MBP Antibody

Maltose-Binding protein, Mouse Antibody
Shipped with Ice Packs
In Stock

Description

Definition and Biological Role

MBP antibodies recognize epitopes on myelin basic protein, which stabilizes myelin compaction. While MBP is essential for maintaining myelin integrity , autoantibodies against it may contribute to demyelination through immune complex formation, complement activation, and cytokine induction .

Autoimmune Disorders

  • Multiple Sclerosis (MS): Elevated anti-MBP IgG/IgM levels are detected in MS patients, though their diagnostic specificity remains debated . A subset of MS patients shows IgA antibodies targeting conformational MBP epitopes, correlating with rapid cognitive decline .

  • Systemic Lupus Erythematosus (SLE): Catalytic MBP antibodies (abzymes) with protease activity are observed, potentially degrading MBP and exacerbating neuroinflammation .

  • Experimental Autoimmune Encephalomyelitis (EAE): MBP immunization in mice increases B-cell infiltration and spinal cord damage, suggesting a pathogenic role .

Healthy Individuals

Natural IgM/IgG anti-MBP antibodies exist in healthy sera, facilitating low-level cytokine production (e.g., TNF-α, IL-10) without triggering overt disease .

Comparative Analysis of MBP Antibodies in Healthy vs. MS Sera

ParameterHealthy SeraMS SeraSource
IgG ReactivityModerateSlightly higher (p=0.10)
IgM ReactivitySignificant depositionSimilar to controls
Cytokine InductionTNF-α, IL-10TNF-α, IL-10, IFN-γ, IL-5
Pathogenic PotentialNon-destructiveCorrelated with demyelination

Mechanistic Insights

  • Immune Complex Formation: MBP-antibody complexes deposit on monocytes, activate complement (C3 fragments), and induce pro-inflammatory cytokines .

  • Epitope Specificity: Antibodies recognize conformational epitopes; denatured MBP fails to trigger responses .

  • Catalytic Activity: Abzymes in MS/SLE hydrolyze MBP at immunodominant sites, potentially accelerating myelin breakdown .

Biomarker Potential

  • Anti-MBP IgA in CSF/serum may identify aggressive MS subtypes .

  • Peptide-based ELISAs using MBP(76–116) improve IgM detection sensitivity .

Therapeutic Strategies

  • B-Cell Depletion: Reduces MBP-reactive B cells and neuroinflammation in EAE models .

  • Complement Inhibition: Blocks antibody-mediated myelin damage .

Controversies and Challenges

  • Detection Variability: Discrepancies arise from assay formats (e.g., denatured vs. native MBP) .

  • Pathogenic Ambiguity: Natural autoantibodies in healthy individuals challenge the direct link to disease .

  • Therapeutic Targeting: Modulating antibody responses without compromising immune surveillance remains complex .

Future Directions

  • Standardize assays for native MBP-reactive antibodies.

  • Investigate abzymes as therapeutic targets in MS and SLE.

  • Explore genetic and environmental triggers of pathogenic antibody subsets.

Product Specs

Introduction
Maltose Binding Protein, part of E. coli's maltose/maltodextrin system, facilitates the uptake and breakdown of maltodextrins. This intricate system employs various proteins and complexes for efficient regulation and transport. MBP often enhances the yield and solubility of its fusion partner polypeptides.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
A 1 mg/ml solution prepared in a buffer containing PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
Store at 4°C for up to 1 month. For extended storage, keep at -20°C. Avoid repeated freezing and thawing cycles.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
The MBP antibody has undergone ELISA and Western blot testing to ensure its specificity and reactivity. However, optimal working concentrations may vary depending on the specific application. It is recommended to perform a titration to determine the optimal reagent concentration for your experiment.
Synonyms
MBP, Maltose-Binding protein.
Purification Method

MBP antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.

Type
Mouse Antibody Monoclonal.
Clone
P10C12AT.
Immunogen
Anti MBP mAb is derived from hybridization of mouse SP2/0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant MBP purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is Myelin Basic Protein and why is it important in research?

Myelin Basic Protein (MBP) is a critical structural component of the myelin sheath that insulates neuronal axons in the central nervous system. In humans, the canonical MBP protein has 304 amino acid residues with a molecular mass of approximately 33.1 kDa . MBP is predominantly expressed in oligodendrocytes and is found abundantly in brain regions including the hippocampus, cerebral cortex, cerebellum, and caudate . The protein's importance in research stems from its role in myelin formation and stability, making it a key marker for studying neurological development, aging processes, and demyelinating disorders such as multiple sclerosis. Up to six different isoforms of MBP have been reported, adding to its complexity and research significance . Studies examining MBP often focus on its relationship to neurological diseases, developmental biology, and nervous system function.

What types of MBP antibodies are available for research, and how do they differ?

Researchers can utilize several types of MBP antibodies, each with distinct characteristics for different experimental applications:

  • Monoclonal antibodies: These antibodies, such as clones 2A1, 3D7 and 932908 , offer high specificity for MBP epitopes and demonstrate consistent performance across experiments. They are produced from single B-cell clones, ensuring homogeneity in binding properties.

  • Polyclonal antibodies: These recognize multiple epitopes on the MBP molecule, providing broader detection but potentially more variability between lots.

  • Species-specific vs. cross-reactive antibodies: Some antibodies are designed to detect MBP from specific species, while others like clone 932908 can detect human, mouse, and rat MBP simultaneously .

  • Application-optimized antibodies: Certain antibodies perform optimally in specific applications such as western blotting, immunoprecipitation, or immunohistochemistry, according to their validation data .

The choice between these antibody types depends on experimental needs, with monoclonals preferred for consistent, reproducible results, while polyclonals might provide higher sensitivity when detecting native proteins across multiple species.

How do I determine the appropriate MBP antibody dilution for my experiments?

Determining the optimal dilution for MBP antibodies requires systematic titration based on your specific application, sample type, and detection method. For western blot applications, starting dilutions typically range from 0.1-1 μg/mL as demonstrated in experiments with mouse and rat brain (cerebellum) tissue samples . For immunocytochemistry, higher concentrations around 10 μg/mL have been effectively used for detecting MBP in fixed rat cortical stem cells differentiated to oligodendrocytes .

A methodical approach to antibody titration includes:

  • Initial range finding: Test a broad range of dilutions (e.g., 1:100, 1:500, 1:1000, 1:5000) with positive and negative control samples.

  • Fine tuning: Once an approximate working range is established, test narrower dilution intervals.

  • Signal-to-noise optimization: Select the dilution that provides the strongest specific signal with minimal background.

  • Validation across replicates: Verify that your chosen dilution performs consistently across multiple experimental repeats.

The optimal dilution is often application-dependent - ELISA typically requires more concentrated antibody solutions than western blotting. In published studies, researchers have used concentrations ranging from 0.1 μg/mL for western blotting to 10 μg/mL for immunofluorescence staining .

How do I optimize western blot protocols for MBP detection?

Optimizing western blot protocols for MBP detection requires special considerations due to its small size isoforms (15-22 kDa) and membrane association. Based on published methodologies, an optimized protocol includes:

  • Sample preparation:

    • For tissue samples: Homogenize brain tissue (particularly cerebellum) in a buffer containing protease inhibitors

    • Use 2-5 μg of cell lysates or 1-2 μg of purified proteins for optimal detection

  • Gel electrophoresis:

    • Use Tris/Glycine/SDS-PAGE gels with appropriate percentage (12-15%) for resolving small proteins

    • Include molecular weight markers spanning 10-25 kDa range to properly identify MBP isoforms

  • Transfer conditions:

    • Use PVDF membranes for optimal protein binding

    • Reduce methanol concentration to 10% in transfer buffer for better transfer of small proteins

    • Consider semi-dry transfer systems for efficient transfer of smaller proteins

  • Blocking and antibody incubation:

    • Block with 5% nonfat dry milk in TBS-T (50 mM Tris-HCl, pH 7.6, 150 mM NaCl, 0.005% Tween-20)

    • Incubate with anti-MBP antibodies at 0.1-1 μg/mL concentration

    • Use species-appropriate secondary antibodies (e.g., IR Dye 800-conjugated anti-mouse for mouse monoclonals)

  • Detection optimization:

    • For enhanced sensitivity, consider chemiluminescent detection systems

    • For quantitative analysis, fluorescence-based systems like Odyssey (LI-COR) provide better linearity

Researchers should note that MBP typically appears as multiple bands between 15-22 kDa due to its various isoforms, with experiment conditions set to reducing conditions using appropriate buffer systems .

What are the key considerations for immunohistochemical detection of MBP?

Successful immunohistochemical detection of MBP requires careful attention to tissue preparation, fixation methods, and antigen retrieval techniques:

  • Tissue preparation and fixation:

    • Immersion fixation in 4% paraformaldehyde provides good preservation of MBP epitopes

    • For paraffin-embedded sections, careful deparaffinization and hydration are essential

    • For frozen sections, post-fixation in acetone or methanol may enhance antibody binding

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval using citrate buffer (pH 6.0) often improves MBP detection

    • For some tissues, enzymatic retrieval with proteinase K might be more effective

    • Optimization of retrieval time and temperature is critical for balancing signal strength with tissue integrity

  • Blocking and antibody incubation:

    • Block with serum from the same species as the secondary antibody (typically 5-10%)

    • Include 0.1-0.3% Triton X-100 for enhanced antibody penetration in tissue sections

    • Validated protocols have used anti-MBP antibodies at concentrations of 10 μg/mL for optimal staining

  • Detection systems:

    • For fluorescent detection, secondary antibodies conjugated with fluorophores like NorthernLights 557 have been effective

    • For chromogenic detection, HRP-based systems with DAB substrate provide permanent staining

    • Counterstaining with DAPI helps to visualize nuclei in relation to MBP expression

  • Controls and validation:

    • Include positive controls (known MBP-expressing tissues like cerebellum)

    • Include negative controls (primary antibody omission and tissues without MBP expression)

    • When possible, validate results with multiple antibodies targeting different MBP epitopes

Researchers should expect to observe MBP staining primarily in white matter tracts, with localization to cell surfaces, cytoplasm, and occasionally nuclei of oligodendrocytes as demonstrated in studies with human brain cortex samples .

How can I use MBP antibodies in ELISA assays for quantitative measurements?

Implementing ELISA assays with MBP antibodies requires careful optimization of multiple parameters to achieve accurate and reproducible quantification. A methodological approach includes:

  • Plate coating optimization:

    • Use purified MBP antigen at concentrations ranging from 0.016-2 μg per well in 0.05 M sodium carbonate buffer (pH 9.6)

    • Perform serial dilutions to establish a standard curve

    • Incubate plates for 2 hours at room temperature for optimal antigen binding

  • Blocking conditions:

    • Block wells overnight with 200 μL of blocking buffer (1% BSA in TBS-T) at 4°C to minimize non-specific binding

    • Ensure complete blocking by washing thoroughly with TBS-T before adding antibodies

  • Antibody concentration optimization:

    • Test multiple antibody concentrations (0.1, 1, and 10 μg/mL) to determine optimal signal-to-noise ratio

    • Incubate antibodies in blocking buffer for 1 hour at room temperature

    • For sandwich ELISA, carefully select capture and detection antibodies recognizing different epitopes

  • Detection system selection:

    • Use HRP-conjugated secondary antibodies (e.g., goat anti-mouse IgG1) for colorimetric detection

    • Develop with TMB substrate solution for approximately 20 minutes at room temperature

    • Stop the reaction with 0.18 M H₂SO₄ and read absorbance at 450 nm using a microplate reader

  • Quantification and analysis:

    • Generate a standard curve using known MBP concentrations

    • Ensure samples fall within the linear range of the standard curve

    • Calculate concentrations using appropriate regression analysis

This methodology has been successfully employed to detect MBP and anti-MBP antibodies in various experimental contexts, including autoimmune encephalomyelitis models .

How can I use MBP antibodies to study demyelinating diseases and models?

MBP antibodies serve as powerful tools for investigating demyelinating diseases through various advanced methodological approaches:

  • Animal model characterization:

    • In experimental autoimmune encephalomyelitis (EAE) models, MBP antibodies can quantify demyelination extent in the central nervous system

    • Use immunohistochemistry with anti-MBP antibodies to visualize and quantify myelin loss in spinal cord sections

    • Compare MBP staining patterns between healthy controls and disease models to assess pathology progression

  • Correlation of antibody levels with disease severity:

    • Measure circulating anti-MBP antibody levels using ELISA to correlate with clinical scores in disease models

    • Track changes in anti-MBP antibody titers throughout disease course to understand immune response dynamics

    • Studies have demonstrated that MBP antibody levels directly correlate with immune cell infiltration in the spinal cord (p < 0.001)

  • Mechanistic investigations:

    • Use double immunofluorescence with MBP antibodies and immune cell markers to visualize the interaction between inflammation and demyelination

    • Employ flow cytometry in combination with tissue staining to quantify B cell responses against MBP

    • Monitor changes in MBP expression patterns following therapeutic interventions

  • Translational research applications:

    • Compare findings from animal models with human patient samples to validate disease mechanisms

    • Use MBP antibodies to assess remyelination following experimental treatments

    • Investigate the relationship between MBP-reactive B cells and disease progression as potential therapeutic targets

This multifaceted approach provides comprehensive insights into demyelinating pathologies, offering both quantitative measurements of disease progression and qualitative assessment of myelin integrity, which are crucial for evaluating potential therapeutic interventions.

What techniques can I use to validate MBP antibody specificity in my research?

Validating MBP antibody specificity is critical for ensuring reliable, reproducible results. A comprehensive validation strategy should include multiple complementary approaches:

  • Western blot validation:

    • Test antibody against purified recombinant MBP and native MBP from tissue lysates

    • Include positive controls (brain tissue, specifically cerebellum) and negative controls (tissues not expressing MBP)

    • Verify the presence of appropriate molecular weight bands (15-22 kDa for MBP isoforms)

    • Use different reducing and non-reducing conditions to assess epitope accessibility

  • Immunoprecipitation cross-validation:

    • Perform IP with one anti-MBP antibody and detect with another targeting a different epitope

    • Validate precipitated proteins by mass spectrometry to confirm identity

    • Compare IP efficiency between different antibody clones (e.g., 2A1 and 3D7)

  • Immunohistochemistry/immunofluorescence validation:

    • Compare staining patterns with established myelin markers

    • Perform antibody pre-absorption with purified MBP to demonstrate specificity

    • Use knockout or knockdown models when available as negative controls

    • Verify proper anatomical distribution (e.g., white matter tracts in brain and spinal cord)

  • Competitive ELISA:

    • Perform assays with and without pre-incubation with purified MBP

    • Construct inhibition curves to demonstrate specific binding

    • Test cross-reactivity with related myelin proteins (e.g., PLP, MOG)

  • Knockout/knockdown controls:

    • When possible, test antibodies on tissues or cells with confirmed MBP knockout/knockdown

    • Compare staining intensity between wild-type and knockout samples

    • This represents the gold standard for antibody validation

This multilateral approach ensures that observed signals truly represent MBP rather than non-specific binding or cross-reactivity with other proteins, significantly enhancing research reliability.

How can I differentiate between MBP isoforms using antibodies?

Differentiating between MBP isoforms requires strategic selection and application of antibodies, combined with appropriate experimental techniques:

This systematic approach allows researchers to distinguish between the six reported MBP isoforms , providing insights into their differential expression and potentially distinct functions in neurological development and pathology.

What are common issues when using MBP antibodies and how can I resolve them?

Researchers frequently encounter several challenges when working with MBP antibodies. Here are methodological solutions to these common problems:

  • Weak or absent signal in western blots:

    • Problem: MBP may be lost during transfer due to its small size (15-22 kDa)

    • Solution: Use 0.2 μm PVDF membranes instead of 0.45 μm; reduce transfer voltage/time; consider semi-dry transfer systems; increase antibody concentration to 0.5-1 μg/mL

  • Multiple unexpected bands:

    • Problem: Could indicate non-specific binding or MBP degradation

    • Solution: Increase blocking stringency (5% milk or BSA); include protease inhibitors during sample preparation; verify antibody specificity with positive controls; note that multiple bands between 15-22 kDa are expected due to natural MBP isoforms

  • High background in immunohistochemistry:

    • Problem: Non-specific binding or insufficient blocking

    • Solution: Extend blocking time (overnight at 4°C); include 0.1-0.3% Triton X-100 in blocking buffer; titrate primary antibody (test 1-10 μg/mL range); increase washing duration and number of washes

  • Inconsistent ELISA results:

    • Problem: Variable antigen coating or antibody binding

    • Solution: Standardize antigen coating concentration (0.016-2 μg per well); ensure consistent incubation times and temperatures; use freshly prepared reagents; include standard curves on each plate; consider sandwich ELISA format for improved consistency

  • Poor immunoprecipitation efficiency:

    • Problem: Inefficient antibody binding to MBP in solution

    • Solution: Optimize buffer conditions (test different salt concentrations); pre-clear lysates; increase antibody amount (10 μg per IP reaction); extend incubation time with antibody-bead complexes

  • Reduced antibody performance over time:

    • Problem: Antibody degradation during storage

    • Solution: Aliquot antibodies to avoid freeze-thaw cycles; store according to manufacturer recommendations; add preservatives for diluted working stocks; validate each new lot against previously working antibodies

Implementing these technical refinements can significantly improve experimental outcomes when working with MBP antibodies across different applications.

How does sample preparation affect MBP antibody detection efficiency?

Sample preparation critically influences the detection of MBP by antibodies across different experimental platforms. Optimizing sample preparation involves:

  • Tissue preservation methods:

    • Fresh vs. fixed samples: Fresh or snap-frozen tissues typically preserve MBP epitopes better than formalin-fixed paraffin-embedded (FFPE) samples

    • Fixation impact: For immunohistochemistry, short fixation times (4-8 hours) with 4% paraformaldehyde preserve MBP antigenicity better than longer protocols

    • Post-fixation processing: Cryoprotection with sucrose gradients prior to freezing helps maintain tissue morphology without compromising antibody binding

  • Protein extraction techniques:

    • Lysis buffer composition: For MBP extraction from tissues, buffers containing 50 mM NaH₂PO₄ (pH 7.4), 1% Triton X-100, 300 mM NaCl, and protease inhibitors have proven effective

    • Sonication parameters: Three 20-second sonication cycles at 1-minute intervals on ice optimally solubilize membrane-associated MBP

    • Cell debris removal: Centrifugation at 10,000 rpm for 5 minutes effectively separates soluble proteins from cellular debris

  • Protein denaturation considerations:

    • Reducing conditions: SDS-PAGE under reducing conditions (with β-mercaptoethanol or DTT) is typically required for optimal MBP detection in western blots

    • Heat denaturation: Gentle heating (70°C for 10 minutes) rather than boiling prevents MBP aggregation

    • Sample dilution: Diluting concentrated samples 1:10 with lysis buffer improves detection in immunoprecipitation experiments

  • Storage impact on sample integrity:

    • Temperature effects: Store protein extracts at -80°C rather than -20°C to maintain MBP integrity

    • Freeze-thaw cycles: Limit to maximum of 2-3 cycles to prevent degradation

    • Protective additives: Adding 10% glycerol to samples prior to freezing helps preserve protein structure

These methodological refinements significantly enhance detection sensitivity and specificity across western blotting, immunohistochemistry, ELISA, and immunoprecipitation applications with MBP antibodies.

How do post-translational modifications affect MBP antibody recognition?

Post-translational modifications (PTMs) of Myelin Basic Protein significantly impact antibody recognition through various mechanisms. Understanding these effects is crucial for experimental design and data interpretation:

  • Impact of specific PTMs on epitope accessibility:

    • Phosphorylation: MBP contains multiple potential phosphorylation sites that can alter protein conformation and epitope exposure

    • Methylation and acetylation: These modifications affect the charge distribution of MBP, potentially masking or revealing epitopes

    • Protein cleavage: Proteolytic processing of MBP can generate fragments that may not be recognized by antibodies targeting epitopes in the cleaved regions

  • Antibody selection strategies:

    • Modification-specific antibodies: Some antibodies specifically recognize phosphorylated, methylated, or acetylated forms of MBP

    • Conformation-dependent antibodies: These recognize structural epitopes that may be altered by PTMs

    • Pan-MBP antibodies: These target conserved regions less affected by PTMs and detect multiple forms of the protein

  • Experimental considerations:

    • Phosphatase treatment: Pre-treating samples with phosphatases before immunoblotting can determine if phosphorylation affects antibody recognition

    • Enrichment techniques: Phospho-protein enrichment columns can isolate modified MBP forms for selective analysis

    • Blocking peptide controls: Using both modified and unmodified peptides as blocking controls can verify PTM-specific recognition

  • Analytical methods for PTM characterization:

    • 2D gel electrophoresis: Separates MBP isoforms based on both molecular weight and isoelectric point, revealing charge changes from PTMs

    • Mass spectrometry validation: Confirms the presence and location of specific modifications on MBP molecules

    • Western blot comparison: Using multiple antibodies targeting different epitopes or modifications provides complementary information

Understanding these interactions is particularly relevant for studying MBP in disease contexts like multiple sclerosis, where altered PTM patterns may affect both antibody recognition and biological function of the protein .

How are MBP antibodies used in multiple sclerosis and related disease research?

MBP antibodies serve as crucial research tools in multiple sclerosis (MS) and related demyelinating disease investigations, offering multiple methodological applications:

  • Quantitative assessment of demyelination:

    • Tissue analysis: MBP immunostaining quantifies myelin loss in brain and spinal cord tissues from experimental models and human samples

    • Comparative studies: Researchers compare MBP staining patterns between normal and diseased tissues to measure demyelination extent

    • Temporal tracking: Sequential analysis reveals progression of demyelination and potential remyelination during disease course or treatment

  • Analysis of autoimmune responses:

    • Anti-MBP antibody detection: ELISA assays using purified MBP quantify autoantibody levels in patient sera or animal models

    • Correlation studies: Research shows MBP antibody levels directly correlate with immune cell infiltration in the spinal cord (p < 0.001)

    • B-cell reactivity: Flow cytometry combined with MBP antigens identifies MBP-reactive B cells in circulation and CNS infiltrates

  • Therapeutic development applications:

    • Remyelination assessment: Anti-MBP antibodies monitor myelin restoration following experimental treatments

    • Target engagement: Verify whether novel therapeutics successfully engage with their molecular targets in myelin

    • Biomarker discovery: Correlate changes in MBP and anti-MBP antibody levels with clinical outcomes to identify potential biomarkers

  • Advanced pathophysiological investigations:

    • Cellular localization studies: Double immunofluorescence with cell-type markers reveals relationships between MBP-expressing cells and infiltrating immune cells

    • Mechanistic experiments: In experimental autoimmune encephalomyelitis (EAE) models, anti-MBP antibodies help elucidate the contributions of B cells to pathogenesis

    • Translational analyses: Compare findings between animal models and human samples to validate disease mechanisms

These methodological approaches have collectively contributed to our understanding that reactive B cells and MBP-induced antibodies significantly contribute to demyelinating disease pathogenesis, potentially informing new therapeutic strategies targeting these mechanisms .

How can I use MBP antibodies to study the developmental biology of myelination?

MBP antibodies provide powerful tools for investigating myelination developmental biology through various methodological approaches:

  • Temporal expression analysis:

    • Developmental time course: Use western blotting with anti-MBP antibodies to quantify MBP expression across different developmental stages

    • Isoform transitions: Track changes in MBP isoform ratios during development, as different isoforms predominate at different developmental stages

    • Regional differences: Compare MBP expression timing across brain regions to map myelination progression

  • Cellular differentiation studies:

    • Oligodendrocyte lineage tracking: Use MBP antibodies to identify mature oligodendrocytes in mixed neural cultures

    • Differentiation assays: Monitor MBP expression in cortical stem cells during differentiation to oligodendrocytes (as demonstrated in rat models)

    • Co-localization analysis: Combine MBP staining with markers of oligodendrocyte precursor cells to visualize maturation sequence

  • High-resolution spatial characterization:

    • Subcellular localization: Confocal microscopy with MBP antibodies reveals protein distribution in cell bodies and processes during myelin formation

    • Myelin sheath visualization: Immunoelectron microscopy shows MBP integration into compact myelin

    • In vivo imaging: Two-photon microscopy with fluorescently labeled antibodies can monitor myelination in living tissue

  • Functional integration experiments:

    • In vitro myelination assays: Co-culture neurons with oligodendrocytes and use MBP antibodies to quantify myelin formation

    • 3D culture systems: Assess MBP expression in brain organoids to model developmental myelination

    • Perturbation studies: Examine how genetic or pharmacological interventions affect MBP expression patterns

  • Comparative developmental biology:

    • Cross-species analysis: Compare MBP expression timing and patterns across human, mouse, rat, and other species

    • Evolutionary perspectives: Study conservation of MBP structure and function across vertebrate lineages

    • Disease model comparisons: Investigate how developmental MBP expression differs in models of developmental disorders

This multifaceted approach provides comprehensive insights into the spatiotemporal dynamics of myelination during development, offering both quantitative measurements and qualitative assessment of myelin formation and maturation.

What is the relationship between MBP antibodies and experimental autoimmune encephalomyelitis (EAE)?

The relationship between MBP antibodies and experimental autoimmune encephalomyelitis (EAE) is multifaceted, involving both mechanistic contributions to disease pathogenesis and applications as research tools:

  • Pathogenic mechanisms of anti-MBP antibodies:

    • Demyelination induction: Anti-MBP antibodies contribute directly to myelin damage by binding to MBP in intact myelin

    • Immune amplification: These antibodies enhance infiltration of inflammatory cells into the CNS, with studies showing direct correlation between antibody levels and immune cell infiltration (p < 0.001)

    • Blood-brain barrier effects: Anti-MBP antibodies may contribute to blood-brain barrier disruption, facilitating further immune cell entry

  • MBP as an immunogen for EAE induction:

    • Immunization protocols: EAE can be induced in C57BL/6 mice using purified MBP with adjuvants like poly(I:C)

    • B cell activation: MBP immunization significantly increases B cell populations producing anti-MBP antibodies

    • Clinical manifestations: The resulting disease mimics aspects of multiple sclerosis, with progressive neurological dysfunction

  • Monitoring disease progression:

    • Antibody titer measurements: ELISA quantification of anti-MBP antibodies serves as a biomarker for disease activity

    • Histopathological assessment: Anti-MBP immunostaining reveals demyelinated areas in spinal cord sections

    • Correlation analysis: Studies demonstrate that MBP antibody levels directly correlate with clinical scores and histopathological findings

  • Therapeutic testing applications:

    • Treatment evaluation: Measuring changes in anti-MBP antibody levels following therapeutic interventions

    • Mechanistic insights: Determining whether treatments function by reducing anti-MBP antibody production or by other mechanisms

    • Predictive biomarkers: Testing whether baseline anti-MBP antibody levels predict treatment response

  • Research methodology considerations:

    • Antibody generation: MBP-immunized mice develop high-titer anti-MBP antibodies that can be harvested for research

    • Epitope mapping: Analyzing which MBP regions are targeted by autoantibodies provides insights into pathogenic mechanisms

    • T-B cell interactions: Studying how T cells specific for MBP help B cells produce anti-MBP antibodies

How can I use MBP antibodies for immunoprecipitation experiments?

Successful immunoprecipitation (IP) of Myelin Basic Protein requires optimization of several critical parameters. Here's a comprehensive methodology:

  • Sample preparation optimization:

    • Cell/tissue lysis: For bacterial expression systems, resuspend pelleted cells in lysis buffer (50 mM NaH₂PO₄ [pH 7.4], 1% Triton X-100, 300 mM NaCl, protease inhibitors)

    • Lysis enhancement: Incubate suspension at 37°C for 30 minutes followed by sonication (three 20-second cycles at 1-minute intervals on ice)

    • Clarification: Remove cell debris by centrifugation at 10,000 rpm for 5 minutes

    • Sample dilution: Dilute lysates 1:10 with lysis buffer to optimize antibody binding conditions

  • Immunoprecipitation protocol:

    • Antibody selection: Use monoclonal antibodies (e.g., clones 2A1 or 3D7) for high specificity

    • Antibody coupling: Bind approximately 10 μg of anti-MBP antibody to Protein G Dynabeads or similar magnetic beads

    • Pre-clearing: Pre-clear lysates with beads alone to reduce non-specific binding

    • Immunoprecipitation reaction: Incubate antibody-bead complexes with prepared lysates for 2-4 hours at 4°C with gentle rotation

  • Washing and elution strategies:

    • Wash optimization: Perform 3-5 washes with decreasing salt concentration to maintain specific interactions while removing non-specific binding

    • Elution conditions: Elute bound proteins using either low pH buffer, high salt, or SDS sample buffer depending on downstream applications

    • Protein recovery: For maximum recovery, elute with SDS sample buffer at 70°C for 10 minutes

  • Analysis and validation:

    • SDS-PAGE separation: Separate immunoprecipitated proteins by SDS-PAGE

    • Detection methods: Analyze by Coomassie blue staining for total protein visualization

    • Western blot verification: Confirm MBP identity using western blotting with a different anti-MBP antibody (e.g., rabbit polyclonal) to avoid detecting the IP antibody

This methodology has been successfully employed to immunoprecipitate both native MBP and MBP fusion proteins (such as MBP-Ror2) from various expression systems, demonstrating the versatility of anti-MBP antibodies for protein complex isolation and characterization .

How can I detect MBP in flow cytometry experiments?

Optimizing flow cytometry protocols for MBP detection requires careful consideration of its intracellular location and the preservation of cellular structure. Here's a methodological approach:

  • Sample preparation considerations:

    • Cell isolation: For CNS tissue, use gentle enzymatic digestion (papain or dispase) followed by mechanical dissociation

    • Cell viability: Include viability dyes (e.g., propidium iodide) to exclude dead cells which can bind antibodies non-specifically

    • Surface marker staining: Perform surface marker staining (e.g., O4 for oligodendrocytes) before fixation when possible

  • Fixation and permeabilization optimization:

    • Fixation protocol: Fix cells with 2-4% paraformaldehyde for 15-20 minutes at room temperature to preserve cellular architecture

    • Permeabilization methods: Compare detergent-based permeabilization (0.1-0.3% Triton X-100) with alcohol-based methods (70-90% methanol) for optimal MBP detection

    • Buffer composition: Use buffers containing 2% FBS or BSA to reduce non-specific binding

  • Antibody staining protocol:

    • Antibody selection: Choose monoclonal antibodies validated for flow cytometry applications

    • Titration: Determine optimal antibody concentration by testing serial dilutions (typically 0.1-10 μg/mL)

    • Incubation conditions: Extend incubation time to 45-60 minutes at room temperature or overnight at 4°C for improved signal

    • Washing steps: Include multiple washing steps with excess buffer volume to reduce background

  • Controls and validation:

    • Isotype controls: Include matched isotype controls at the same concentration as the primary antibody

    • Fluorescence minus one (FMO): Prepare controls with all antibodies except anti-MBP to establish gating boundaries

    • Positive controls: Include cell populations known to express high levels of MBP (e.g., mature oligodendrocytes)

    • Blocking controls: Pre-incubate antibody with purified MBP to confirm specificity

  • Analytical considerations:

    • Compensation: Carefully set up compensation when using multiple fluorochromes

    • Gating strategy: First gate on viable cells, then identify relevant populations using lineage markers before assessing MBP expression

    • Quantification methods: Record both percentage of positive cells and mean fluorescence intensity (MFI)

This approach has been successfully employed in experimental autoimmune encephalomyelitis studies to quantify B cells and assess MBP reactivity, providing valuable insights into disease mechanisms .

What are the considerations for using MBP antibodies in live cell imaging?

Live cell imaging with MBP antibodies presents unique challenges due to the intracellular localization of MBP and the need to maintain cell viability. Here's a methodological framework for successful implementation:

  • Antibody fragment selection and modification:

    • Fragment types: Use Fab or scFv fragments rather than whole IgG for better penetration and reduced interference with cellular functions

    • Fluorophore conjugation: Directly conjugate antibody fragments with bright, photostable fluorophores (e.g., Alexa Fluor dyes) rather than using secondary detection

    • Validation: Verify that conjugation doesn't alter binding specificity or affinity through parallel fixed-cell experiments

  • Cell membrane permeabilization strategies:

    • Gentle detergents: Use low concentrations (0.01-0.05%) of digitonin or saponin for selective membrane permeabilization while maintaining cell viability

    • Reversible permeabilization: Employ systems allowing temporary pore formation for antibody entry followed by resealing

    • Alternative delivery methods: Consider microinjection, cell-penetrating peptides, or liposome-based delivery systems for antibody internalization

  • Live imaging optimization:

    • Phototoxicity minimization: Use low laser power and longer exposure times to reduce phototoxicity

    • Environment control: Maintain physiological temperature, pH, and CO2 levels during imaging

    • Temporal resolution: Balance acquisition frequency with cell viability concerns

    • Z-stack parameters: Optimize step size and range to capture MBP distribution while minimizing light exposure

  • Controls and validation:

    • Viability markers: Include non-toxic viability indicators to monitor cell health during imaging

    • Specificity controls: Use non-binding antibody fragments with matched fluorophores to control for non-specific signal

    • Functional assays: Verify that antibody binding doesn't disrupt normal MBP function or oligodendrocyte behavior

  • Alternative approaches:

    • MBP-fluorescent protein fusions: Consider transfection with MBP-GFP fusion constructs as an alternative to antibody labeling

    • Hybrid approaches: Combine live imaging of other markers with post-fixation MBP staining for correlation

    • Super-resolution techniques: Implement stimulated emission depletion (STED) or structured illumination microscopy (SIM) for enhanced resolution of myelin structures

This methodology enables visualization of MBP dynamics in living oligodendrocytes, providing insights into myelination processes that complement findings from fixed tissue studies.

What are the current limitations of MBP antibodies in research?

Despite their invaluable contributions to neuroscience research, MBP antibodies present several methodological limitations that researchers should consider:

  • Epitope accessibility challenges:

    • MBP's integration within compact myelin makes epitopes inaccessible in some contexts

    • Post-translational modifications can mask antibody binding sites, particularly in disease states where MBP modification patterns change

    • Different fixation methods variably affect epitope preservation, requiring protocol optimization

  • Isoform detection limitations:

    • Many commercially available antibodies cannot differentiate between the six reported MBP isoforms

    • Cross-reactivity between isoforms complicates interpretation of expression patterns

    • Lack of isoform-specific antibodies limits understanding of their distinct functions

  • Species cross-reactivity considerations:

    • While some antibodies detect MBP across human, mouse, and rat samples , complete cross-species validation is often lacking

    • Epitope conservation varies between species, affecting antibody performance in comparative studies

    • Limited validation in less common research species restricts evolutionary and comparative studies

  • Technical application constraints:

    • Small size of MBP (15-22 kDa) makes western blot transfer and detection challenging

    • Membranous nature of MBP requires specialized extraction procedures for optimal yields

    • Live cell applications remain difficult due to MBP's intracellular localization and membrane association

  • Standardization issues:

    • Variability between antibody lots affects reproducibility

    • Lack of standardized validation methods makes comparing antibody performance difficult

    • Quantitative applications require careful calibration and may not be comparable between different antibody clones

Understanding these limitations is essential for experimental design, data interpretation, and troubleshooting. Researchers should implement appropriate controls and validation steps to address these challenges and ensure reliable, reproducible results when working with MBP antibodies.

What future developments can we expect in MBP antibody technology?

The future of MBP antibody technology is likely to see significant advancements that address current limitations while expanding research capabilities:

  • Enhanced antibody engineering:

    • Isoform-specific antibodies: Development of monoclonal antibodies with enhanced specificity for individual MBP isoforms, enabling more precise developmental and pathological studies

    • PTM-sensitive detection: Advanced antibodies capable of distinguishing between different post-translational modification states of MBP

    • Smaller antibody formats: Continued development of single-domain antibodies (nanobodies) and aptamers for improved tissue penetration and live imaging applications

  • Advanced imaging applications:

    • Super-resolution compatible probes: Next-generation fluorophore conjugates optimized for techniques like STORM, PALM, and STED microscopy

    • Multiplexing capabilities: Development of antibodies compatible with highly multiplexed imaging techniques like CODEX and Imaging Mass Cytometry

    • In vivo imaging probes: Blood-brain barrier penetrant antibody fragments for non-invasive visualization of myelin in living organisms

  • Single-cell analysis integration:

    • Compatibility with CyTOF: Metal-conjugated anti-MBP antibodies for high-dimensional single-cell proteomics

    • Spatial transcriptomics integration: Combined detection of MBP protein and mRNA in tissue contexts

    • Multi-omics approaches: Antibodies enabling simultaneous analysis of MBP expression with other cellular parameters

  • Therapeutic and diagnostic applications:

    • Improved prognostic tools: Standardized antibody-based assays for measuring anti-MBP autoantibodies as biomarkers in multiple sclerosis

    • Therapeutic antibody development: Engineered antibodies targeting pathogenic MBP epitopes or blocking harmful autoantibody binding

    • Remyelination monitoring: Non-invasive imaging with specialized antibodies to track myelin repair in response to treatments

  • Technical refinements:

    • Increased standardization: Development of reference standards and validation protocols for anti-MBP antibodies

    • Improved recombinant antibodies: Shift toward recombinant production for enhanced reproducibility and reduced batch variation

    • Cross-species validation: Comprehensive validation across diverse species to enable evolutionary and comparative studies

These anticipated developments will significantly enhance our ability to study myelin biology in both normal and pathological contexts, potentially leading to improved diagnostics and therapeutics for demyelinating disorders.

How should researchers choose the optimal MBP antibody for their specific research questions?

Selecting the optimal MBP antibody requires systematic evaluation of multiple factors aligned with specific research objectives. Here's a comprehensive decision framework:

  • Research question alignment:

    • Target specificity needs: Determine whether you need pan-MBP detection or isoform-specific recognition

    • Application requirements: Different applications (WB, IHC, IP, ELISA, flow cytometry) may require specialized antibodies

    • Species considerations: Ensure the antibody recognizes MBP in your species of interest (human, mouse, rat, etc.)

    • PTM sensitivity: Consider whether detection of post-translationally modified MBP is relevant to your research question

  • Validation evidence assessment:

    • Published validation: Review literature for independent validation in applications similar to yours

    • Manufacturer data: Evaluate the comprehensiveness of validation data provided by suppliers

    • Citation history: Consider antibodies with established track records in peer-reviewed publications

    • Reproducibility reports: Check for any reported issues with batch-to-batch variability

  • Technical specifications evaluation:

    • Antibody format: Choose between monoclonal (higher specificity) and polyclonal (potentially higher sensitivity) based on your needs

    • Isotype considerations: Different isotypes (IgG, IgM) have different properties for certain applications

    • Conjugation options: Consider pre-conjugated antibodies for direct detection or unconjugated for flexibility

    • Clone identification: For monoclonals, specific clones like 932908, 2A1, or 3D7 have documented performance characteristics

  • Protocol compatibility:

    • Buffer requirements: Ensure compatibility with your experimental buffers (e.g., TBS-T for western blotting)

    • Fixation sensitivity: For histological applications, verify compatibility with your fixation method

    • Concentration recommendations: Review suggested working concentrations (0.1-1 μg/mL for WB; up to 10 μg/mL for IHC)

    • Incubation conditions: Consider optimal temperature and duration requirements

  • Practical considerations:

    • Cost-benefit analysis: Balance antibody cost against quality and validation evidence

    • Availability: Consider consistent supply for long-term projects

    • Technical support: Evaluate manufacturer support for troubleshooting

    • Reproducibility planning: For critical experiments, test multiple antibodies in parallel

Product Science Overview

Introduction

Maltose-binding protein (MBP) is a well-characterized protein from Escherichia coli that plays a crucial role in the uptake and catabolism of maltodextrins. It is part of the maltose/maltodextrin system, which is a complex regulatory and transport system involving multiple proteins and protein complexes . MBP has been extensively studied for its structural, functional, and biotechnological applications.

Structure and Function

MBP is a monomeric protein with an approximate molecular mass of 42.5 kilodaltons . It is encoded by the malE gene of E. coli, which produces a precursor polypeptide consisting of 396 amino acid residues. Upon cleavage of the NH2-terminal extension (26 residues), the mature MBP (370 residues) is formed . The protein is divided into two distinct globular domains connected by three short polypeptide segments. These domains are separated by a deep groove that contains the maltose/maltodextrin binding site .

The binding of maltose induces a significant conformational change in MBP, closing the groove by a rigid motion of the two domains around the linking polypeptide hinge . Both precursor and mature forms of MBP are functional for maltose binding, although the NH2-terminal extension decreases the folding rate of the precursor form relative to its mature form .

Localization and Export

MBP is exported into the periplasmic space of E. coli . The NH2-terminal extension, also known as the signal peptide, has two primary roles: it slows down the folding of the newly synthesized polypeptide and directs it to the membrane and SecYEG translocon . Once folded, the precursor can no longer enter the translocation pathway . Mutations in the signal peptide can block export, highlighting its importance in the protein’s localization .

Control of Expression

The malE gene, coding for MBP, is part of the Mal regulon of E. coli, which consists of ten genes involved in the efficient uptake and utilization of maltose and maltodextrins . These genes are clustered in the malB region of E. coli and organized into two divergent operons: malE-malF-malG and malK-lamB .

Biotechnological Applications

MBP is widely used as a fusion tag to enhance the solubility and expression of recombinant proteins in bacterial cells . Fusion of a target protein to MBP allows for a simple capture affinity step on amylose resin, resulting in a protein that is often 70-90% pure . Additionally, MBP fusion proteins exhibit increased protein production in mammalian cell lines and reduced cell death upon transient transfection . This makes MBP a versatile expression tag for protein production in various expression systems .

Mouse Antibody

Mouse antibodies are immunoglobulins produced by mice in response to antigens. They are commonly used in research and therapeutic applications due to their specificity and effectiveness . However, the use of mouse antibodies in humans can lead to the development of human anti-mouse antibodies (HAMA), which can reduce the effectiveness of the treatment and cause allergic reactions . Despite these challenges, mouse antibodies remain valuable tools in preclinical studies and therapeutic research .

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.