mtrm Antibody

Shipped with Ice Packs
In Stock

Description

MTR Antibody: Function, Applications, and Research Findings

Methionine Synthase (MTR) catalyzes the transfer of a methyl group from methylcobalamin (MeCbl) to homocysteine, producing methionine and regenerating cob(I)alamin. This enzyme is essential for maintaining methionine and tetrahydrofolate pools, playing a role in DNA synthesis and epigenetic regulation.

Key Antibody Properties and Applications

SupplierProduct IDTypeReactivityApplications
Abcamab66039Rabbit PolyclonalHuman, Mouse, RatWB, IHC-P, ICC/IF
Sino Biological200844-T46Rabbit PolyclonalHumanICC, IF, IHC-P, IP, WB

Research Insights:

  • Western Blot Validation: Anti-MTR (ab66039) detects a ~90 kDa band in human TE 671 (rhabdomyosarcoma), mouse liver, rat thymus, and Jurkat cell lysates. Treatment with staurosporine or bleomycin did not alter MTR expression levels in HeLa cells, suggesting stability under stress .

  • Pathological Relevance: MTR deficiency is linked to homocystinuria and neurodegenerative disorders. Antibodies enable study of enzyme activity in metabolic diseases.

MTRR Antibody: Target, Mechanism, and Clinical Relevance

Methionine Synthase Reductase (MTRR) regenerates methylcobalamin from cob(I)alamin, ensuring continuous MTR activity. MTRR deficiency causes elevated homocysteine and methylmalonic acid, leading to megaloblastic anemia and neurological issues.

Research Applications:

  • Diagnostic Use: MTRR antibodies enable detection of enzyme expression in tissues (e.g., liver, bone marrow) to diagnose cobalamin deficiency .

  • Therapeutic Monitoring: Polyclonal antibodies are used in ELISA to quantify MTRR levels in clinical samples, aiding in personalized treatment plans .

Comparative Analysis of MTR and MTRR Antibodies

ParameterMTR AntibodyMTRR Antibody
TargetMethionine SynthaseMethionine Synthase Reductase
Key FunctionMethionine synthesis, folate cycleRegeneration of methylcobalamin
Disease LinkHomocystinuria, cancerMegaloblastic anemia, neurological defects
Primary UseMetabolic disorder research, cancer studiesDiagnostic assays, biochemical analyses

Challenges and Future Directions

  • Antibody Specificity: Polyclonal antibodies (e.g., ab66039, HPA038113) may cross-react with homologous proteins, necessitating validation with knockout cell lines .

  • Therapeutic Potential: MTRR-targeted antibodies are not yet in clinical trials, but research focuses on modulating cobalamin metabolism in metabolic disorders .

Table 1: MTR Antibody Performance in Western Blot (ab66039)

SampleTreatmentMTR Band (kDa)
TE 671 (Human)None~90
Mouse LiverNone~90
HeLa (Human)Staurosporine (24hr)~90
Jurkat (Human)Bleomycin (20U/ml)~90

Table 2: MTRR Antibody Suppliers and Applications

SupplierProduct IDApplications
Sino Biological200844-T46ICC, IF, IHC-P, IP, WB
Atlas AntibodiesHPA038113IHC-F, IHC-P
EpigentekA51387-050ELISA

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
mtrm antibody; anon-D52 antibody; CG18543 antibody; Protein matrimony antibody; Cell cycle arrest protein D52 antibody
Target Names
mtrm
Uniprot No.

Target Background

Function
Mtrm Antibody is a Polo kinase inhibitor essential for maintaining G2 arrest during the female meiotic cell cycle. It plays a crucial role in holding heterochromatically paired homologs together from the end of pachytene until metaphase I. This locus exhibits haploinsufficiency for homologous achiasmate segregation and may be required for the maintenance of heterochromatic pairings.
Gene References Into Functions
  1. Research has shown that Mtrm undergoes APC(Cort)-mediated proteasomal degradation. A putative APC/C recognition motif within Mtrm has been identified, and mutations within this motif partially stabilize the protein in the embryo. PMID: 24019759
  2. The binding of Drosophila Polo kinase to its regulator Matrimony is noncanonical and involves two distinct functional domains. PMID: 23479640
  3. During female meiosis, Mtrm regulates Polo's function in promoting nuclear envelope breakdown, ultimately leading to the oocyte's entry into prometaphase. PMID: 18245949
Database Links

KEGG: dme:Dmel_CG18543

STRING: 7227.FBpp0076340

UniGene: Dm.891

Subcellular Location
Nucleus. Chromosome.

Q&A

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

MTR (Methionine synthase) is an essential enzyme that catalyzes the transfer of a methyl group from methylcob(III)alamin (MeCbl) to homocysteine, yielding enzyme-bound cob(I)alamin and methionine in the cytosol. MeCbl, an active form of cobalamin (vitamin B12), functions as a cofactor for methionine biosynthesis. The cob(I)alamin form is regenerated to MeCbl through methyl transfer from 5-methyltetrahydrofolate. MTR operates within a multiprotein complex in the cytosol that includes MMACHC, MMADHC, and MTRR (methionine synthase reductase), which collectively ensure efficient cobalamin utilization for methionine production . Due to its critical role in one-carbon metabolism and amino acid synthesis, MTR is a significant target for studying metabolic disorders and related pathologies.

What types of MTR antibodies are commercially available for research?

Commercial MTR antibodies are primarily available in polyclonal and monoclonal formats. Polyclonal antibodies, such as the rabbit polyclonal ab66039 and ab238483, target different epitopes of the MTR protein. The ab238483 antibody recognizes a recombinant fragment within the human methionine synthase protein from amino acid 900 to the C-terminus , while ab66039 has demonstrated reactivity with human, mouse, and rat samples . These antibodies are validated for multiple applications including Western Blot (WB), Immunohistochemistry-Paraffin (IHC-P), Immunoprecipitation (IP), and Immunocytochemistry/Immunofluorescence (ICC/IF), providing researchers with options for different experimental requirements.

How should researchers validate MTR antibodies before experimental use?

Effective antibody validation requires a multi-faceted approach to ensure specificity and reliability:

  • Knockout/Knockdown Controls: Utilize CRISPR-generated knockout (KO) or RNAi knockdown (KD) cell lines as negative controls. According to research by YCharOS, KO cell lines provide superior control conditions compared to other methods, particularly for Western blot and immunofluorescence applications .

  • Multi-Application Testing: Validate the antibody across multiple applications (WB, IHC-P, ICC/IF) as performance may vary by application context.

  • Cross-Reactivity Assessment: Test against closely related proteins to confirm specificity.

  • Batch Testing: For polyclonal antibodies, test each new lot against a reference standard to account for batch variability.

  • Literature Validation: Review published data citing the specific antibody catalog number to confirm its reliability in similar experimental contexts. For example, ab66039 has been cited in 9 publications, providing additional validation evidence .

What are the optimal conditions for using MTR antibodies in Western blotting?

Optimizing Western blot conditions for MTR antibodies requires careful consideration of several parameters:

ParameterRecommended ConditionsNotes
Sample PreparationStandard RIPA or NP-40 lysis buffer with protease inhibitorsAvoid harsh detergents that might denature the epitope
Sample Loading20-40 μg of total protein per laneMay vary based on MTR expression levels in your samples
Blocking Solution5% non-fat milk or BSA in TBSTTest both to determine optimal background reduction
Primary Antibody Dilution1:500 to 1:2000Optimize for each antibody; start with manufacturer's recommendation
Incubation ConditionsOvernight at 4°C or 2 hours at room temperatureOvernight incubation often yields cleaner results
Detection MethodHRP or fluorescence-based secondary antibodiesChoose based on desired sensitivity and equipment availability
Expected Band Size~140 kDaVerify against positive control samples

Remember to include appropriate positive and negative controls, particularly KO cell lysates if available, as these have been shown to be superior controls for specificity verification .

How can researchers accurately differentiate between specific and non-specific binding when using MTR antibodies?

Differentiating between specific and non-specific binding is critical for accurate data interpretation. A comprehensive approach includes:

  • Knockout Cell Line Controls: The YCharOS study demonstrated that using CRISPR-generated knockout cell lines is the most effective control method, revealing that approximately 12 publications per protein target included data from antibodies that failed to recognize the relevant target protein .

  • Pre-adsorption Controls: Incubate the antibody with excess purified MTR protein before application to determine if the signal disappears (indicating specificity).

  • Multiple Antibodies Approach: Use at least two antibodies targeting different epitopes of the MTR protein. Concordant results strengthen confidence in specificity.

  • Signal Correlation Analysis: Compare antibody signal intensity with known MTR expression levels across different tissues or cell types.

  • Mass Spectrometry Validation: For critical experiments, confirm target identity through immunoprecipitation followed by mass spectrometry analysis, as detailed in the interlaboratory study on monoclonal antibody characterization .

How do recombinant antibodies compare to traditional monoclonal and polyclonal MTR antibodies?

Recombinant antibodies offer several advantages over traditional formats:

  • Consistency: Recombinant antibodies show superior batch-to-batch consistency compared to hybridoma-derived monoclonals and polyclonals, which can vary over time.

  • Performance: The YCharOS study demonstrated that recombinant antibodies outperformed both monoclonal and polyclonal antibodies across multiple assays on average .

  • Specificity: The defined sequence of recombinant antibodies eliminates the risk of hybridoma lines expressing multiple antibodies, which has been documented as a concern with some monoclonal preparations .

  • Renewability: Unlike polyclonal antibodies, which are non-renewable and subject to batch variability, recombinant antibodies can be consistently reproduced.

  • Customizability: Computational approaches like those described in the in silico maturation study can further optimize recombinant antibody affinity and specificity through techniques such as the Metropolis algorithm for sequence space exploration .

Antibody TypeConsistencySpecificityRenewabilityApplication Performance
RecombinantExcellentExcellentExcellentSuperior across multiple applications
MonoclonalGoodGoodGoodVariable, depending on hybridoma stability
PolyclonalPoorVariablePoorOften higher background, batch-dependent

How should researchers address inconsistent results when using MTR antibodies across different experimental platforms?

Inconsistent results across platforms require systematic troubleshooting:

  • Application-Specific Validation: An antibody that works well in Western blot may perform poorly in IHC-P. According to the YCharOS study, 50-75% of proteins were covered by at least one high-performing commercial antibody, but performance varied significantly by application . Verify that your chosen antibody is validated for your specific application.

  • Epitope Accessibility Analysis: In different applications, the target epitope may have different accessibility. For example, the MTR antibody ab238483 recognizes a C-terminal fragment , which might be differently exposed in native versus denatured conditions.

  • Protocol Optimization Matrix: Create a systematic optimization matrix for each platform:

ParameterWestern BlotIHC-PICC/IF
Sample PreparationTest different lysis buffersCompare fixation methods (formalin vs. PFA)Test different fixation times
Antibody ConcentrationDilution seriesDilution seriesDilution series
Blocking AgentCompare milk vs. BSACompare normal sera optionsTest different blocking buffers
Incubation Time1h RT vs. overnight 4°CTest 1h vs. 2h vs. overnightCompare incubation temperatures
Detection SystemHRP vs. fluorescenceDAB vs. fluorescenceTest different mounting media
  • Cross-Validation: Use orthogonal methods (e.g., RT-PCR for mRNA expression) to verify protein expression patterns.

What are the most common false positive scenarios with MTR antibodies and how can they be identified?

False positives can arise from several sources:

  • Cross-Reactivity: The antibody may recognize proteins with similar epitopes. This is particularly common with polyclonal antibodies, which contain multiple antibody species that can introduce false positives and increased background noise .

  • Non-Specific Binding: High concentrations of antibody can lead to non-specific binding. The in silico maturation study demonstrated that even when an antibody has nanomolar affinity, non-specific binding can occur if used at excessive concentrations .

  • Sample Preparation Artifacts: Certain fixation methods may create epitopes that are recognized by the antibody but are not biologically relevant.

Identification strategies include:

  • Using knockout controls as negative controls

  • Performing antibody dilution series to find the optimal concentration that maximizes signal-to-noise ratio

  • Including peptide competition controls

  • Comparing results with multiple antibodies targeting different epitopes of MTR

  • Correlating results with known MTR expression patterns in different tissues or cell types

How can computational approaches improve MTR antibody selection and development?

Computational methods offer powerful tools for antibody optimization:

  • In Silico Maturation: The study on in silico maturation of antibodies demonstrated that Markov chain Monte Carlo simulations with the Metropolis algorithm can effectively explore sequence space to improve antibody affinity. This approach resulted in antibodies with improved binding energies through optimized electrostatic interactions and CDR flexibility .

  • Structure Prediction: Deep learning methods like H3-OPT, AlphaFold2, and protein language models can predict CDR-H3 loop structures with high accuracy, enabling better understanding of antibody-antigen interactions without costly experimental structure determination .

  • Binding Energy Calculations: Molecular dynamics simulations coupled with MM-PBSA (Molecular Mechanics Poisson-Boltzmann Surface Area) calculations can predict binding free energies between antibodies and their targets, guiding the selection of optimal candidates .

  • Epitope Mapping: Computational epitope mapping can identify the most accessible and immunogenic regions of MTR, informing better antibody design.

What emerging technologies are enhancing the validation and application of MTR antibodies?

Several cutting-edge technologies are transforming antibody research:

  • CRISPR-Generated Knockout Cell Lines: As demonstrated by the YCharOS initiative, CRISPR technology enables creation of knockout cell lines that serve as gold-standard negative controls for antibody validation .

  • Top-Down and Middle-Down Mass Spectrometry: These techniques allow characterization of monoclonal antibody primary structures, including modifications, providing more accurate validation than traditional methods .

  • Quantitative Modeling and Simulation Frameworks: Approaches like those used for bamlanivimab provide physiologically-based pharmacokinetic (PBPK) models that can predict antibody distribution in tissues, informing experimental design for in vivo studies .

  • Next-Generation Sequencing of Antibody Repertoires: This approach enables comprehensive analysis of antibody diversity and can inform the development of more specific antibody reagents.

  • Microfluidic and Single-Cell Technologies: These allow for high-throughput screening of antibody binding characteristics at unprecedented resolution.

What resources exist for researchers to share MTR antibody validation data?

Several community resources support antibody validation data sharing:

  • Antibody Registries: Resources like Antibodypedia, the Antibody Registry, and CiteAb catalog antibodies and associated validation data.

  • Collaborative Initiatives: The YCharOS group's open science approach to antibody validation provides a model for community-based validation efforts, having analyzed 614 antibodies targeting 65 proteins and sharing these results publicly .

  • Publication Requirements: Many journals now require detailed antibody validation information, including catalog numbers and RRID identifiers, creating a growing literature-based validation resource.

  • Open Data Repositories: Platforms like Zenodo and FigShare allow for sharing of raw validation data, including images and protocols.

  • Knockout Cell Line Repositories: While not yet comprehensive, efforts to create repositories for knockout cell lines are underway, which would greatly enhance antibody validation capabilities.

How can researchers contribute to improving the reliability of MTR antibody research?

Individual researchers can significantly advance the field through:

  • Rigorous Validation and Reporting: Always include comprehensive validation data with publications, following the five pillars of antibody validation (genetic strategies, orthogonal methods, independent antibodies, expression patterns, and immunocapture-MS).

  • Data Sharing: Submit validation data to public repositories, even negative results showing antibody limitations.

  • Protocol Optimization and Publication: Share optimized protocols for specific applications to reduce variability between labs.

  • Collaboration with Vendors: Provide feedback to commercial suppliers about antibody performance, as demonstrated by the YCharOS initiative where vendor participation led to removal of ~20% of failed antibodies and modification of recommended applications for ~40% .

  • Education and Training: Implement proper training in antibody selection, validation, and use in research laboratories, addressing a key gap identified in the antibody characterization crisis .

By following these guidelines and contributing to community efforts, researchers can help address the estimated $0.4-1.8 billion annual losses in the United States alone attributed to inadequately characterized antibodies .

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.