LEO1 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Composition

LEO1 Antibody, Biotin Conjugated refers to a polyclonal antibody raised against the LEO1 protein (UniProt ID: LEO1_HUMAN) and chemically linked to biotin. Key characteristics include:

PropertyDescription
TargetLEO1 protein (amino acids 159–209 in humans)
Host SpeciesRabbit
ConjugationBiotin via covalent linkage using NHS ester chemistry
ApplicationsWestern blot (WB), ELISA, immunohistochemistry (IHC), immunofluorescence (IF)
SpecificityDetects endogenous LEO1 in human, mouse, and rat samples

Production and Validation

The antibody is generated through:

  1. Immunogen Design: A synthesized peptide corresponding to residues 159–209 of human LEO1 .

  2. Biotinylation: Achieved using kits that activate biotin for efficient conjugation to antibodies. Typical protocols involve:

    • Reaction completion in 10 minutes at room temperature .

    • Biotin-to-antibody ratios optimized for minimal steric interference .

  3. Quality Control: Validated via WB (showing a 105 kDa band) and ELISA (detection limits: 2,000–20,000 dilutions) .

Transcriptional Regulation Studies

LEO1 is a core component of the PAF1 complex, which regulates RNA polymerase II activity and histone modifications . The biotin conjugate facilitates:

  • Chromatin immunoprecipitation (ChIP) to map LEO1’s binding sites .

  • Co-immunoprecipitation (Co-IP) to study interactions with partners like KMT2A/MLL1 .

Disease Research

  • Cancer: LEO1 supports leukemogenesis via KMT2A/MLL1 fusion proteins .

  • Neurodegeneration: Implicated in transcriptional dysregulation in C9orf72-associated FTD .

Technical Advantages

ApplicationProtocol DetailBenefit
ELISASandwich assays with streptavidin-HRP Enhanced sensitivity (pg/mL detection)
Multiplex IHCSimultaneous detection using biotin-streptavidin systems Reduced cross-reactivity
High-ThroughputScalable biotinylation (0.1–1 g IgG per reaction) Cost-effective production

Research Case Studies

  • Leukemia Mechanisms: Demonstrated LEO1’s role in maintaining MLL1-rearranged oncoprotein activity .

  • Stem Cell Pluripotency: Identified LEO1 as critical for embryonic stem cell self-renewal via H3K4me3 modulation .

  • Viral Interactions: Revealed PAF1C-NS1 protein interactions in influenza H3N2 transcription .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
Gm185 antibody; LEO1 antibody; LEO1_HUMAN antibody; LOC123439 antibody; Paf1/RNA polymerase II complex component antibody; Replicative senescence down regulated leo1 like protein antibody; Replicative senescence down-regulated leo1-like protein antibody; Replicative senescence downregulated leo1 like antibody; RNA polymerase associated protein LEO1 antibody; RNA polymerase-associated protein LEO1 antibody; Senescence downregulated leo1 like antibody
Target Names
LEO1
Uniprot No.

Target Background

Function
The LEO1 protein is a component of the PAF1 complex (PAF1C), a multi-functional complex involved in RNA polymerase II-mediated transcription. PAF1C plays a crucial role in regulating development and maintaining embryonic stem cell pluripotency. It interacts with RNA polymerase II, specifically with both phosphorylated (Ser-2 and Ser-5) and non-phosphorylated forms of the C-terminal domain (CTD) of POLR2A. This interaction facilitates transcriptional elongation, often synergistically with TCEA1 and in cooperation with the DSIF complex and HTATSF1. PAF1C is essential for the transcription of Hox and Wnt target genes and plays a significant role in hematopoiesis, stimulating the transcriptional activity of KMT2A/MLL1. Furthermore, its association with KMT2A/MLL1-rearranged oncoproteins (e.g., KMT2A/MLL1-MLLT3/AF9 and KMT2A/MLL1-MLLT1/ENL) contributes to leukemogenesis. PAF1C is involved in histone modifications, including H2B ubiquitination and H3K4me3 (histone H3 lysine 4 trimethylation). It recruits the RNF20/40 E3 ubiquitin-protein ligase complex and the E2 enzyme UBE2A or UBE2B to chromatin, mediating the monoubiquitination of H2BK120 (histone H2B lysine 120). This H2B ubiquitination, facilitated by UBE2A/B, is thought to be coupled to transcription. PAF1C also participates in mRNA 3' end formation, likely through interaction with cleavage and poly(A) factors, and connects PAF1C to Wnt signaling. In influenza A (H3N2) infection, PAF1C interacts with the viral NS1 protein, influencing gene transcription. It is also involved in the polyadenylation of mRNA precursors.
Gene References Into Functions
  1. A positive correlation between PRL-3 and LEO1 levels was observed in AML patient samples. PMID: 24686170
  2. Analysis of glioblastomas and oligodendrogliomas revealed a fusion gene between LEO1 exon 11 and SLC12A1 exon 10, identified through aberrant 5'-3' gene expression in regions of copy number change. PMID: 20196086
Database Links

HGNC: 30401

OMIM: 610507

KEGG: hsa:123169

STRING: 9606.ENSP00000299601

UniGene: Hs.567662

Protein Families
LEO1 family
Subcellular Location
Nucleus.
Tissue Specificity
Highly expressed in skeletal muscle and heart. Weakly expressed in placenta and liver.

Q&A

What is LEO1 protein and why is it significant in molecular research?

LEO1 (RNA polymerase-associated protein LEO1) is a 666 amino acid protein belonging to the LEO1 family. It functions as a critical component of the PAF1 complex (PAF1C), which plays multiple roles during transcription by RNA polymerase II and is implicated in regulating development and maintaining embryonic stem cell pluripotency .

While LEO1's calculated molecular weight is approximately 75 kDa, it typically appears at around 105 kDa in Western blots due to post-translational modifications . The protein is encoded by the LEO1 gene (Gene ID: 123169) , making it an important target for research in gene expression regulation and developmental biology.

What molecular information is available for LEO1 antibodies?

LEO1 antibodies are available in various formats with detailed molecular information:

PropertyDetails
UniProt IDQ8WVC0
GenBank AccessionBC018147
Gene SymbolLEO1
Gene ID (NCBI)123169
Calculated MW666 aa, 75 kDa
Observed MW105 kDa
RRIDAB_10640429 (for catalog #12281-1-AP)

This molecular information provides the foundation for research targeting LEO1 in various experimental systems and validates the specificity of antibodies used in research applications.

How does biotin conjugation enhance antibody functionality in research applications?

Biotin conjugation significantly enhances antibody functionality through multiple mechanisms:

Biotin, a small vitamin molecule, has an extremely high affinity for streptavidin and avidin proteins. In antibody applications, biotin is typically conjugated to primary amines (lysine residues) on the antibody molecule . Typically, 3-8 biotin molecules are incorporated per antibody , creating multiple binding sites for detection reagents.

This conjugation creates a powerful amplification system when used with streptavidin-linked reporter molecules (enzymes, fluorophores, or quantum dots). As noted in recent research, "biotin-labeled antibodies, often paired with streptavidin-HRP or AP, are used by researchers in ELISA, Western blotting, and immunohistochemistry" .

The biotin-streptavidin system provides several advantages:

  • Significantly enhanced sensitivity for detecting low-abundance targets

  • Versatility across multiple detection platforms

  • Compatibility with various amplification strategies

  • Stable binding that withstands stringent washing conditions

This amplification capability makes biotin-conjugated antibodies particularly valuable for detecting proteins like LEO1 that may be expressed at relatively low levels in certain cell types or tissues.

What are the optimal working dilutions for biotin-conjugated LEO1 antibodies in various applications?

Based on the manufacturer's recommendations and experimental validation data, the optimal dilution ranges for biotin-conjugated LEO1 antibodies vary by application:

ApplicationRecommended Dilution RangeNotes
Western Blot (WB)1:500-1:2400 (polyclonal) 1:5000-1:50000 (monoclonal) Sample-dependent, should be titrated for optimal results
Immunohistochemistry (IHC)1:50-1:500 Suggested antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0
Immunofluorescence (IF)/ICC1:10-1:100 Validated in HeLa cells
ELISA1:500-1:1000 Higher dilutions may be used for sensitive detection systems

For optimal results, researchers should perform titration experiments with positive controls (e.g., HeLa cells, Jurkat cells, mouse brain tissue) to determine the ideal working concentration for their specific experimental setup. The dilution producing the highest signal-to-noise ratio while minimizing background should be selected.

As noted in product documentation: "It is recommended that this reagent should be titrated in each testing system to obtain optimal results" .

How can researchers prepare their own biotin-conjugated LEO1 antibody?

Preparing a custom biotin-conjugated LEO1 antibody from an unconjugated antibody involves several critical steps:

Materials required:

  • Purified LEO1 antibody (concentration ≥2 mg/ml recommended)

  • NHS-Biotin or Sulfo-NHS-Biotin reagent

  • Amine-free buffer (PBS pH 7.4)

  • Desalting column (50 kDa cutoff)

  • Storage buffer components

Procedure:

  • Antibody preparation:

    • Remove sodium azide and amine-containing compounds through dialysis

    • Exchange into appropriate reaction buffer (typically PBS)

  • Conjugation reaction:

    • Calculate appropriate biotin:antibody ratio (80 μg biotin per mg antibody is standard, but can use 10-400 μg range for optimization)

    • Dissolve biotin reagent immediately before use (unstable once solubilized)

    • Add dissolved biotin to antibody solution

    • Incubate at room temperature for 30-60 minutes

  • Purification:

    • Remove unreacted biotin using a desalting column (50 kDa cutoff)

    • Collect and pool protein-containing fractions

    • For streptavidin-biotin antibody preparation, wash on 50 kDa cutoff column per manufacturer's protocol

  • Storage:

    • Add glycerol to 50% final concentration

    • Store at -20°C in small aliquots to prevent freeze-thaw damage

For researchers requiring simplified workflows, commercial biotinylation kits like the LYNX Rapid Plus Biotin Antibody Conjugation Kit provide pre-optimized reagents and protocols .

First-time users should prepare a range of conjugates with varying biotin:antibody ratios and test each for optimal performance in their specific application.

What are the critical storage and handling requirements for maintaining biotin-conjugated LEO1 antibody stability?

Proper storage and handling are essential for maintaining the functionality of biotin-conjugated LEO1 antibodies:

Storage conditions:

  • Store at -20°C for long-term preservation

  • Avoid repeated freeze-thaw cycles by preparing small, single-use aliquots

  • Most products remain stable for one year after shipment when stored properly

Buffer composition:

  • Typical storage buffer: PBS or TBS (pH 7.3-7.4) with 50% glycerol

  • Preservatives: 0.02-0.03% sodium azide or 0.03% Proclin 300

  • Some formulations include stabilizers like 0.1-1% BSA

Handling recommendations:

  • Thaw frozen aliquots completely before use

  • Briefly centrifuge vials before opening to collect contents

  • Keep on ice when working with diluted antibody solutions

  • Return to -20°C immediately after use

  • For working solutions, short-term storage (up to one week) at 4°C is acceptable

  • Protect biotinylated antibodies from strong light exposure

Following these guidelines will help maintain antibody performance and extend shelf life, ensuring reproducible results across experiments.

How can researchers validate the specificity of biotin-conjugated LEO1 antibodies?

Rigorous validation is essential for ensuring the specificity of biotin-conjugated LEO1 antibodies. A comprehensive validation approach should include:

1. Positive and negative control samples:

  • Positive controls: Test with samples known to express LEO1, such as HeLa, Jurkat, MCF-7, HEK-293, HepG2, or A549 cells

  • Negative controls: Use LEO1 knockdown/knockout cells or tissues

  • Multiple sample types: Test across human, mouse, and rat samples if antibody is reported to cross-react with these species

2. Western blot verification:

  • Confirm band at the expected molecular weight (~105 kDa, despite calculated MW of 75 kDa)

  • Compare with unconjugated LEO1 antibody to ensure biotinylation hasn't altered specificity

  • Use multiple antibodies targeting different epitopes of LEO1

3. Competitive inhibition:

  • Pre-incubate antibody with immunogen peptide (if available)

  • Observe elimination of specific signal in peptide-blocked samples

4. Cross-platform validation:

  • Verify consistent staining patterns across multiple techniques (WB, IHC, IF)

  • For IHC, confirm appropriate cellular/subcellular localization pattern

  • Compare results with published literature on LEO1 expression and localization

5. Technical controls:

  • Include streptavidin-only controls (no primary antibody)

  • Use isotype control antibodies with biotin conjugation

  • Include endogenous biotin blocking steps to eliminate non-specific binding

The validation process should be systematic and well-documented, with results compared against known biological characteristics of LEO1 as a component of the PAF1 complex involved in transcriptional regulation.

What approaches can resolve high background issues when using biotin-conjugated LEO1 antibodies?

High background is a common challenge when using biotin-conjugated antibodies. Researchers can implement several methodological solutions:

1. Address endogenous biotin interference:

  • Endogenous biotin in tissues can bind directly to streptavidin detection reagents

  • Implement avidin/biotin blocking kit before primary antibody application

  • For severe cases, consider using a biotin-free detection system as an alternative

2. Optimize antibody concentration:

  • Perform careful titration experiments to determine minimal effective concentration

  • Start with higher dilutions (1:500 for IHC, 1:2000 for WB) and adjust as needed

  • Balance signal strength with background reduction

3. Enhance blocking protocols:

  • Extend blocking time to 1-2 hours at room temperature

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

  • Include 0.1-0.3% Triton X-100 in antibody diluent to reduce non-specific binding

4. Modify washing procedures:

  • Increase number and duration of wash steps

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

  • Use TBS instead of PBS for washing if phosphate interferes with detection

5. Optimize antigen retrieval:

  • Test both recommended retrieval methods: TE buffer pH 9.0 and citrate buffer pH 6.0

  • Adjust retrieval duration and temperature

  • Include a cool-down period after retrieval

6. Refine detection system:

  • Dilute streptavidin-conjugated detection reagent further

  • Consider alternative streptavidin conjugates (HRP vs. AP)

  • Use detection systems with lower background characteristics

7. Modify incubation conditions:

  • Incubate primary antibody at 4°C overnight instead of at room temperature

  • Use humidity chambers to prevent section drying

  • Include carrier proteins in antibody diluent

Through systematic optimization of these parameters, researchers can significantly improve signal-to-noise ratio when working with biotin-conjugated LEO1 antibodies.

How can researchers quantitatively assess the degree of biotinylation in LEO1 antibody preparations?

Precise quantification of biotin incorporation is essential for consistent experimental results. Researchers can employ several methodological approaches:

1. Spectrophotometric Analysis:

  • Perform non-destructive UV scan (220-400 nm) of the biotinylated antibody

  • Calculate the biotin-to-protein ratio using established extinction coefficients

  • Optimal conjugation typically shows 3-8 biotin molecules per antibody

2. Competitive Binding Assay:

  • As described in recent literature, use a competitive biotin-streptavidin reaction methodology

  • Employ a reference antibody dually labeled with biotin and electrochemiluminescent (ECL) luminophores

  • The reference and test antibodies compete for streptavidin binding sites on magnetic beads

  • ECL signal strength correlates inversely with biotinylation degree of test antibody

  • This approach provides functional assessment rather than just molecular counting

3. HABA Assay:

  • Based on displacement of 4'-hydroxyazobenzene-2-carboxylic acid (HABA) from avidin

  • Measure absorbance decrease at 500 nm as biotin displaces HABA

  • Calculate molar ratio by comparing to protein concentration

4. Lot-to-Lot Comparison:

  • When evaluating new batches, compare biotinylation levels to reference standards

  • As noted in published research: "The validation experiments and aforementioned proof-of-concept experiments establish the applicability of the method"

  • Document biotinylation degree for each batch to ensure experimental reproducibility

The optimal degree of biotinylation balances detection sensitivity with antibody functionality - too few biotin molecules reduces sensitivity, while excessive biotinylation can alter antibody binding characteristics and lead to aggregation.

What methodological approaches can characterize batch-to-batch variation in biotin-conjugated LEO1 antibodies?

Batch-to-batch variation can significantly impact experimental reproducibility. Researchers should implement rigorous characterization methods:

1. Functional Binding Capability Assessment:

  • Employ the competitive biotin-streptavidin binding methodology described in recent literature

  • This approach evaluates the actual binding capability rather than just molecular counts

  • Analyze competition curves to detect subtle variations between lots

  • As noted in research: "The slightly deviating curve of lot 1 indicates a slightly stronger biotin–streptavidin competition and, thus, a higher degree of biotinylation"

2. Direct Performance Comparison:

  • Run parallel experiments with both old and new lots using identical conditions

  • Include consistent positive controls (e.g., HeLa cells for LEO1 detection)

  • Compare signal intensity, specificity, background, and reproducibility

  • Document performance metrics for future reference

3. Analytical Characterization:

  • Perform spectrophotometric analysis to determine biotin incorporation level

  • Compare protein concentration and biotin:protein ratios between batches

  • Assess physical characteristics like aggregation tendency and stability

4. Application-Specific Testing:

  • Evaluate each new lot in the specific application(s) where it will be used

  • For WB: Compare band intensity and specificity at fixed protein loading

  • For IHC/IF: Compare staining patterns, intensity, and background

  • For ELISA: Generate standard curves with both lots and compare sensitivity/dynamic range

5. Standardization Approach:

  • Maintain reference material from well-characterized lots

  • Use internal standards for normalization across experiments

  • Document lot-specific optimal working dilutions

As emphasized in published research: "Whether or not the three lots are acceptable from the quality control point of view depends on the setup of a standard, which is based on the specific needs of a researcher" . Establishing clear acceptance criteria based on research requirements is essential for maintaining experimental consistency.

What are the considerations for using biotin-conjugated LEO1 antibodies in multiplex immunofluorescence?

Multiplex immunofluorescence with biotin-conjugated LEO1 antibodies requires careful experimental design:

1. Panel Design Considerations:

  • Position biotin-streptavidin detection strategically within multiplex panel

  • Reserve biotin-conjugated LEO1 antibody for targets requiring signal amplification

  • Select streptavidin conjugates with minimal spectral overlap with other fluorophores

  • Consider using LEO1 antibody in the near-infrared channel if available

2. Sequential Staining Strategy:

  • Apply biotinylated LEO1 antibody and streptavidin detection first

  • Block remaining biotin binding sites (with unconjugated streptavidin or free biotin)

  • Proceed with directly labeled antibodies for other targets

  • This approach minimizes cross-reactivity between detection systems

3. Endogenous Biotin Management:

  • Implement avidin/biotin blocking before applying any biotinylated antibodies

  • Particularly critical for biotin-rich tissues (liver, kidney, adipose tissue)

  • Consider tissue-specific optimization of blocking protocols

4. Signal Balancing:

  • Adjust biotin-conjugated LEO1 antibody concentration to match signal intensity with other targets

  • Biotin-streptavidin amplification may create signal imbalance with directly labeled antibodies

  • Optimize exposure settings for each channel during image acquisition

5. Validation Controls:

  • Include single-staining controls for each antibody in the panel

  • Perform fluorescence-minus-one (FMO) controls to assess spectral overlap

  • Test for potential cross-interactions between detection systems

This methodological approach enables successful integration of biotin-conjugated LEO1 antibodies into complex multiplex panels while maintaining specificity and quantitative accuracy.

How do researchers optimize conditions for biotin-conjugated LEO1 antibody in chromatin immunoprecipitation studies?

While standard LEO1 antibodies have been validated for applications including WB, IHC, and IP , optimizing biotin-conjugated versions for chromatin immunoprecipitation (ChIP) requires specific considerations:

1. Pre-Clearing Strategy:

  • Implement rigorous pre-clearing with streptavidin beads before immunoprecipitation

  • This removes materials with non-specific affinity for streptavidin

  • Include appropriate blocking agents (BSA, salmon sperm DNA) to reduce background

2. Antibody Titration:

  • Test multiple antibody concentrations (2-8 μg per reaction)

  • For IP applications, manufacturer data suggests using 2-8 μg/mg lysate

  • Optimize for maximum signal-to-noise ratio with minimal antibody consumption

3. Streptavidin Bead Selection:

  • Use magnetic streptavidin beads for better recovery and handling

  • Pre-block beads to minimize non-specific binding

  • Consider bead capacity when calculating optimal antibody amounts

4. Buffer Optimization:

  • Test different salt concentrations in wash buffers to balance specificity and yield

  • Include detergents at appropriate concentrations to reduce background

  • Consider adding competitors for non-specific interactions

5. Elution Strategy Development:

  • The exceptionally strong biotin-streptavidin interaction presents unique challenges

  • Test competitive elution with excess free biotin

  • Consider harsh elution conditions (e.g., high temperature in SDS) if necessary

  • For DNA analysis, direct PCR from bead-bound complexes may be most efficient

6. Validation Approach:

  • Compare to results using non-biotinylated LEO1 antibodies

  • Perform ChIP-qPCR on known LEO1-associated genomic regions

  • Include appropriate controls (IgG, input, non-specific genomic regions)

This systematic optimization strategy enables researchers to leverage biotin-conjugated LEO1 antibodies for chromatin studies despite the inherent challenges of the biotin-streptavidin system in ChIP applications.

What factors influence the choice between biotin-HRP and biotin-AP detection systems with LEO1 antibodies?

The choice between horseradish peroxidase (HRP) and alkaline phosphatase (AP) detection systems with biotin-conjugated LEO1 antibodies depends on multiple experimental factors:

1. Sensitivity Requirements:

ParameterStreptavidin-HRPStreptavidin-AP
Detection LimitExcellent with chemiluminescent substratesGood, can be enhanced with extended development
Signal Development SpeedRapid (minutes)Slower (10-30 minutes)
Signal StabilityShort-lived with chemiluminescent substratesLonger-lasting, stable signal
Amplification PotentialExcellent with tyramide amplificationGood with BCIP/NBT substrates

2. Application-Specific Considerations:

ApplicationPreferred SystemRationale
Western BlotStreptavidin-HRPFaster development, better with chemiluminescent detection
IHC (Permanent)Streptavidin-APCreates permanent, stable precipitate
ELISAEither (HRP more common)Both provide good sensitivity
Multiplex IFDepends on other fluorophoresChoose based on spectral compatibility

3. Sample-Specific Factors:

  • For tissues with high endogenous peroxidase activity: Consider streptavidin-AP

  • For tissues with high endogenous phosphatase activity: Consider streptavidin-HRP

  • For tissues with high biotin content: Additional blocking steps needed regardless of enzyme choice

4. Practical Laboratory Considerations:

  • Available detection instruments (e.g., chemiluminescent imagers)

  • Substrate availability and cost

  • Experience with specific detection systems

As noted in recent research, "biotin-labeled antibodies, often paired with streptavidin-HRP or AP, are used by researchers in ELISA, Western blotting, and immunohistochemistry" . The optimal choice should be determined based on specific experimental requirements, sample characteristics, and available detection systems.

What innovations are emerging in the application of biotin-conjugated antibodies for single-cell analysis?

Recent technological advances have expanded the utility of biotin-conjugated antibodies in single-cell analysis techniques:

1. Antibody-Oligonucleotide Conjugates for CITE-seq:

  • Biotin-streptavidin linkage enables creation of antibody-oligonucleotide conjugates for single-cell applications

  • As described in recent protocols: "For experiments involving conjugation via a streptavidin-biotin-linkage, we therefore recommend simply ordering biotinylated oligos"

  • This approach enables simultaneous protein and RNA profiling at single-cell resolution

  • Biotinylated antibodies serve as the foundation for creating these advanced reagents

2. Protocol Optimization for Single-Cell Applications:

  • Specialized conjugation protocols have been developed for single-cell applications

  • Methodology includes: "Merge streptavidin-antibodies with biotinylated-oligos in PBS/0.5M NaCl"

  • Each antibody can be labeled with "~2 streptavidin molecules" which can bind "800pmol (biotin binding sites)"

  • This approach enables multiplexed protein detection in single-cell RNA sequencing experiments

3. Panel Design for Single-Cell Protein Profiling:

  • For single-cell applications: "Make antibody panel by pooling all streptavidin-biotin antibodies"

  • Use "1-2 μg of each antibody-oligo complex, comparable to what is recommended for flow cytometry per test"

  • Special consideration for biotin blocking: "Add 2 μl of 10 mM biotin to block unoccupied biotin-binding sites"

4. Technical Challenges and Solutions:

  • Biotin-streptavidin antibody-oligo conjugates require special storage: "have to be stored separately and should only be pooled shortly before the run"

  • Purification is essential: "clean pooled panel on 50kDa cutoff column per manufacturer's protocol to remove unbound oligos shortly before CITE-seq run"

These innovations represent the cutting edge of biotin-conjugated antibody applications, enabling integration of protein detection (including LEO1) with genomic and transcriptomic analysis at single-cell resolution.

What are the key considerations for selecting the optimal biotin-conjugated LEO1 antibody for research applications?

When selecting a biotin-conjugated LEO1 antibody for research, researchers should consider:

  • Application compatibility: Verify validation data for your specific application (WB, IHC, IF, ELISA)

  • Species reactivity: Confirm reactivity with your experimental system (human, mouse, rat)

  • Epitope information: Consider antibody epitope location within LEO1 protein:

    • Some target N-terminal regions (e.g., amino acids 2-24)

    • Others target mid-protein regions (e.g., residues 550-600)

  • Validation data: Review available validation data showing specificity in relevant cell lines:

    • For human: HeLa, Jurkat, MCF-7, HEK-293 cells

    • For mouse: NIH 3T3, TCMK-1 cells, mouse tissues

  • Biotin conjugation quality: Consider degree of biotinylation (optimal range: 3-8 biotin molecules per antibody)

  • Format and storage: Evaluate buffer composition, concentration, and stability:

    • Most are supplied in PBS/glycerol with preservatives

    • Storage at -20°C is typically recommended

  • Technical support: Consider manufacturer's technical resources and application support

Through careful evaluation of these parameters, researchers can select the biotin-conjugated LEO1 antibody most likely to yield successful results in their specific experimental system.

How can researchers ensure reproducibility when working with biotin-conjugated LEO1 antibodies?

Ensuring reproducibility with biotin-conjugated LEO1 antibodies requires systematic approaches:

  • Detailed record-keeping:

    • Document complete antibody information (catalog number, lot number, source)

    • Record exact experimental conditions for each application

    • Maintain detailed protocols including all buffer compositions

  • Standardized validation:

    • Establish consistent validation protocols for each new lot

    • Use the same positive controls across experiments (e.g., HeLa cells for human LEO1)

    • Document validation results systematically

  • Quantitative batch comparison:

    • Implement functional comparison between lots using competitive binding assays

    • As noted in research: "It is worth noting that the ECL detection is a highly sensitive technique... but the high sensitivity or very low limit of detection methodology is not a prerequisite for this type of application"

    • Document optimal working dilutions for each lot and application

  • Optimized handling and storage:

    • Store consistently at -20°C with minimal freeze-thaw cycles

    • Prepare single-use aliquots to maintain antibody integrity

    • Follow consistent handling protocols across experiments

  • Comprehensive controls:

    • Include positive and negative controls in each experiment

    • Use isotype controls for background assessment

    • Implement biotin blocking controls to account for endogenous biotin

  • Application-specific optimization:

    • Determine optimal working concentration for each application

    • Document optimized protocols for specific sample types

    • Consider sample-specific factors (fixation, processing 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.