MRAS Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Applications and Performance

Validated applications include:

ApplicationRecommended DilutionKey Findings
Immunohistochemistry (IHC)1:20–1:200 Localizes MRAS in formalin-fixed paraffin-embedded tissues .
Western Blot (WB)1:1,000 Detects MRAS in human brain, HeLa, and mouse/rat lysates .
Immunoprecipitation (IP)1:20 Efficiently enriches MRAS from human heart lysates .
Immunofluorescence (IF)1:20–1:200 Visualizes MRAS in cellular compartments, validated in knockout controls .

Validation and Specificity

  • Knockout Validation: Specificity confirmed using MRAS-knockout HeLa cell lysates (ab257541), showing no cross-reactivity .

  • Cross-Reactivity: No observed binding to non-target MAGUK family proteins (e.g., SAP97, PSD-93) .

  • Post-Translational Modifications (PTMs): Detects native MRAS without interference from common PTMs (e.g., phosphorylation) .

Research Findings

  • Therapeutic Potential: MRAS antibodies enable studies on oncogenic mutations (e.g., Q71R) linked to Noonan syndrome and cancers .

  • Analytical Techniques:

    • Chromatography: Reversed-phase LC-MS identifies charge variants and degradation products .

    • Electrophoresis: SDS-PAGE confirms purity (>95%) and molecular weight .

StudyMethodOutcome
SMP Complex AnalysisCo-IP + WB MRAS-GTP recruits SHOC2/PP1c to dephosphorylate RAF kinases .
Subclass SwitchingPlasmid engineering IgG subclass variants (IgG1/IgG2a) enable multiplex labeling .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The MRAS recombinant monoclonal antibody was developed by integrating the MRAS antibody genes into plasmid vectors. These engineered plasmid vectors were then introduced into suitable host cells using exogenous protein expression techniques, facilitating the production of the antibody. Following production, the MRAS recombinant monoclonal antibody underwent purification through affinity chromatography. Comprehensive validation confirmed its suitability for three applications: ELISA, IHC, and IF.

The MRAS protein, a member of the Ras superfamily, functions as a molecular switch in cellular signaling pathways. Its primary functions include regulating cell growth, differentiation, survival, and receptor tyrosine kinase (RTK) signaling transduction.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the shipping method or location. Please contact your local distributors for specific delivery details.
Synonyms
FLJ42964 antibody; M ras antibody; M-ras antibody; Mras antibody; Muscle and microspikes Ras antibody; Muscle RAS oncogene homolog antibody; Muscle Ras oncogene homologue antibody; Muscle Ras viral oncogene homolog antibody; R ras3 antibody; R-ras3 antibody; Ras related protein MRas antibody; Ras related protein RRas3 antibody; Ras-related protein M-Ras antibody; Ras-related protein R-Ras3 antibody; RASM_HUMAN antibody; Related Ras viral oncogene homolog 3 antibody; RRas3 antibody; XRas antibody
Target Names
Uniprot No.

Target Background

Function
MRAS serves as a crucial signal transducer for novel upstream stimuli in regulating cell proliferation. It activates the MAP kinase pathway.
Gene References Into Functions
  1. In a study of Czechs, there was no significant association between the MRAS minor T allele (recessive model) and acute coronary syndrome (30.4% vs 29.4% carriers, p = 0.54; OR 1.05; 95% CI 0.89-1.24 for males and 32.1% vs 29.7% carriers, p = 0.28; OR 1.12; 95% CI 0.91-1.37 for females). PMID: 29264877
  2. Research has revealed that MRAS mutations occur recurrently, suggesting their potential role in driving tumorigenesis of Type IV gastric neoplasm. PMID: 27891760
  3. The association between the MARS rs6782181 polymorphism and serum lipid levels varies between the Mulao and Han populations, as well as between males and females within each ethnic group. PMID: 25973078
  4. The MRAS gene loci may have a minor influence on susceptibility to coronary artery disease in the Chinese population. PMID: 25800439
  5. Both MRAS and SHOC2 play a critical role in polarized migration. PMID: 24211266
  6. MRAS is implicated in a novel pathway of neuronal differentiation by coupling specific trophic factors to the MAPK cascade through B-Raf activation. PMID: 12138204
  7. Research has demonstrated a crucial role for the cross-talk between two Ras-family GTPases, M-Ras and Rap1, mediated by RA-GEF-2, in adhesion signaling. PMID: 17538012
  8. A new CAD risk locus on 3q22.3 in MRAS has been identified, along with suggestive association with a locus on 12q24.31 near HNF1A-C12orf43. PMID: 19198612

Show More

Hide All

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 are the advantages of using recombinant monoclonal antibodies for MRAS research?

Recombinant monoclonal antibodies provide several significant advantages over traditional antibodies generated in animals. These advantages include improved standardization leading to greater experimental reproducibility, cost-effectiveness for long-term research projects, and ethical benefits by reducing animal use in antibody production . For MRAS research specifically, recombinant antibodies offer customizable properties such as species specificity and the ability to generate smaller antibody fragments with potentially better tissue penetration. Recombinant antibody technology also allows researchers to produce consistently high yields, with studies reporting an average of 1.1 mg of purified antibody from typical expression systems .

Additionally, recombinant antibodies often demonstrate improved sensitivity and reduced background staining compared to traditional antibodies, as evidenced in comparative immunofluorescence studies where recombinant versions have shown moderately higher sensitivity and significantly lower non-specific binding .

How are recombinant monoclonal antibodies against proteins like MRAS typically generated?

The generation of recombinant monoclonal antibodies involves several key steps:

  • Sequence Identification: For existing hybridoma cell lines, mRNA transcriptome sequencing is performed to identify antibody variable region sequences . Alternatively, single B cell isolation techniques can be used to identify antigen-specific antibody sequences.

  • Gene Assembly: Variable region sequences are combined with constant region sequences to construct expression vectors for both heavy and light chains .

  • Expression System Selection: Human Expi293F cells are commonly used for expression due to their efficient protein production and human-like post-translational modifications .

  • Transfection and Expression: Expression plasmids are transfected into the selected cells, which then express and secrete the antibody into the culture medium .

  • Purification: The antibodies are typically purified using Protein A Sepharose columns, which have high affinity for the Fc region of antibodies .

A more rapid approach recently developed involves generating antibodies directly from single antigen-specific antibody secreting cells (ASCs) using ferrofluid technology and transcriptionally active PCR (TAP), which can produce functional recombinant antibodies in less than 10 days .

What validation methods should be used to confirm the specificity of a new MRAS recombinant monoclonal antibody?

Comprehensive validation is essential to confirm the specificity of any recombinant monoclonal antibody. For MRAS antibodies, the following validation methods should be employed:

  • Immunofluorescence: Test antibody recognition of endogenous MRAS in relevant cell types, establishing expected subcellular localization patterns .

  • RNA Interference: Perform siRNA-mediated knockdown of MRAS and demonstrate reduced antibody reactivity in both immunofluorescence and immunoblotting assays. This control is crucial for confirming target specificity .

  • Western Blotting: Verify that the antibody recognizes a band of the correct molecular weight in control samples that is absent or diminished in MRAS-depleted samples .

  • Comparison to Established Antibodies: When possible, compare the staining pattern and sensitivity of the new recombinant antibody to well-characterized traditional antibodies against the same target using equivalent antibody concentrations .

  • Cross-Reactivity Testing: Assess potential cross-reactivity with closely related proteins, particularly other RAS family members that share sequence homology with MRAS .

  • Functional Assays: For antibodies intended to detect specific functional states of MRAS (e.g., GTP-bound active form), perform functional validation using activators or inhibitors of relevant signaling pathways .

What are the typical yields and purification efficiencies for MRAS recombinant monoclonal antibodies?

Expression yields for recombinant monoclonal antibodies vary depending on the specific antibody and expression system used, but studies have reported consistent production levels:

Antibody TypeExpression SystemTypical Yield (mg/L)Purification MethodFinal Purity
Full-length IgGExpi293F cells0.8-1.5 mgProtein A Sepharose>95%
Antibody fragments (scFv, Fab)E. coli0.2-0.5 mgImmobilized metal affinity>90%
Bivalent formatsExpi293F cells0.6-1.2 mgProtein A Sepharose>95%

Based on reported data, a typical yield from a 30 mL culture of Expi293F cells is approximately 1.1 mg of purified antibody , which is sufficient for multiple experimental applications including immunofluorescence, immunoblotting, and functional assays. The purification efficiency using Protein A Sepharose columns is generally high, with minimal loss of functional antibody during the purification process .

How can researchers optimize expression systems for difficult-to-express MRAS recombinant antibodies?

Optimization of expression systems for challenging recombinant antibodies requires a multifaceted approach:

  • Vector Engineering: Use codon optimization based on the expression host's preference to enhance translation efficiency. For MRAS antibodies, consider using native signal peptides which can improve secretion efficiency .

  • Host Cell Selection: While Expi293F cells are commonly used, CHO cells may provide better yields for some difficult-to-express antibodies due to their robust protein folding machinery. A comparative evaluation of different expression hosts is recommended for challenging constructs .

  • Culture Conditions Optimization: Implement fed-batch cultures with optimized feed strategies and temperature shifts (typically to 32-34°C) during the production phase to enhance protein folding and reduce aggregation .

  • Co-expression of Chaperones: For antibodies with folding challenges, co-express molecular chaperones such as BiP or PDI to improve folding efficiency and reduce aggregation .

  • Transfection Protocol Optimization: Adjust the DNA:transfection reagent ratio and test different transfection reagents to improve transfection efficiency without compromising cell viability .

  • Post-translational Modification Engineering: For antibodies requiring specific glycosylation patterns, employ glycoengineered cell lines or supplement cultures with glycosylation modulators .

A systematic approach involving the testing of multiple conditions in parallel is recommended to identify the optimal expression parameters for challenging MRAS antibodies.

What strategies exist for developing recombinant antibodies against challenging MRAS epitopes?

Developing antibodies against difficult epitopes of MRAS requires specialized strategies:

  • Peptide Immunization Approach: For conformational or phospho-epitopes, use synthetic peptides containing the specific epitope of interest as immunogens. For phospho-epitopes, implement a phosphatase inhibitor strategy during screening to preserve the phosphorylation state .

  • Phage Display with Tailored Selection: Perform selections with gradual reduction in antigen concentration and increased washing stringency to isolate high-affinity binders to rare epitopes .

  • Single B Cell Technology: Isolate antigen-specific B cells directly from immunized animals or human donors using antigen-coated magnetic beads, followed by single-cell PCR of antibody genes. This approach preserves natural heavy and light chain pairing and can identify rare specificities .

  • Rational Antibody Engineering: For epitopes with high sequence conservation across species, introduce targeted mutations in the complementarity-determining regions (CDRs) of existing antibodies to alter specificity .

  • Negative Selection Strategies: Include closely related proteins (e.g., other RAS family members) in the screening process to deplete cross-reactive antibodies and enrich for MRAS-specific binders .

For phospho-epitopes specifically, validation should include treatment with relevant kinase inhibitors (such as MEK/ERK pathway inhibitors for MRAS) to confirm phospho-specificity, as demonstrated with other phospho-specific antibodies .

How can recombinant antibody technology be used to generate species-specific MRAS antibodies?

Species-specific antibody generation is particularly valuable for comparative studies of MRAS across different model organisms. The following approaches can be employed:

This approach has been successfully demonstrated with other targets, where researchers have modified antibodies to recognize epitopes in specific species while eliminating cross-reactivity with homologous proteins in other species .

What approach should be used for forced degradation studies of MRAS recombinant monoclonal antibodies?

Forced degradation studies are essential for understanding antibody stability and establishing stability-indicating analytical methods. For MRAS recombinant antibodies, a comprehensive approach includes:

  • Stress Conditions: Subject antibodies to various stress conditions including:

    • Thermal stress (40-60°C)

    • pH extremes (pH 3-4 and pH 9-10)

    • Oxidation (0.1-3% hydrogen peroxide)

    • Light exposure (ICH Q1B guidelines)

    • Mechanical stress (agitation, freeze-thaw cycles)

  • Time-Point Analysis: Collect samples at multiple time points (0, 24, 48, 72 hours) to track degradation kinetics under each stress condition .

  • Analytical Methods: Employ multiple orthogonal methods to characterize degradation:

    • Size-exclusion chromatography for aggregation assessment

    • Cation-exchange chromatography for charge variant analysis

    • Capillary electrophoresis for fragmentation detection

    • Mass spectrometry for detailed degradation mapping

  • Functional Analysis: Compare binding activity of stressed and unstressed antibodies using:

    • ELISA to determine EC50 shifts

    • Surface plasmon resonance for binding kinetics

    • Cell-based assays for functional activity preservation

  • Stability-Indicating Method Development: Use degradation profiles to develop and validate analytical methods capable of detecting all relevant degradation products .

The degradation pathways identified through these studies provide critical information for formulation development and stability prediction under real-time storage conditions, which is especially important for maintaining the specificity and functionality of MRAS-targeting antibodies .

What are the considerations for converting single chain antibody fragments to full-length antibodies for MRAS research?

Converting single chain antibody fragments (scFv) to full-length antibodies requires careful consideration of several factors:

  • Framework Selection: Choose appropriate framework regions compatible with the variable domains to minimize aggregation and maximize expression. Human IgG1 frameworks are commonly used for their robust expression and well-characterized effector functions .

  • Linker Design: When designing the junction between variable domains and constant regions, ensure proper folding by using established linker sequences. This is critical for maintaining the structural integrity of the antigen-binding site .

  • Format Optimization: Consider different antibody formats based on research needs:

    • Standard IgG for maximum stability and bivalency

    • Fab fragments for applications requiring smaller size

    • Bispecific formats for simultaneous targeting of MRAS and another protein

  • Expression System Selection: While bacterial systems are often suitable for scFv production, mammalian expression systems (e.g., Expi293F cells) are generally required for proper folding and glycosylation of full-length antibodies .

  • Functional Validation: Compare binding characteristics of the original scFv and the converted full-length antibody using surface plasmon resonance to confirm that specificity and affinity are preserved .

  • Avidity Assessment: Evaluate the impact of increased valency on apparent affinity and function, as conversion from monovalent scFv to bivalent IgG typically enhances target binding through avidity effects .

This conversion process has been successfully demonstrated for various antibodies, with the resulting full-length antibodies maintaining target specificity while gaining advantages in stability, half-life, and functional capabilities .

How can the rapid generation of recombinant antibodies from single ASCs be applied to MRAS research?

Recent advances in generating recombinant antibodies directly from single antigen-specific antibody secreting cells (ASCs) offer significant advantages for MRAS antibody development:

  • Rapid Timeline: This approach allows identification and expression of MRAS-specific monoclonal antibodies in less than 10 days, dramatically accelerating research timelines compared to traditional methods .

  • Methodology Implementation:

    • Isolate CD138+ ASCs from peripheral blood using ferrofluid technology

    • Perform single-cell RT-PCR to amplify immunoglobulin heavy and light chain variable regions

    • Generate linear Ig heavy and light chain gene expression cassettes ("minigenes") via transcriptionally active PCR (TAP)

    • Express functional antibodies without traditional cloning procedures

  • Advantages for MRAS Research:

    • Preserves natural heavy and light chain pairing from responding B cells

    • Enables pre-screening of individual ASCs for MRAS binding before antibody cloning

    • Allows selection of antibodies with desired functional characteristics

    • Facilitates comprehensive analysis of variable region repertoires alongside functional testing

  • Application to MRAS Signaling Studies: This approach could rapidly generate antibodies recognizing specific conformational states of MRAS (GTP-bound active form versus GDP-bound inactive form), enabling dynamic studies of MRAS activation in different cellular contexts .

  • Protocol Optimization for MRAS: The PCR conditions for generating TAP minigenes involve an initial activation step at 95°C for 3 minutes, followed by 35 cycles of 30-second denaturation at 95°C, 30-second annealing at 55°C, and 5-minute extension at 72°C, with a final extension at 72°C for 10 minutes .

This methodology is particularly valuable for generating antibodies against transient or rare epitopes, making it highly applicable for studying dynamic MRAS signaling states in different cellular contexts .

What next-generation sequencing approaches can enhance MRAS recombinant antibody development?

Next-generation sequencing (NGS) technologies offer powerful approaches to enhance recombinant antibody development for MRAS research:

  • Repertoire Analysis: Deep sequencing of B cell repertoires from immunized animals allows identification of expanded B cell clones responding to MRAS immunization, highlighting promising antibody candidates .

  • Hybridoma Deconvolution: For existing hybridoma cell lines producing anti-MRAS antibodies, NGS enables accurate identification of productive heavy and light chain pairs, resolving issues with hybridoma instability or contamination .

  • Paired Chain Sequencing: Single-cell sequencing technologies maintain natural heavy and light chain pairing information, critical for reconstructing functional antibodies with preserved specificity .

  • Affinity Maturation Tracking: NGS can track somatic hypermutation patterns during antibody affinity maturation, identifying key mutations that enhance MRAS binding and informing rational antibody engineering strategies .

  • Integration with Structural Analysis: Combining NGS data with computational structural modeling helps predict antibody-MRAS interactions and guide optimization of binding properties through targeted mutations .

  • Protocol Implementation: The workflow typically involves:

    • Isolation of antigen-specific B cells (using MRAS protein as bait)

    • RNA extraction and library preparation preserving paired VH-VL information

    • Deep sequencing using platforms like Illumina

    • Bioinformatic analysis to identify promising antibody candidates

    • Recombinant expression and functional testing

This integration of NGS with recombinant antibody technology creates a powerful platform for developing highly specific antibodies against different functional domains and conformational states of MRAS .

How should researchers address cross-reactivity issues with MRAS recombinant antibodies?

Cross-reactivity with related RAS family proteins is a common challenge when developing MRAS-specific antibodies. A systematic approach to address this includes:

When cross-reactivity issues arise with an existing antibody, researchers have successfully addressed them by combining the constant regions from one antibody with the variable regions from another, as demonstrated with the CENP-C antibody that exhibited cross-reactivity with rabbit secondary antibodies .

What quality control parameters should be monitored during long-term production of MRAS recombinant monoclonal antibodies?

Consistent quality control is essential for ensuring reproducible results with recombinant antibodies. For MRAS antibodies, key parameters to monitor include:

  • Analytical Parameters:

    • Purity: ≥95% by SDS-PAGE and size-exclusion chromatography

    • Aggregation: <5% high molecular weight species by SEC

    • Charge Variants: Consistent charge profile by IEX-HPLC

    • Glycosylation Pattern: Consistent glycan profile by HILIC or mass spectrometry

    • Endotoxin Levels: <1 EU/mg for cell-based applications

  • Functional Parameters:

    • Binding Affinity: <10% variation in KD by SPR or BLI

    • EC50 in ELISA: <20% variation between batches

    • Epitope Recognition: Consistent epitope mapping profile

    • Specificity Profile: Consistent performance in specificity assays

  • Stability Indicators:

    • Thermal Stability: Consistent Tm by DSC or DSF

    • pH Stability: Consistent pH-dependent stability profile

    • Freeze-Thaw Stability: <10% activity loss after 5 cycles

  • Cell Culture Parameters:

    • Cell Viability: >90% at harvest

    • Metabolite Profiles: Consistent glucose consumption and lactate production

    • Production Kinetics: Consistent time-course of antibody expression

  • Reference Standard Comparison:

    • Maintain a reference standard from an early production batch

    • Compare each new batch to the reference using multiple orthogonal 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 2024 Thebiotek. All Rights Reserved.