Super-resolution microscopy for nanoscale SRPX localization

    • Single-molecule pull-down assays for precise interaction analysis

    • Quantum dot-streptavidin conjugates for enhanced sensitivity and stability

  • Microfluidic and organ-on-chip platforms:

    • Real-time monitoring of SRPX expression in controlled microenvironments

    • Integrated detection systems for continuous measurement

    • Single-cell resolution in complex tissue mimetics

  • DNA-barcoded antibody technologies:

    • CODEX multiplexed imaging for spatial protein profiling

    • Antibody-oligonucleotide conjugates for ultra-high-plex detection

    • Integration with spatial transcriptomics for multi-omic analysis

  • AI-assisted image analysis:

    • Automated pattern recognition for SRPX localization

    • Deep learning approaches for multiplexed marker quantification

    • Predictive modeling of SRPX associations with cellular phenotypes

  • Engineered biotin analogs and alternatives:

    • Click chemistry-based conjugation strategies

    • Photoactivatable biotin derivatives for spatiotemporal control

    • Orthogonal binding pairs with biotin-like properties but distinct specificity

  • In vivo applications:

    • Bioorthogonal chemistry for in vivo labeling

    • Near-infrared fluorophore-streptavidin for deep tissue imaging

    • Targeted nanoparticle delivery systems using biotin-streptavidin bridges

  • Integration with multi-omic approaches:

    • Spatial proteogenomics combining antibody detection with nucleic acid analysis

    • Single-cell protein and RNA co-detection systems

    • Computational integration frameworks for multi-modal data

These emerging technologies will expand the applications of biotin-conjugated SRPX antibodies beyond current capabilities, enabling more sensitive, specific, and comprehensive analysis of SRPX biology in complex biological systems.

What represents the current consensus on best practices for utilizing SRPX Antibody, Biotin conjugated across different experimental platforms?

Consensus Best Practices for SRPX Antibody, Biotin Conjugated Applications:

  • Comprehensive validation strategy:

    • Perform Western blot validation to confirm specificity

    • Include appropriate positive and negative controls

    • Validate in relevant experimental systems before proceeding with full studies

  • Application-specific optimization:

    • ELISA: Determine optimal working dilution (typically 1:2000-1:10000)

    • IHC/ICC: Optimize fixation and antigen retrieval for SRPX epitope preservation

    • Flow cytometry: Validate appropriate permeabilization protocols for intracellular SRPX detection

  • Signal amplification calibration:

    • Start with manufacturer's recommended dilution

    • Titrate both biotinylated antibody and streptavidin conjugate

    • Determine optimal signal-to-noise ratio for specific application

  • Storage and handling protocols:

    • Store at -20°C or -80°C in small aliquots

    • Avoid repeated freeze-thaw cycles

    • Prepare working dilutions fresh for each experiment

  • Documentation and reporting standards:

    • Record complete antibody information (manufacturer, catalog number, lot)

    • Document all experimental conditions in detail

    • Present both positive and negative controls alongside experimental data

  • Endogenous biotin management:

    • Apply biotin blocking steps for tissues with high endogenous biotin

    • Consider alternative detection systems if endogenous biotin issues persist

    • Include biotin-blocked negative controls

  • Data interpretation guidelines:

    • Interpret results in context of biological knowledge about SRPX

    • Apply appropriate statistical analysis for quantitative measurements

    • Verify findings with orthogonal detection methods when possible

These consensus best practices represent the current standard in the field, though specific protocols may require adaptation based on particular experimental systems and research questions.

What future directions are likely to advance our understanding of SRPX biology through improved antibody technologies?

Future Directions in SRPX Antibody Technology and Research:

  • Development of conformation-specific antibodies:

    • Antibodies targeting specific structural states of SRPX protein

    • Tools to distinguish between active/inactive or complexed/free SRPX

    • Enabling studies of SRPX conformational dynamics in different cellular contexts

  • Single-domain antibody development:

    • Nanobodies or single-domain antibodies against SRPX epitopes

    • Improved penetration in tissues and access to restricted cellular compartments

    • Enhanced multiplexing capabilities due to smaller size

  • Degradation-resistant antibody formats:

    • Engineered antibody fragments with enhanced stability

    • Resistance to proteolytic degradation in challenging sample types

    • Extended shelf-life and experimental reliability

  • Site-specific biotin conjugation:

    • Precisely controlled biotin:antibody ratios

    • Oriented biotin presentation for optimal streptavidin binding

    • Reduced impact on antibody paratope function

  • Integrated technological approaches:

    • Combining antibody detection with CRISPR screening for functional studies

    • Spatial multi-omics integration of protein, RNA, and epigenetic data

    • Temporal dynamics of SRPX expression and localization in living systems

  • Systems biology integration:

    • Network analysis of SRPX interactions and pathways

    • Computational modeling of SRPX function in cellular processes

    • Predictive frameworks for SRPX involvement in disease mechanisms

  • Translational applications:

    • Development of SRPX as a potential biomarker for specific conditions

    • Therapeutic targeting strategies based on SRPX biology

    • Diagnostic tools utilizing advanced SRPX detection methods

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.