ST5 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Biological Relevance

ST5 (Suppression of Tumorigenicity 5) is a protein encoded by the ST5 gene, implicated in tumor suppression and cellular signaling. It interacts with MAPK1/ERK2 kinase and modulates tumorigenic phenotypes . The HRP-conjugated ST5 antibody is a polyclonal antibody (rabbit-derived) that binds specifically to human ST5, facilitating its detection via enzymatic signal amplification .

Conjugation Methodology

The conjugation process often involves:

  1. Activation of HRP: Oxidation of carbohydrate moieties on HRP using sodium periodate to generate aldehyde groups .

  2. Lyophilization: Freeze-drying activated HRP to enhance binding capacity and stability .

  3. Antibody Binding: Conjugation of lyophilized HRP to ST5 antibodies under controlled pH (6.5–8.5) and buffer conditions .

Critical Buffer Requirements for Conjugation4:

ComponentRecommended Level
pH6.5–8.5
BSA<0.1%
Tris<50 mM
Glycerol<50%

Applications and Performance

ST5 Antibody, HRP conjugated is validated for:

Key Applications

  • ELISA: Effective at dilutions up to 1:10,000 .

  • Western Blot: Detects endogenous ST5 at ~60 kDa .

Example Protocol (ELISA)

  1. Coating: Immobilize antigen (e.g., His-tagged protein) at 1 μg/mL .

  2. Detection: Use ST5-HRP conjugate at 0.2–2 μg/mL .

  3. Signal Development: Employ substrates like TMB or DAB for colorimetric detection .

Research Findings and Advancements

  • Enhanced Sensitivity: Lyophilization during conjugation increases HRP binding capacity, improving ELISA sensitivity (1:5000 dilution vs. 1:25 in classical methods) .

  • Functional Validation: ST5-HRP conjugate successfully detects recombinant LAG-3/CD223 protein in Western blot .

  • Thermal Stability: Conjugates retain activity after lyophilization and long-term storage at 4°C .

Limitations and Future Directions

  • Species Restriction: Limited to human samples in current formulations .

  • Research Gaps: Broader studies needed to validate the method across diverse IgG antibodies .

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 orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For specific delivery timelines, please consult your local distributors.
Synonyms
DENND2B antibody; DENN domain containing protein 2B antibody; DENN domain-containing protein 2B antibody; DENN/MADD domain containing 2B antibody; HeLa tumor suppression 1 antibody; HTS1 antibody; p126 antibody; ST 5 antibody; ST5 antibody; ST5_HUMAN antibody; Suppression of tumorigenicity 5 antibody; Suppression of tumorigenicity 5 protein antibody
Target Names
ST5
Uniprot No.

Target Background

Function
ST5 may play a role in cytoskeletal organization and tumorigenesis. It appears to be involved in a signaling transduction pathway leading to the activation of MAPK1/ERK2. ST5 plays a role in EGFR trafficking from recycling endosomes back to the cell membrane. ST5 functions as a guanine nucleotide exchange factor (GEF) that may activate RAB9A and RAB9B. It promotes the exchange of GDP to GTP, converting inactive GDP-bound Rab proteins into their active GTP-bound form. ST5 may block ERK2 activation stimulated by ABL1 (likely). It may also alter cell morphology and cell growth (likely).
Gene References Into Functions
  1. Research has shown that intersectin-s binds DENND2B, a guanine nucleotide exchange factor for the exocytic GTPase Rab13. This interaction promotes the recycling of ligand-free EGFR to the cell surface. This finding reveals a novel mechanism for controlling the fate of internalized EGFR, which has significant implications for cancer. PMID: 29030480
  2. DENND2B interacts with the Rab13 effector MICAL-L2 at the cell periphery. This interaction is essential for the dynamic remodeling of the cell's leading edge. PMID: 25713415
  3. Using RNA in situ hybridization in mice, researchers found that St5 is expressed in the frontal cortex during embryonic development. In adult mouse brains, expression of St5 was particularly high in the hippocampal area and cerebellum. PMID: 19843505
Database Links

HGNC: 11350

OMIM: 140750

KEGG: hsa:6764

STRING: 9606.ENSP00000319678

UniGene: Hs.117715

Subcellular Location
[Isoform 1]: Cytoplasm, cell cortex. Cell membrane. Recycling endosome.
Tissue Specificity
Widely expressed with the exception of peripheral blood lymphocytes. Isoform 1 is expressed in several epithelial and fibroblast (including tumorigenic) but absent in lymphoid cell lines (at protein level). Isoform 3 is expressed in primary cell or weakly

Q&A

What is ST5 protein and why is it an important research target?

ST5 (Suppression of tumorigenicity 5) protein, also known as DENND2B or HTS1, functions as a DENN domain-containing protein involved in signal transduction pathways . The protein (UniProt ID: P78524) has been studied for its potential tumor suppressive properties, making ST5 antibodies valuable tools for investigating cancer biology and signaling mechanisms. ST5 antibodies help researchers detect, quantify, and localize this protein in various experimental systems, with HRP conjugation providing a direct enzymatic detection capability without requiring secondary antibody steps .

What are the optimal storage conditions for maintaining ST5 antibody-HRP conjugate activity?

For maximum retention of both immunological specificity and enzymatic activity, ST5 antibody-HRP conjugates should be stored at -20°C to -80°C for long-term preservation . The antibody is typically supplied in a stabilizing buffer containing glycerol (50%), which prevents freeze-thaw damage, along with phosphate-buffered saline (0.01M, pH 7.4) and preservatives like Proclin 300 (0.03%) . While refrigeration at 4°C is suitable for short-term storage (1-2 weeks), repeated freeze-thaw cycles should be strictly avoided as they significantly diminish both antibody binding capacity and enzymatic activity of the HRP moiety . For working solutions, aliquoting into single-use volumes before freezing is strongly recommended. Activity assessment through small-scale test runs is advisable after extended storage periods or when using older lots.

What are the optimal conditions for using ST5 antibody-HRP conjugate in Western blot applications?

For optimal Western blot results with ST5 antibody-HRP conjugate, protocols should be carefully optimized across multiple parameters. Sample preparation should include complete protein denaturation using standard SDS buffer with DTT or β-mercaptoethanol heating at 95°C for 5 minutes, as the ST5 protein (UniProt ID: P78524) may form complexes that affect epitope accessibility . The recommended antibody dilution range starts at 1:1000 in blocking buffer (5% non-fat milk or BSA in TBST) . Incubation should be performed at 4°C overnight with gentle rocking for maximum sensitivity, followed by 4-5 stringent washes with TBST to minimize background .

Detection should utilize enhanced chemiluminescence (ECL) substrates matched to experiment sensitivity requirements - standard ECL for abundant targets or enhanced/super-sensitive variants for low-abundance detection . When troubleshooting weak signals, consider extending exposure times, increasing antibody concentration (up to 1:500), or using signal enhancers containing oxidation stabilizers to prolong HRP activity. Importantly, HRP activity is inhibited by azides, cyanides, and sulfides, so buffers containing these compounds must be avoided throughout the experimental workflow .

How can I optimize the ST5 antibody-HRP conjugate for ELISA applications?

Optimizing ST5 antibody-HRP conjugate for ELISA requires systematic evaluation of multiple parameters to achieve maximum sensitivity and specificity. Begin with coating plates using purified recombinant ST5 protein (3-280AA region) for direct ELISA or appropriate capture antibodies for sandwich ELISA formats . Antibody titration is essential - test dilutions from 1:500 to 1:5000 in manufacturer-recommended or optimized diluent to determine the minimum concentration providing maximum signal-to-noise ratio .

The substrate selection significantly impacts detection sensitivity: TMB (3,3',5,5'-tetramethylbenzidine) provides excellent sensitivity with blue color development (changing to yellow after stop solution addition), while ABTS (2,2'-azino-di-[3-ethylbenzthiazoline-6-sulfonic acid]) offers a more stable green endpoint . Optimize incubation parameters thoroughly - most protocols recommend room temperature for 1-2 hours or 4°C overnight, followed by substrate development for precisely timed intervals (typically 5-30 minutes depending on concentration) . For quantitative work, always include a standard curve using recombinant ST5 protein at 7-8 concentration points with at least duplicate measurements. Signal saturation should be avoided through appropriate dilution selection to ensure readings fall within the linear range of detection.

What controls are essential when using ST5 antibody-HRP conjugate in immunoassays?

A comprehensive control strategy is crucial for meaningful interpretation of results when working with ST5 antibody-HRP conjugate. Primary negative controls should include: (1) isotype-matched HRP-conjugated control antibodies to assess non-specific binding, (2) samples known to be negative for ST5 expression to establish baseline signal, and (3) blocking peptide competition controls where excess immunogen peptide (recombinant human ST5 protein, 3-280AA) pre-incubated with the antibody should eliminate specific signal .

Essential positive controls include: (1) validated ST5-expressing cell lines or tissues, (2) recombinant ST5 protein for direct detection assays, and (3) previously validated detection methods for comparison . Technical controls should evaluate the integrity of the HRP conjugate by including: (1) enzyme activity controls using HRP substrate directly on a small antibody aliquot, (2) secondary-only controls where appropriate to distinguish conjugate-specific signal from potential endogenous peroxidase activity, and (3) stepwise omission controls to identify protocol steps contributing to background . For multiplex assays, single-marker controls are essential to rule out signal bleed-through or unexpected cross-reactivity with other detection systems.

What are the most common causes of high background when using ST5 antibody-HRP conjugates, and how can they be addressed?

High background with ST5 antibody-HRP conjugates typically stems from several identifiable sources that can be systematically addressed. Endogenous peroxidase activity in samples (particularly in tissues or cells with high peroxidase content) requires effective quenching using 0.3-3% hydrogen peroxide treatment for 10-30 minutes before antibody application . Non-specific antibody binding can be minimized by optimizing blocking conditions - testing different blocking agents (BSA, casein, normal serum, commercial blockers) at various concentrations (1-5%) and ensuring adequate blocking time (1-2 hours at room temperature) .

Insufficient washing represents another common cause of high background - protocols should incorporate at least 4-5 washes with appropriate buffer (TBST or PBST) after antibody incubation, with each wash lasting 5-10 minutes with gentle agitation . Antibody concentration must be titrated carefully as excess conjugate significantly contributes to background - typical starting dilutions around 1:1000 should be adjusted based on signal-to-noise performance . For tissue sections or cell preparations, evaluating fixation protocols is crucial as overfixation can increase non-specific HRP binding while inadequate fixation may produce artifactual staining patterns. Finally, substrate incubation requires careful timing control as extended development periods to compensate for weak signals often result in increased background.

How can I determine if my ST5 antibody-HRP conjugate has maintained its dual functionality after storage?

Assessing the dual functionality (both antigen recognition and enzymatic activity) of stored ST5 antibody-HRP conjugates requires a two-pronged approach. For enzymatic activity evaluation, perform a simple HRP activity test using a small aliquot (1-2 μl) of the conjugate added directly to TMB or other HRP substrate - rapid color development indicates preserved peroxidase function . Quantitative comparison to a reference standard or previous lot can provide more precise activity measurements by spectrophotometric analysis of reaction kinetics.

Immunological functionality should be assessed using known positive controls such as recombinant ST5 protein or validated ST5-expressing cell lysates (referencing UniProt ID: P78524) . Comparative testing against fresh conjugate or previously validated detection methods using identical samples provides the most reliable assessment. When both activities need simultaneous evaluation, dot blot analysis offers an efficient approach - spotting known quantities of target protein on membranes followed by antibody incubation and substrate development allows rapid assessment of both binding specificity and signal generation capacity . For quantitative applications, developing a standardized quality control workflow with defined acceptance parameters ensures experimental reproducibility and comparability across studies.

What strategies can be employed when ST5 antibody-HRP conjugate shows weak signal in Western blots?

When encountering weak signals with ST5 antibody-HRP conjugate in Western blots, multiple enhancement strategies can be implemented to improve detection sensitivity. Sample preparation modifications include increasing protein loading (up to 50-100 μg per lane), using protein enrichment techniques (immunoprecipitation, subcellular fractionation), and ensuring complete protein denaturation to fully expose epitopes . Transfer efficiency should be verified using reversible total protein stains such as Ponceau S before immunoblotting.

Antibody incubation conditions can be optimized by increasing concentration (using dilutions up to 1:500), extending incubation time to overnight at 4°C, and adding signal enhancers such as 5% polyethylene glycol to the antibody diluent . Detection system enhancements include switching to high-sensitivity ECL substrates designed for femtogram-level detection, utilizing signal accumulation technology with longer exposure times, or considering alternative detection methods like near-infrared fluorescence if HRP activity appears compromised .

Technical modifications such as reducing membrane pore size (0.22 μm PVDF maintains small proteins better than 0.45 μm), optimizing transfer conditions (longer transfer times or lower amperage for larger proteins), and using low-fluorescence PVDF for reduced background can significantly improve signal-to-noise ratio . When working with challenging samples, consider native conditions if protein conformation affects epitope accessibility, or test alternative sample preparation buffers that may better preserve the target epitope.

How can ST5 antibody-HRP conjugate be effectively integrated into multiplex detection systems?

Integrating ST5 antibody-HRP conjugate into multiplex detection systems requires strategic approaches to overcome the limitations of single-enzyme detection. For chromogenic multiplexing, sequential detection protocols can be implemented using HRP substrates producing distinct colors (DAB for brown, AEC for red, TMB for blue) with complete inactivation steps between cycles using hydrogen peroxide treatment . This approach allows discrimination between multiple targets on the same sample but requires careful optimization of each detection cycle.

For fluorescence-based multiplexing, tyramide signal amplification (TSA) offers significant advantages - the HRP conjugate catalyzes deposition of fluorophore-labeled tyramide molecules, which can be performed with different fluorophores in sequential rounds after HRP inactivation . This technique provides substantially higher sensitivity than direct fluorophore conjugation and enables detection of multiple targets with clear spectral separation. The ST5 antibody-HRP conjugate can be combined with other enzyme-conjugated antibodies (such as alkaline phosphatase) targeting different proteins to achieve dual colorimetric detection in a single assay .

Mass cytometry approaches represent the most advanced multiplexing option - although traditionally using metal-conjugated antibodies, HRP-conjugated antibodies can be utilized in sequential metal deposition protocols where the enzymatic activity precipitates specific metal ions onto the bound antibody location . For any multiplexing approach, comprehensive validation using single-target controls is essential to ensure signal specificity and rule out cross-reactivity between detection systems.

What considerations are important when using ST5 antibody-HRP conjugate for studying protein-protein interactions?

When employing ST5 antibody-HRP conjugate for protein-protein interaction studies, several critical factors must be addressed to ensure valid results. Epitope accessibility represents a primary concern - the antibody targets a specific region (amino acids 3-280) of the ST5 protein (UniProt ID: P78524), so researchers must consider whether potential interaction partners might mask this region . Preliminary structural analysis or epitope mapping can help predict such interference.

For co-immunoprecipitation applications, gentler lysis conditions (non-ionic detergents like NP-40 or Triton X-100 at 0.1-1%) help preserve protein complexes, but may require optimization to balance complex stability with antibody accessibility . The direct detection capability of HRP conjugates offers advantages in proximity-dependent enzymatic assays where HRP can catalyze biotinylation or other modifications of proteins in close proximity to ST5, allowing identification of interaction partners without relying on antibodies for each target.

Controls are particularly crucial in interaction studies - competitive blocking with immunizing peptide confirms signal specificity, while isotype controls rule out non-specific binding . Reciprocal co-immunoprecipitation using antibodies against suspected interaction partners provides validation of detected interactions. When used in combination with cross-linking techniques, ST5 antibody-HRP conjugate can help distinguish between direct and indirect interactions through careful analysis of complex composition under different stabilization conditions. Researchers should also consider how the HRP moiety (44kDa) might sterically hinder or affect certain protein-protein interactions, particularly in native non-denaturing systems.

How can ST5 antibody-HRP conjugate be adapted for high-throughput screening applications?

Adapting ST5 antibody-HRP conjugate for high-throughput screening (HTS) requires optimization for automation compatibility, miniaturization, and robust signal generation. Microplate-based ELISA represents the most straightforward adaptation, with the conjugate directly detecting ST5 protein in 96, 384, or 1536-well formats . Protocol modifications for HTS include reduced volumes (maintaining concentrations while minimizing reagent consumption), accelerated incubation times through increased temperature or orbital shaking, and selection of rapid-developing HRP substrates compatible with automated plate readers.

For more advanced applications, the conjugate can be adapted to bead-based multiplex assays where ST5 antibody-HRP is used to detect its target on uniquely identifiable microspheres, allowing simultaneous measurement of multiple analytes in each sample well . Microfluidic platforms offer another HTS-compatible format, with the HRP conjugate incorporated into miniaturized immunoassay channels where small sample volumes (nL-μL range) can be rapidly analyzed with significant reagent savings.

Signal detection optimization is crucial for HTS - chemiluminescent HRP substrates provide excellent sensitivity for plate reader detection, while colorimetric substrates offer stability advantages for batch processing . Statistical validation becomes particularly important in HTS contexts - establishing Z-factor values (ideally >0.5) through extensive positive and negative control testing ensures assay robustness across large sample sets. Finally, automated data analysis pipelines should include quality control metrics to flag wells with anomalous signal patterns, ensuring reliable hit identification despite the throughput scale.

How should researchers quantitatively analyze results obtained with ST5 antibody-HRP conjugate?

Quantitative analysis of results obtained with ST5 antibody-HRP conjugate requires rigorous standardization and careful consideration of the detection system's characteristics. For Western blot densitometry, signal linearity must be established across the range of expected protein concentrations by creating standard curves with recombinant ST5 protein . Normalization to housekeeping proteins such as β-actin (detected with antibodies like CST #5125) is essential, but researchers should verify that treatments don't affect these reference proteins .

For ELISA quantification, standard curves using purified recombinant ST5 protein (3-280AA region) should be prepared with 7-8 concentration points in at least duplicate measurements . Four-parameter logistic regression typically provides better curve fitting than linear models for ELISA data. The limits of detection (LOD) and quantification (LOQ) should be formally established through statistical analysis of background signals (typically LOD = mean background + 3SD; LOQ = mean background + 10SD) .

Signal normalization strategies differ by application: for cell-based assays, normalization to cell number (using DNA content or cell counting) provides more reliable standardization than protein content when cell size may vary . For tissue samples, considering section thickness, tissue area analyzed, and cell density improves comparison accuracy. When developing novel quantitative protocols, method validation should include precision assessment (intra- and inter-assay CV%), accuracy evaluation (spike-and-recovery experiments), and linearity testing (dilutional parallelism) to ensure robust quantitative performance across the analytical range.

What approaches help distinguish between specific and non-specific signals when using ST5 antibody-HRP conjugate?

Distinguishing specific from non-specific signals requires implementing multiple complementary validation strategies. Immunological validation should include competitive inhibition experiments where excess immunizing peptide (recombinant human ST5 protein, 3-280AA) pre-incubated with the antibody should eliminate specific signals while leaving non-specific background unaffected . Isotype control experiments using irrelevant HRP-conjugated antibodies of the same isotype (rabbit IgG-HRP) administered at identical concentrations help establish the baseline non-specific binding level .

Technical controls to differentiate signal sources include enzyme activity assessment through selective peroxidase inhibition (using sodium azide or other specific inhibitors) to distinguish between conjugate activity and endogenous peroxidase signals . Signal pattern analysis provides additional evidence - specific ST5 staining should correspond to expected subcellular localization patterns and show consistency across technical replicates, while non-specific signals often present as diffuse background or edge artifacts.

Advanced validation approaches include correlation with orthogonal detection methods (e.g., mass spectrometry, RNA expression) to confirm that signals correspond to actual ST5 protein levels . When working with tissues or complex samples, signal absorption testing (pre-absorbing the antibody with tissues or lysates from known negative sources) can reduce cross-reactivity. For quantitative applications, dose-response relationships provide compelling evidence for specificity - signals that change proportionally with known ST5 concentration changes suggest specific detection, while non-specific background typically remains constant regardless of target concentration.

How can researchers effectively compare ST5 protein expression data across different experimental systems?

Effective comparison of ST5 protein expression across different experimental systems requires standardization at multiple levels. For quantitative comparisons, absolute quantification using purified recombinant ST5 protein standards can provide system-independent measurements, preferably expressed as molar concentrations or molecules per cell rather than arbitrary units . When absolute quantification isn't feasible, relative quantification using identical reference standards across all experiments enables normalized comparisons.

Technical standardization is equally important - developing a standard operating procedure (SOP) that specifies key parameters (antibody lot, dilution, incubation conditions, detection system, image acquisition settings) minimizes method-induced variability . For Western blot comparisons, loading the same positive control lysate on each blot and normalizing experimental samples to this reference improves inter-blot comparability . In ELISA and other plate-based formats, including identical control samples on each plate enables calculation of plate correction factors to account for run-to-run variability .

Cross-platform normalization requires careful validation - when comparing different detection methods (e.g., ELISA vs. Western blot), performing method comparison studies with identical samples helps establish conversion factors . For tissue analysis, considering cellularity differences through normalization to cell-specific markers rather than total protein improves biological relevance. Finally, statistical approaches such as z-score normalization or quantile normalization can be applied to datasets from different experimental systems to enable integrated analysis while acknowledging the technical limitations of cross-system comparisons.

What are the key performance characteristics of ST5 antibody-HRP conjugate compared to other detection methods?

ST5 antibody-HRP conjugate offers distinct performance advantages and limitations compared to alternative detection approaches. Sensitivity analysis reveals that HRP-conjugated antibodies typically provide detection limits in the low picogram to high femtogram range (approximately 10^-12 to 10^-15 moles) when used with enhanced chemiluminescent substrates, comparable to fluorophore-conjugated antibodies but without requiring specialized detection equipment . The signal amplification inherent in the enzymatic reaction provides superior sensitivity compared to direct immunofluorescence methods.

The following table compares key performance aspects across detection methods:

Performance ParameterST5 Antibody-HRP ConjugateUnconjugated Primary + HRP-SecondaryFluorophore-Conjugated AntibodyMass Cytometry (Metal-Conjugated)
Sensitivity (Approximate Detection Limit)1-10 pg0.5-5 pg5-50 pg0.1-1 pg
Protocol Duration4-5 hours6-8 hours3-4 hours8-10 hours
Signal Persistence1-30 minutes (substrate dependent)1-30 minutes (substrate dependent)Months (if protected from light)Permanent
Equipment RequirementsStandard chemiluminescence imagerStandard chemiluminescence imagerFluorescence microscope/scannerMass cytometer (specialized)
Multiplexing CapacityLimited (sequential only)Limited (sequential only)High (4-6 simultaneous)Very high (30+ simultaneous)
Relative Cost Per AssayModerateLowModerate-HighVery High

What factors affect the enzyme kinetics of HRP in ST5 antibody-HRP conjugates, and how do they impact experimental design?

The enzymatic activity of HRP in ST5 antibody conjugates is governed by multiple factors that significantly influence experimental design and detection sensitivity. Substrate concentration exhibits typical Michaelis-Menten kinetics with the optimal working range dependent on the specific substrate - TMB typically shows higher sensitivity with Km values in the 0.1-0.3 mM range, while DAB substrates may require higher concentrations for equivalent signal development . Temperature dramatically affects reaction kinetics, with activity approximately doubling with each 10°C increase between 4-40°C, making temperature control critical for quantitative applications.

pH conditions substantially impact HRP activity, with the enzyme showing optimal performance between pH 6.0-6.5 for most substrates, though enhanced chemiluminescent (ECL) systems often operate optimally at higher pH ranges (7.5-8.5) . Buffer composition effects are equally important - phosphate buffers at concentrations above 20mM can inhibit HRP activity, while certain detergents may either enhance (low concentrations of Tween-20) or inhibit (ionic detergents) enzymatic function .

The relationship between incubation time and signal development follows non-linear patterns, with initial rapid signal increase followed by plateauing as substrate depletion occurs. This necessitates careful timing optimization, particularly for quantitative applications. Inhibitory compounds present in biological samples (azides, cyanides, sulfides) can dramatically reduce HRP activity, requiring sample pretreatment when these compounds cannot be avoided . For experimental design, these kinetic considerations dictate optimal substrate selection, development timing, and signal detection parameters that balance sensitivity requirements against practical workflow constraints and equipment limitations.

How does the degree of HRP conjugation affect antibody performance, and what are the optimal ratios?

The degree of HRP conjugation to ST5 antibody fundamentally influences multiple performance parameters, creating a critical balance between enhanced detection capability and preserved immunoreactivity. The molar ratio of HRP to antibody typically ranges from 2:1 to 4:1 in commercial conjugates, with higher ratios providing increased sensitivity up to a threshold, beyond which performance declines . Excessive conjugation (>6 HRP molecules per antibody) frequently leads to decreased immunoreactivity through steric hindrance or epitope masking, particularly when conjugation chemistry preferentially targets amino groups in or near the antigen binding region.

Optimal conjugation ratios vary by application - for enzyme immunoassays where detection sensitivity is paramount, higher HRP:antibody ratios (3-4:1) may be preferred despite some loss of binding affinity . For applications requiring precise epitope recognition or working with structurally complex antigens, lower conjugation ratios (1-2:1) better preserve binding characteristics. Researchers should consider that while manufacturers optimize conjugation chemistry for balanced performance, custom conjugation services can provide tailored HRP:antibody ratios optimized for specific experimental requirements when working with challenging targets or specialized detection systems .

How might advances in bioconjugation chemistry improve next-generation ST5 antibody-HRP conjugates?

Emerging bioconjugation technologies promise significant improvements for next-generation ST5 antibody-HRP conjugates through enhanced control over attachment chemistry and conjugate architecture. Site-specific conjugation using engineered antibodies with incorporated unnatural amino acids (such as p-azidophenylalanine) enables precise control over HRP attachment locations, potentially preserving the ST5 binding domain while optimizing HRP orientation for maximum enzymatic efficiency . Enzymatic conjugation methods utilizing sortase or transglutaminase enzymes offer similar site-specificity without requiring antibody engineering, allowing controlled HRP attachment at defined positions away from antigen-binding regions.

Click chemistry approaches (copper-free strain-promoted azide-alkyne cycloaddition) provide conjugation advantages through highly specific, orthogonal reactions that proceed rapidly under physiological conditions without damaging either protein component . These methods could enable production of more homogeneous ST5 antibody-HRP conjugates with consistent HRP:antibody ratios and orientations, improving batch-to-batch reproducibility. Linker technology advancements, including cleavable linkers that release HRP upon environmental triggers (pH changes, reducing conditions, enzymatic cleavage), could create "smart conjugates" that activate only in specific cellular compartments or under particular conditions.

The integration of recombinant HRP variants with enhanced catalytic efficiency, increased stability, or modified substrate specificity would further advance detection capabilities . These engineered enzymes, when site-specifically attached to optimized recombinant ST5 antibodies, could dramatically improve sensitivity while maintaining the convenience of direct detection. Combined with computational antibody engineering to identify optimal conjugation sites that maximize both binding affinity and enzymatic accessibility, these approaches represent the frontier of antibody-enzyme conjugate development for next-generation research applications.

What potential applications exist for ST5 antibody-HRP conjugates in emerging single-cell analysis techniques?

ST5 antibody-HRP conjugates hold substantial potential for adaptation to emerging single-cell analysis platforms through innovative detection strategies. In microfluidic single-cell western blotting, the HRP conjugate could enable direct detection of ST5 protein in cellular lysates separated within microchambers, facilitating protein expression profiling at single-cell resolution . The signal amplification inherent in HRP enzymatic activity is particularly valuable for detecting low-abundance signaling proteins like ST5 in individual cells, where protein quantities may fall below the detection limits of fluorescence-based methods.

For spatial proteomics applications, the conjugate could be integrated into multiplexed ion beam imaging (MIBI) or imaging mass cytometry workflows through metal deposition techniques, where HRP activity catalyzes the precipitation of lanthanide metals that can be detected by mass spectrometry . This approach would allow visualization of ST5 protein distribution within tissue architectures at subcellular resolution while preserving spatial context. In cyclic immunofluorescence (CycIF) applications, tyramide signal amplification using the ST5 antibody-HRP conjugate could substantially enhance detection sensitivity for rare or low-abundance targets.

Single-cell secretion analysis represents another promising application, where the conjugate could detect secreted ST5 protein variants captured on antibody-coated surfaces in techniques like microengraving or droplet-based secretion assays . The catalytic amplification provided by HRP enables detection of extremely low protein quantities secreted by individual cells. Additionally, the development of photocatalytic properties of HRP opens possibilities for light-controlled enzymatic reactions at single-cell resolution, potentially enabling selective labeling or modification of cells based on their ST5 expression patterns. These applications would benefit from ongoing optimization of conjugation chemistry to maximize sensitivity while minimizing steric effects that might compromise antigen recognition in spatially constrained single-cell contexts.

How might computational approaches enhance the design and application of ST5 antibody-HRP conjugates in complex research systems?

Computational approaches offer transformative potential for optimizing ST5 antibody-HRP conjugates through multiple dimensions of analysis and prediction. Molecular dynamics simulations can model the conformational impact of HRP conjugation on antibody structure, predicting how different attachment sites affect binding kinetics and accessibility of the ST5 epitope (amino acids 3-280) . These simulations enable rational design of conjugates with optimal spatial arrangements that maximize both enzymatic activity and immunological specificity while minimizing steric hindrance.

Machine learning algorithms trained on experimental performance data could predict optimal conjugation parameters (HRP:antibody ratio, linker length, attachment chemistry) for specific applications, accelerating optimization processes that currently require extensive empirical testing . Automated image analysis using deep learning approaches can enhance signal quantification from HRP-generated signals, improving discrimination between specific staining and background while enabling more sensitive detection of subtle expression differences across experimental conditions or biological samples.

Systems biology integration represents another computational frontier - correlating ST5 protein detection data from antibody-HRP conjugates with transcriptomic, proteomic, and metabolomic datasets to place findings within broader biological contexts . This multi-omics integration helps researchers understand how ST5 protein functions within signaling networks and how its expression patterns correlate with other biological parameters. Additionally, computational modeling of reaction kinetics can optimize experimental protocols by predicting signal development profiles under various conditions, allowing researchers to determine ideal substrate concentrations, incubation times, and detection parameters for specific sensitivity requirements. These computational approaches collectively transform conjugate development from empirical trial-and-error into hypothesis-driven rational design, significantly accelerating research progress in understanding ST5 biology.

How does ST5 antibody-HRP conjugate compare with fluorophore-conjugated alternatives for different research applications?

The comparison between ST5 antibody-HRP and fluorophore conjugates reveals distinct advantages for specific research contexts. HRP conjugates excel in applications requiring signal amplification, as the enzymatic reaction can generate thousands of detectable molecules per antibody, providing detection limits in the low picogram to high femtogram range for Western blots and ELISAs . This amplification makes HRP conjugates particularly valuable for detecting low-abundance targets like ST5 in limited samples or when target expression is minimal.

Equipment requirements represent another key differentiator - HRP detection requires only basic laboratory equipment (standard light microscopes for chromogenic detection, widely available chemiluminescence imagers for enhanced sensitivity), while fluorophore detection necessitates specialized fluorescence microscopes or scanners with appropriate excitation sources and filter sets . The signal stability profile differs significantly - HRP signals are transient (minutes to hours depending on substrate) but can be permanently preserved through precipitation reactions (DAB), while fluorophore signals can persist for months when properly protected from photobleaching.

The following table summarizes the comparative advantages for different research applications:

ApplicationST5 Antibody-HRP AdvantageFluorophore Conjugate Advantage
Western BlottingHigher sensitivity with chemiluminescent detection; permanent record possible; stripping and reprobing feasibleDirect digital quantification; linear dynamic range; multiplexing capability with different fluorophores
ImmunohistochemistrySignal amplification for low-expression targets; permanent staining; conventional microscopy compatibleMultiplexing with other markers; no substrate development required; superior colocalization analysis
Flow CytometryNot typically usedStandard detection method; multiplexing capability; quantitative signal proportional to expression
Live Cell ImagingNot compatibleEnables real-time monitoring; subcellular trafficking studies; minimal interference with cellular processes
High-Content ScreeningCompatible with colorimetric plate readers; robust assay developmentSuperior multiplexing; subcellular resolution; direct quantification without substrate development

What are the comparative advantages of directly conjugated ST5 antibody-HRP versus primary-secondary antibody detection systems?

Directly conjugated ST5 antibody-HRP offers distinct workflow and performance advantages compared to two-step detection systems using unconjugated primary antibodies followed by HRP-labeled secondary antibodies. Protocol efficiency represents a primary advantage - direct conjugates eliminate the 1-2 hour secondary antibody incubation and associated wash steps, reducing total protocol time by approximately 25-40% while minimizing hands-on requirements . This time efficiency becomes particularly valuable in high-throughput or time-sensitive research contexts.

Cross-reactivity elimination provides another significant advantage for complex samples - direct conjugates avoid potential non-specific binding from secondary antibodies, which is particularly problematic in multi-species samples or tissues with endogenous immunoglobulins . This specificity enhancement is especially valuable when studying ST5 in heterogeneous tissue environments or samples containing multiple species components. Signal-to-noise ratios typically improve with direct conjugates due to the elimination of secondary antibody background contribution, though this must be balanced against the signal amplification that secondary systems provide through multiple secondary antibodies binding each primary.

The following comparison table highlights key differences between the approaches:

ParameterST5 Antibody-HRP Direct ConjugatePrimary ST5 + Secondary-HRP System
Protocol Duration4-5 hours6-8 hours
Reagent Cost Per AssayHigher initial cost, lower per-assay cost for high-volume usersLower initial cost, higher reagent consumption
SensitivityModerate to high (depends on conjugation ratio)Higher (due to signal amplification from multiple secondaries)
Background in Complex SamplesLower (no secondary cross-reactivity)Potentially higher (secondary antibody cross-reactivity)
Species FlexibilityLimited to one species systemSecondary antibodies available for multiple host species
Quantitative LinearityExcellent (direct relationship to target abundance)Good, but affected by primary-secondary binding variability
Batch-to-Batch ConsistencyModerate (conjugation process variability)High (primary and secondary can be QC tested separately)
Multiplexing CapabilityLimited (one conjugate per experiment without sequential protocols)Better (different primaries can be detected with same secondary)

Under what conditions would mass spectrometry-based approaches be preferred over ST5 antibody-HRP detection?

Mass spectrometry (MS) approaches offer distinct advantages over ST5 antibody-HRP immunodetection in specific research contexts, particularly when experimental questions extend beyond simple target verification. Comprehensive protein characterization represents a primary advantage - MS can simultaneously identify post-translational modifications, sequence variants, and isoforms of ST5 protein, providing detailed molecular characterization that antibody-based methods cannot achieve . This becomes crucial when investigating how modifications affect ST5 function or when studying novel variants with potentially altered epitopes.

Unbiased discovery applications strongly favor MS approaches - antibody detection inherently requires prior knowledge of the target, while MS can identify unexpected ST5 interactions, novel binding partners, or previously uncharacterized protein complexes through immunoprecipitation followed by MS analysis (IP-MS) . This discovery capability is particularly valuable in signaling pathway research where the complete interactome of ST5 may not be fully characterized. Absolute quantification represents another MS strength - techniques like multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM) with isotope-labeled standards enable precise absolute quantification of ST5 protein with excellent reproducibility and standardization across laboratories.

The decision framework for selecting between these approaches should consider these experimental factors:

Research QuestionPreferred ApproachRationale
Simple verification of ST5 presence in samplesST5 Antibody-HRPFaster, more cost-effective, higher throughput for simple detection
Quantification across large sample setsST5 Antibody-HRPBetter suited for routine high-throughput screening with established protocols
Identification of novel ST5 protein interactionsMass SpectrometryUnbiased discovery of binding partners without requiring antibodies for each potential interactor
Characterization of ST5 post-translational modificationsMass SpectrometryComprehensive mapping of modification sites and types with site-specific quantification
ST5 isoform discriminationMass SpectrometrySuperior detection of sequence variations and proteoforms that may share antibody epitopes
Spatial localization within tissues or cellsST5 Antibody-HRPBetter spatial resolution and compatibility with morphological analysis
Absolute quantification with cross-laboratory standardizationMass SpectrometryMore consistent absolute quantification using isotope-labeled standards
Detection in samples with extremely limited materialST5 Antibody-HRPGenerally better sensitivity for single-protein detection in trace samples

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 2024 Thebiotek. All Rights Reserved.