MRAS Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to MRAS and Biotin-Conjugated Antibodies

MRAS (Muscle RAS Oncogene Homolog) is a member of the Ras superfamily of small GTPases, functioning as a molecular switch in signaling pathways regulating cell proliferation, survival, and receptor tyrosine kinase (RTK) signaling . The biotin-conjugated MRAS antibody is a specialized reagent designed for high-sensitivity detection of MRAS in assays such as ELISA, Western blotting (WB), and immunofluorescence (IF). Biotin conjugation leverages the exceptionally high-affinity interaction between biotin and streptavidin (K<sub>D</sub> ≈ 10<sup>−14</sup>–10<sup>−15</sup> M), enabling robust signal amplification in immunoassays .

Biotin Conjugation Technology

Biotin is covalently attached to the MRAS antibody, typically via lysine residues or N-terminal amines, depending on the conjugation method. Key features include:

  • Site-specific conjugation: Some protocols employ reductive alkylation or engineered tags to label the N-terminus, minimizing interference with antigen-binding sites .

  • Random conjugation: Traditional methods target lysine residues, which may alter epitope recognition in antibodies with limited lysine availability .

  • Stability: Biotin-streptavidin interactions resist proteolysis, temperature extremes, and organic solvents, enhancing assay reliability .

3.1. Enzyme-Linked Immunosorbent Assay (ELISA)

In sandwich ELISA, biotin-conjugated MRAS antibodies bind target antigens, followed by streptavidin-linked enzymes (e.g., HRP) for signal detection. For example:

  • Protocol: Capture antibodies immobilize MRAS, biotin-labeled detection antibodies bind, and streptavidin-HRP amplifies the signal .

  • Sensitivity: Enables detection of low-concentration MRAS in complex matrices like serum .

3.2. Western Blotting (WB)

Biotin-conjugated MRAS antibodies are used to probe polyacrylamide gels:

  • Dilution Range: 1:300–1:5000 .

  • Signal Enhancement: Streptavidin-HRP conjugates enhance chemiluminescence for precise band visualization .

3.3. Immunofluorescence (IF)

Used to localize MRAS in cellular compartments:

  • Applications: Cytoplasmic and membrane staining in cancer cells or signaling studies .

5.1. Sensitivity and Specificity

  • ELISA: Biotin-conjugated antibodies enable titration of MRAS in solution, with optimal performance in sandwich assays .

  • WB: Efficient detection in mouse and rat models, with minimal background noise .

5.2. Comparative Analysis

  • Site-Specific Conjugation: Reduces epitope interference, critical for antibodies with limited lysine residues (e.g., variable heavy domains) .

  • Biotin Interference: High endogenous biotin in samples may suppress signal; streptavidin blocking protocols mitigate this .

Challenges and Considerations

  • Cross-Reactivity: Polyclonal antibodies may bind non-specific epitopes; monoclonal variants (e.g., bsm-62693r-biotin) offer higher specificity .

  • Conjugation Efficiency: Random labeling risks compromising antibody activity; site-specific methods (e.g., N-terminal biotinylation) improve yield .

  • Stability: Long-term storage at -20°C is required to preserve activity .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product dispatch occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
Ras-related protein M-Ras (Ras-related protein R-Ras3), MRAS, RRAS3
Target Names
Uniprot No.

Target Background

Function

This antibody targets MRAS, a critical signal transducer involved in regulating cell proliferation via a novel upstream signaling pathway. MRAS activates the MAP kinase pathway.

Gene References Into Functions

MRAS's functional role is supported by the following research:

  • Association with acute coronary syndrome in Czech populations (PMID: 29264877).
  • Identification as a recurrently mutated gene in Type IV gastric neoplasm, suggesting a role in tumorigenesis (PMID: 27891760).
  • Population-specific association between the MRAS rs6782181 polymorphism and serum lipid levels (PMID: 25973078).
  • Minor effect on coronary artery disease susceptibility in Chinese populations (PMID: 25800439).
  • Key role, alongside SHOC2, in polarized cell migration (PMID: 24211266).
  • Involvement in neuronal differentiation via coupling of trophic factors to the MAPK cascade through B-Raf activation (PMID: 12138204).
  • Crucial role in adhesion signaling through cross-talk with Rap1 mediated by RA-GEF-2 (PMID: 17538012).
  • Identification of a CAD risk locus on 3q22.3 within the MRAS gene (PMID: 19198612).

Further details available upon request.

Database Links

HGNC: 7227

OMIM: 608435

KEGG: hsa:22808

STRING: 9606.ENSP00000289104

UniGene: Hs.527021

Protein Families
Small GTPase superfamily, Ras family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side.
Tissue Specificity
Expression highly restricted to the brain and heart.

Q&A

What is MRAS and what cellular functions does it regulate?

MRAS (Muscle RAS oncogene homolog) is a small GTPase protein belonging to the RAS superfamily. It serves as an important signal transducer for upstream stimuli in controlling cell proliferation and activates the MAP kinase pathway . Located in both the cytoplasm and cell membrane, MRAS (also known as R-ras3 or RRAS3) participates in signal transduction cascades that regulate cellular growth, differentiation, and survival . Unlike some other RAS family members, MRAS has been characterized as having a relatively weaker activation effect on the MAP kinase pathway, suggesting potential regulatory differences in signaling mechanisms . In research contexts, studying MRAS provides insights into fundamental cellular signaling networks and their dysregulation in various pathological conditions.

What are the key specifications of commercially available MRAS antibodies?

Commercially available MRAS antibodies come in different forms with specific characteristics that researchers should consider when selecting reagents for their experiments:

SpecificationMonoclonal MRAS AntibodyPolyclonal MRAS Antibody
Catalog #bsm-62693r-biotinbs-22921R-Biotin
HostRabbitRabbit
ReactivityHuman, Mouse, RatMouse, Rat
SourceKLH conjugated synthetic peptide (human MRAS)KLH conjugated synthetic peptide (mouse MRAS)
Immunogen RangeNot specified201-259/259
ConcentrationLot dependent1ug/ul
ApplicationsWBWB
StorageStore at -20°C for 12 monthsStore at -20°C for 12 months
Storage BufferAqueous buffered solution with 0.01M TBS (pH 7.4), 1% BSA, 0.02% Proclin300, 50% GlycerolAqueous buffered solution with 0.01M TBS (pH 7.4), 1% BSA, 0.03% Proclin300, 50% Glycerol

These specifications help researchers select the appropriate antibody based on target species, experimental applications, and storage conditions .

How does biotin conjugation enhance antibody utility in experimental protocols?

Biotin conjugation of antibodies provides significant experimental advantages through the exploitation of the strong biotin-avidin/streptavidin interaction, which has an extremely low dissociation constant (kd) of approximately 10^-15 M . This modification enables multiple methodological improvements:

First, biotinylated antibodies serve as primary detection reagents that can be subsequently targeted with streptavidin or avidin conjugated to various reporter molecules (enzymes, fluorophores, or radiolabels), allowing for signal amplification without compromising antibody binding specificity . Second, the small size of biotin (244 Da) means that multiple biotin molecules can be conjugated to each antibody without measurably affecting immunoreactivity . Third, the biotin-streptavidin system provides exceptional flexibility for experimental design, as biotinylated antibodies can be used in multi-step targeting approaches where the initial antibody-biotin complex serves as a scaffold for subsequent targeting with avidin/streptavidin-conjugated molecules .

Additionally, biotin-conjugated antibodies facilitate more efficient purification and enrichment protocols, particularly for complex biological samples where specific targets must be isolated from heterogeneous mixtures .

What is the recommended working dilution range for Western blot applications?

For Western blot applications, the recommended working dilution range for biotin-conjugated MRAS antibodies is 1:300-1:5000 . This relatively wide range allows researchers to optimize antibody concentration based on specific experimental parameters including protein abundance, detection system sensitivity, and signal-to-noise ratio requirements.

It is strongly recommended to perform initial titration experiments to determine the optimal working dilution for each specific application and sample type. Start with a mid-range dilution (approximately 1:1000) and adjust based on signal intensity and background levels. When establishing optimal dilutions, consider variables such as protein loading amount, transfer efficiency, blocking conditions, and the sensitivity of the detection system being employed.

How do monoclonal and polyclonal MRAS antibodies compare in experimental applications?

Monoclonal and polyclonal MRAS antibodies present different advantages and limitations that should guide their selection for specific research applications:

Monoclonal antibodies represent a single population of antibodies that bind to a specific epitope, offering superior consistency between experiments and batches . The monoclonal anti-MRAS antibody (bsm-62693r-biotin) is derived from a KLH-conjugated synthetic peptide of human MRAS, providing high specificity for the target epitope . This specificity is particularly valuable in applications requiring precise epitope recognition or when distinguishing between closely related proteins.

A significant advantage of monoclonal antibodies is their status as a renewable resource with uniform performance characteristics, whereas polyclonal antibodies represent a limited resource with potential variation between production lots . Research published in 2024 demonstrated that well-characterized monoclonal antibodies can significantly outperform polyclonal counterparts in identification efficiency, with one study showing a monoclonal anti-biotin antibody discovering 3425 ± 6.6% biotin peptides compared to 1425 ± 9.0% for a polyclonal alternative .

What strategies can improve elution efficiency when using anti-biotin antibodies for immunoaffinity enrichment?

Recent research has identified critical improvements to elution strategies for immunoaffinity enrichment of biotinylated peptides. A significant advancement involves the addition of highly organic solvents during the elution phase, which substantially increases recovery of biotinylated molecules .

An optimized protocol published in 2024 demonstrates the following methodological improvements:

  • After sample incubation with anti-biotin antibody beads (typically 1 hour at 4°C), wash beads thoroughly with 1X PBS (four washes recommended) to remove non-specifically bound molecules.

  • Elute bound biotinylated peptides using a dual elution approach with 50 μL of 0.15% TFA (trifluoroacetic acid) applied twice, combining the eluents for subsequent analysis.

  • Further purify the eluent using C18 stage tips, washing with 100 μL of 0.1% FA (formic acid) twice before final elution with 50 μL of 50% MeCN (acetonitrile) containing 0.1% FA .

This improved elution strategy takes advantage of the relatively lower binding affinity of anti-biotin antibodies to biotin (compared to streptavidin/avidin), allowing for more efficient recovery while maintaining specificity . Research indicates that anti-biotin antibody-based methods provide superior results compared to neutravidin-based approaches for site-specific biotin-related research .

How can signal amplification be achieved when using biotin-conjugated antibodies in detection systems?

Signal amplification with biotin-conjugated antibodies can be strategically implemented through several methodologies that exploit the unique properties of the biotin-avidin/streptavidin interaction:

The primary amplification approach utilizes the tetravalent nature of avidin/streptavidin, which can bind up to four biotin molecules per protein . This characteristic enables the construction of detection complexes where multiple reporter molecules interact with each primary biotinylated antibody. Implementation typically involves a multi-step procedure:

  • Apply the biotin-conjugated primary antibody to the target tissue or sample.

  • Follow with avidin/streptavidin conjugated to detection molecules (enzymes, fluorophores, radionuclides).

  • For further amplification, utilize biotinylated secondary detection molecules that can bind additional avidin/streptavidin complexes .

Research indicates that pre-targeting approaches can significantly enhance target-to-background ratios. For example, administering biotinylated antibodies several days prior to introducing radiolabeled avidin/streptavidin has demonstrated improved targeting in experimental systems . The targeted signal can be calculated using the formula:

Targeted signal = (%ID/g) × (moles of antibody injected)

Where %ID/g represents the percentage of injected dose per gram of tissue .

For optimal amplification, researchers should determine the maximum number of biotin molecules that can be conjugated per antibody without compromising immunoreactivity, with studies showing that several biotin molecules can typically be added without measurable losses in binding capacity .

What factors should be considered when designing proximity labeling experiments using biotin-conjugated antibodies?

Proximity labeling experiments with biotin-conjugated antibodies require careful consideration of multiple experimental parameters to ensure reliable results:

First, the specificity of the biotinylated antibody for its target must be rigorously validated to avoid false positive identifications. This is particularly important when studying protein interaction sites, such as those demonstrated for the β2 adrenergic receptor (β2AR) using proximity labeling approaches in living cells .

Second, researchers must optimize the enrichment and recovery of biotinylated peptides or proteins. Recent methodological improvements include the development of monoclonal anti-biotin antibodies with superior reproducibility compared to polyclonal alternatives, and enhanced elution strategies incorporating organic solvents to maximize peptide recovery for LC-MS/MS analysis .

Third, experimental controls must account for potential non-specific biotinylation. This includes performing parallel experiments with non-specific antibodies of matching isotype or using systems where the labeling enzyme is absent or inactive .

Fourth, the temporal dynamics of labeling should be considered, with optimization of labeling time to balance specific signal against increasing background. This is especially relevant for dynamic cellular processes where timing of labeling can significantly impact results .

Finally, downstream analytical methods must be sensitive enough to detect biotinylated targets, particularly when studying low-abundance proteins or transient interactions. Advanced mass spectrometry techniques have been developed specifically for site-specific biotin-related research, with methodological improvements continuing to enhance detection sensitivity and specificity .

How does the binding kinetics of biotin-conjugated antibodies compare with other targeting approaches?

The binding kinetics of biotin-conjugated antibodies represents a critical parameter that distinguishes this approach from alternative targeting strategies:

When biotin-conjugated antibodies are used in conjunction with avidin/streptavidin systems, the resulting complex demonstrates exceptional stability due to the remarkably low dissociation constant (kd) of approximately 10^-15 M . This affinity is substantially stronger than typical antibody-antigen interactions (kd ~10^-9 M), providing exceptional stability for detection systems .

Research utilizing Surface Plasmon Resonance (SPR) with instruments such as the Biacore 8K has enabled precise characterization of binding kinetics in single-cycle kinetic formats, typically using concentrations around 30 nM for biotin-conjugated peptides . These studies demonstrate that while the native antibody-antigen interaction determines initial binding specificity, the subsequent biotin-avidin/streptavidin interaction dramatically enhances retention time and complex stability.

The strong binding affinity of the biotin-avidin/streptavidin complex presents both advantages and challenges. The exceptional stability ensures robust signal retention in detection systems, but can complicate elution in purification applications . This has led to the development of specialized elution strategies for immunoaffinity enrichment of biotinylated molecules, as traditional methods used for conventional antibody-antigen complexes may prove insufficient .

For quantitative analyses, researchers must consider that binding kinetics can be influenced by:

  • The density of biotin conjugation on the antibody

  • Potential steric hindrance affecting biotin accessibility

  • The specific configuration of the detection system (direct vs. bridged detection schemes)

  • Temperature and buffer conditions that may affect binding rates

What are common challenges when using biotin-conjugated MRAS antibodies and how can they be addressed?

Several challenges may arise when working with biotin-conjugated MRAS antibodies, each requiring specific optimization strategies:

High Background Signal: This common issue can result from endogenous biotin in biological samples or non-specific binding. To address this, implement more stringent blocking protocols using biotin-free blocking reagents (avoid avidin/biotin blocking kits for biotin-conjugated primary antibodies). Additionally, increase washing stringency using buffers containing 0.1-0.3% Tween-20, and optimize antibody dilution to reduce non-specific binding .

Poor Signal Intensity: If signal strength is suboptimal, first verify protein expression levels of MRAS in your samples. For enhanced detection, consider signal amplification using streptavidin-conjugated reporter systems, or adjust working dilution within the recommended range (1:300-1:5000) . Loading higher protein amounts may help when detecting low-abundance targets.

Inconsistent Results: Variability between experiments may reflect lot-to-lot differences, particularly with polyclonal antibodies. To address this, standardize experimental conditions, including incubation times and temperatures. Consider switching to monoclonal antibodies, which offer greater consistency between experiments, particularly for longitudinal studies .

Cross-Reactivity Issues: MRAS antibodies may show cross-reactivity with related RAS family proteins. Validate specificity using positive and negative controls, including overexpression systems or knockout/knockdown models. If cross-reactivity persists, explore alternative antibody clones or more specific detection methods .

Inefficient Elution in Immunoprecipitation: When using biotin-conjugated antibodies for pulldown experiments, elution efficiency may be compromised. Implement the improved elution strategy using 0.15% TFA followed by purification with C18 stage tips as described in recent literature .

How should experimental controls be designed for studies utilizing biotin-conjugated MRAS antibodies?

Robust experimental design requires appropriate controls to validate findings and eliminate potential artifacts when using biotin-conjugated MRAS antibodies:

Positive Controls: Include samples with known MRAS expression, such as cell lines with documented MRAS activity or recombinant MRAS protein standards. This verifies antibody functionality and provides a reference signal intensity .

Negative Controls: Incorporate samples lacking MRAS expression (knockout models or cell lines with minimal MRAS expression). Additionally, use isotype control antibodies (biotin-conjugated IgG from the same species but without specific targeting) to assess non-specific binding .

Loading Controls: For Western blot applications, always include appropriate loading controls (e.g., β-actin, GAPDH) to normalize MRAS signals across samples and ensure consistent protein loading .

Blocking Controls: To verify signal specificity, perform competitive inhibition experiments where the biotin-conjugated antibody is pre-incubated with excess unconjugated MRAS antibody or with the immunizing peptide before application to samples .

Specificity Validation: For critical applications, conduct antibody validation using multiple methods. Compare results from different antibody clones targeting distinct MRAS epitopes, or validate using complementary techniques like qPCR to correlate protein and mRNA levels .

Biotin Controls: In tissues with high endogenous biotin (e.g., liver, kidney), perform additional controls to distinguish specific antibody binding from endogenous biotin interactions with detection reagents. Pre-block sections with streptavidin before antibody incubation to mask endogenous biotin .

Elution Controls: For immunoprecipitation experiments, include mock elution controls to verify that detected signals arise from specific antibody-mediated capture rather than non-specific binding to beads or other components .

What are the optimal storage and handling recommendations to maintain antibody performance?

Proper storage and handling of biotin-conjugated MRAS antibodies is essential for maintaining immunoreactivity and ensuring experimental reproducibility:

Storage Temperature: Store antibodies at -20°C for long-term stability. Both monoclonal and polyclonal MRAS antibodies have a documented shelf life of 12 months when stored properly at this temperature .

Storage Buffer Composition: The optimal buffer for MRAS antibodies contains 0.01M TBS (pH 7.4) with 1% BSA, 0.02-0.03% Proclin300 as preservative, and 50% glycerol to prevent freeze-thaw damage . This formulation maintains antibody stability while preventing microbial contamination.

Aliquoting Strategy: Upon receipt, divide antibodies into small single-use aliquots to minimize freeze-thaw cycles. Each freeze-thaw event can reduce antibody activity by 5-10%, so limiting these cycles is crucial for maintaining performance .

Working Dilution Preparation: When preparing working dilutions, use fresh, sterile buffers. For Western blot applications, dilute antibodies in blocking buffer containing 0.05-0.1% Tween-20 to reduce non-specific binding. Prepare working dilutions immediately before use rather than storing diluted antibody .

Temperature Transitions: Allow antibodies to warm gradually to room temperature before opening vials to prevent condensation, which can introduce contaminants and accelerate degradation. After use, return antibodies promptly to -20°C storage .

Contamination Prevention: Use sterile technique when handling antibodies to prevent microbial contamination. Avoid exposure to strong light sources, particularly for fluorescently labeled secondary detection reagents used in conjunction with biotin-conjugated antibodies .

Transport Considerations: If antibodies must be transported between laboratories, use dry ice to maintain freezing temperatures. Brief exposure to room temperature during shipping or handling should not significantly impact antibody performance if properly managed .

How can biotin-conjugated MRAS antibodies be utilized in multiplexed detection systems?

Biotin-conjugated MRAS antibodies offer significant advantages in multiplexed detection systems through their compatibility with diverse reporting methodologies:

In fluorescence-based multiplexing, biotin-conjugated MRAS antibodies can be paired with differentially labeled streptavidin conjugates (e.g., streptavidin-Alexa Fluor dyes) alongside antibodies with direct fluorophore conjugation targeting other proteins of interest . This approach allows visualization of MRAS in relation to multiple other cellular components simultaneously, providing spatial context for protein interactions and cellular localization.

For mass spectrometry applications, biotin-conjugated antibodies enable selective enrichment of MRAS and its interaction partners from complex biological samples. Recent methodological improvements in elution strategies significantly enhance the recovery of biotinylated peptides, with monoclonal antibodies demonstrating superior performance in identifying biotin-modified sites compared to polyclonal alternatives (3425 ± 6.6% vs 1425 ± 9.0%) .

Particularly powerful applications combine biotin-conjugated MRAS antibodies with proximity labeling techniques to identify protein-protein interactions. In this approach, MRAS antibodies can be used to validate interactions identified through techniques like BioID or APEX, where proteins in close proximity to MRAS would be biotinylated and subsequently detected .

For chromogenic multiplexing in tissue sections, researchers can employ sequential detection protocols where biotin-conjugated MRAS antibodies are visualized first using one chromogen, followed by additional antibodies detected with different visualization systems. Between detection rounds, complete stripping or blocking of previous detection components is essential to prevent cross-reactivity .

What considerations are important when using biotin-conjugated MRAS antibodies in proximity ligation assays?

Proximity ligation assays (PLAs) using biotin-conjugated MRAS antibodies require careful optimization to generate reliable data about protein-protein interactions:

First, when designing PLA experiments, researchers must consider the epitope location on MRAS to ensure that antibody binding does not interfere with potential protein interaction sites. This is particularly important for small proteins like MRAS, where antibody binding may create steric hindrance affecting natural protein-protein interactions .

Second, the biotin conjugation density must be optimized to provide sufficient biotin molecules for detection while avoiding over-conjugation that could impair antibody binding specificity. Typical conjugation protocols aim for 3-8 biotin molecules per antibody to maintain immunoreactivity while providing adequate detection sensitivity .

Third, appropriate controls are essential to validate PLA results, including:

  • Negative controls omitting one primary antibody

  • Controls using antibodies against proteins known not to interact with MRAS

  • Positive controls targeting known MRAS interaction partners

  • Validation using alternative methods (co-immunoprecipitation, FRET)

Fourth, careful optimization of PLA reaction conditions is necessary, including:

  • Antibody concentrations (typically starting with the middle of the recommended range, 1:1000)

  • Incubation times and temperatures

  • Washing stringency to minimize background

  • Rolling circle amplification duration for optimal signal-to-noise ratio

Finally, quantitative analysis of PLA signals should account for the cellular context, as MRAS localization in both cytoplasm and cell membrane compartments may affect interaction patterns . Signal quantification should consider not only the number of PLA puncta but also their distribution relative to cellular structures and other proteins of interest.

How do biotin-conjugated monoclonal and polyclonal antibodies compare in immunohistochemistry applications?

In immunohistochemistry (IHC) applications, biotin-conjugated monoclonal and polyclonal MRAS antibodies present distinct performance characteristics that impact experimental outcomes:

Monoclonal antibodies offer superior specificity by targeting a single epitope, which minimizes cross-reactivity with related proteins and provides consistent staining patterns across different tissue samples . This specificity is particularly valuable when studying MRAS in tissues where other RAS family members are expressed, as it helps distinguish specific MRAS signals from potential cross-reactive background .

Polyclonal antibodies recognize multiple epitopes on the MRAS protein, potentially providing enhanced sensitivity in detecting low-abundance targets and greater tolerance for epitope modifications that might affect monoclonal antibody binding . This multi-epitope recognition can be advantageous when examining fixed tissues where protein conformation or epitope accessibility may be altered by fixation procedures.

A significant consideration in IHC applications is endogenous biotin, which is abundant in certain tissues (particularly liver, kidney, and some brain regions). When using biotin-conjugated antibodies in these tissues, researchers must implement effective blocking strategies to minimize background from endogenous biotin interaction with detection reagents . Alternative detection methods using polymer-based systems rather than avidin-biotin complexes may be preferable for tissues with high endogenous biotin content.

Research comparing monoclonal and polyclonal antibody performance has demonstrated that well-characterized monoclonal antibodies generally provide more consistent results with greater reproducibility between experiments and between different researchers . This advantage is particularly relevant for longitudinal studies or multi-center research projects where standardization is critical.

What are the emerging applications for biotin-conjugated antibodies in single-cell analysis?

Biotin-conjugated antibodies, including those targeting MRAS, are enabling several innovative approaches in single-cell analysis:

In mass cytometry (CyTOF) applications, biotin-conjugated primary antibodies serve as detection reagents that can be visualized using metal-tagged streptavidin conjugates. This approach enables integration of MRAS detection into comprehensive cellular phenotyping panels with minimal channel overlap, allowing simultaneous examination of MRAS expression alongside numerous other markers at single-cell resolution .

Single-cell proteomics techniques now incorporate biotin-conjugated antibodies for targeted protein enrichment prior to analysis. The improved elution strategies developed for biotin-antibody systems enhance recovery of low-abundance proteins like MRAS from individual cells, facilitating more comprehensive protein profiling at the single-cell level . This approach is particularly valuable for heterogeneous populations where bulk analysis would obscure important cellular subpopulations.

Proximity labeling approaches using biotin-conjugated antibodies enable spatial proteomic mapping in single cells, revealing protein interaction networks with subcellular resolution. These techniques address the significant challenge of characterizing protein interactions within intact cellular environments, providing insights into functional protein complexes and their dynamics .

For spatial transcriptomics, biotin-conjugated antibodies enable simultaneous visualization of MRAS protein expression alongside mRNA detection. This multimodal approach correlates post-transcriptional regulation with protein expression patterns, revealing potential regulatory mechanisms affecting MRAS functionality at the single-cell level .

Advanced multiplexed imaging techniques using cyclic immunofluorescence now incorporate biotin-conjugated antibodies to expand the number of detectable targets. For MRAS studies, this enables visualization of expression patterns in relation to numerous other cellular components, providing contextual information about signaling pathway activation and cellular phenotypes .

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.