MYD88 antibodies are immunoreagents designed to bind specifically to the MYD88 protein, an adapter molecule critical for Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) signaling . These antibodies are used to:
Detect MYD88 expression in cells and tissues via Western blot (WB), immunohistochemistry (IHC), or immunofluorescence (IF) .
Study MYD88-dependent signaling pathways, including NF-κB and JAK/STAT activation .
Identify MYD88 L265P and other mutations in lymphoma diagnostics .
MYD88 antibodies have elucidated the protein’s function in TLR/IL-1R pathways:
MYD88 recruits IRAK kinases to activate NF-κB and proinflammatory cytokines .
In MyD88−/− mice, impaired IFN-γ production and CD4+ T cell recruitment exacerbate viral encephalomyelitis .
MYD88 L265P is a gain-of-function mutation detected in >90% of Waldenström’s macroglobulinemia (WM) cases and ~20% of diffuse large B-cell lymphomas (DLBCL) .
Antibodies help identify mutant MYD88 in cerebrospinal fluid for diagnosing Bing-Neel syndrome .
MYD88 signaling in B cells and dendritic cells (DCs) drives autoantibody production in Lyn−/− mice, a lupus model .
Inhibiting MYD88 in DCs reduces IL-12 and IL-6 expression, attenuating inflammation .
Liquid Biopsies: MYD88 L265P detection in blood or CSF aids WM diagnosis and reduces invasive biopsies .
Prognostic Marker: MYD88-mutated DLBCL correlates with activated B-cell (ABC) subtype and poorer survival .
BTK inhibitors (e.g., ibrutinib) show efficacy in MYD88 L265P-positive WM by disrupting BCR-MYD88 supercomplexes .
Preclinical studies suggest blocking MYD88 improves lupus outcomes by halting autoantibody production .
Specificity: Some antibodies cross-react with non-target proteins, necessitating KO validation .
Mutation Heterogeneity: Non-L265P MYD88 mutations (e.g., S219C, M232F) occur in DLBCL but lack prognostic significance .
MyD88 is a key adaptor molecule containing a cytoplasmic Toll/interleukin-1 receptor (TIR) domain that plays a pivotal role in innate immune responses . It functions by recruiting IRAK to the IL-1 receptor complex in response to IL-1 stimulation, leading to NF-κB activation and inflammatory gene expression . MyD88 was originally identified as a myeloid differentiation primary response gene rapidly induced during IL-6 stimulated differentiation of myeloleukemic cells into macrophages .
MyD88 is essential for:
Mediating TLR/IL-1 receptor signaling responses
Regulating IL-18 receptor signaling
Supporting antibody responses with specific IgG subclass profiles
Enabling efficient cross-presentation of viral antigens
Selection criteria should include:
For example, the polyclonal antibody against residues 279-296 of human MyD88 recognizes both human and mouse antigens , while some monoclonal antibodies may have more restricted specificity.
MyD88 has distinctive structural features that influence antibody detection:
Contains an amino-terminal death domain separated from a carboxyl-terminal TIR domain
Primarily localized in the cytoplasm, as confirmed by immunofluorescence studies
Expressed in diverse tissues including heart, kidney, liver, and various immune cells (monocytes, T cells, B cells, NK cells, dendritic cells)
For effective Western blot analysis:
For validation, include recombinant human MyD88 (1 ng) as a reference control alongside cellular lysates . Western blot analysis can detect endogenous levels of MyD88 protein, with specific bands appearing at approximately 35 kDa .
For successful cellular localization studies:
Fixation: Use paraformaldehyde fixation (typically 4%)
Permeabilization: Saponin is effective for intracellular access
Antibody concentration: 3-15 μg/mL depending on the antibody
Incubation time: 3 hours at room temperature is often sufficient
Counterstaining: DAPI for nuclear visualization
MyD88 appears predominantly in the cytoplasm as demonstrated in Raji human Burkitt's lymphoma cells, consistent with its role as a cytoplasmic adaptor protein .
For intracellular flow cytometry:
Cell fixation: Paraformaldehyde (typically 2-4%)
Permeabilization: Saponin buffer (0.1-0.5%)
Blocking: Normal serum corresponding to secondary antibody host
Primary antibody: Titrate for optimal signal-to-noise ratio
Secondary detection: Fluorophore-conjugated secondary antibodies like Allophycocyanin-conjugated Anti-Goat IgG
Controls: Include isotype control antibody for background assessment
Flow cytometric analysis has been successfully demonstrated in Raji cells, providing quantitative assessment of MyD88 expression at the single-cell level .
To study MyD88's function in cross-presentation:
Experimental design:
Key findings from previous research:
MyD88 is strictly required for cross-priming when virally infected cells are used as an antigen source
Direct infection of DCs can activate antigen-specific CD8+ T cells in a MyD88-independent manner
The effects of innate immunity stimulation via the MyD88 pathway control cross-presentation efficiency, but not direct presentation
This approach reveals how MyD88 signaling impacts different antigen presentation pathways with implications for viral immunity and vaccine design.
To examine cell-intrinsic roles of MyD88 in antibody responses:
These approaches highlight how MyD88 signaling in specific immune cell subsets influences the quality and magnitude of antibody responses.
To investigate MyD88's impact on antibody isotype profiles:
Experimental approach:
Notable findings:
MyD88 is necessary for the production of specific IgG2b, IgG2c, and IgG3 antibodies against various AAV serotypes
TLR9 is not critically required for total anti-AAV IgG responses but maintains a high IgG2c/IgG1 ratio typical of Th1-supported antibody responses
MyD88 is essential for developing neutralizing antibody responses against rAAV1
This research demonstrates how specific innate signaling pathways shape adaptive antibody responses, with implications for vaccine design and immunotherapy.
To ensure antibody specificity:
Validation approaches:
Use MyD88-knockout cells or tissues as negative controls
Test cross-reactivity with recombinant MyD88 protein
Compare results from multiple antibodies targeting different epitopes
Perform peptide competition assays
Validate across multiple applications
Cross-reactivity considerations:
For pathway discrimination studies:
Key experimental controls:
Include MyD88-deficient cells alongside wild-type
Employ selective TLR/IL-1R pathway inhibitors
Test multiple TLR ligands that signal through distinct pathways
Monitor downstream signaling events (NF-κB activation, cytokine production)
Analytical approaches:
Assess both early (minutes to hours) and late (hours to days) responses
Examine multiple readouts, including signaling intermediate phosphorylation
Consider compensatory mechanisms in knockout systems
Combine in vitro and in vivo analyses for comprehensive understanding
When encountering inconsistent results:
Potential causes:
Epitope accessibility differences in various applications
Post-translational modifications affecting antibody recognition
Sample preparation variations (fixation, extraction methods)
Antibody lot-to-lot variability
Cell type-specific expression patterns or isoforms
Resolution strategies:
Test multiple antibodies targeting different regions of MyD88
Validate with complementary techniques (PCR, mass spectrometry)
Standardize experimental conditions rigorously
Consider cell type and context-specific effects on MyD88 expression
Advanced approaches to study MyD88 complex formation:
Temporal analysis techniques:
Time-course immunoprecipitation with MyD88 antibodies following stimulation
Proximity ligation assays to visualize protein-protein interactions
FRET/BRET analysis with labeled antibodies or fusion proteins
Super-resolution microscopy to visualize signalosome assembly
Co-localization studies:
Multi-color immunofluorescence with antibodies against MyD88 and associated proteins
Live-cell imaging with fluorescently tagged antibody fragments
Correlation with functional outcomes (NF-κB nuclear translocation, cytokine production)
Innovative research applications include:
Biomarker development:
Assessment of MyD88 expression/activation as a predictor of immunotherapy response
Monitoring pathway activation in patient samples during treatment
Correlation with clinical outcomes in inflammatory and infectious diseases
Therapeutic targeting:
For investigating novel MyD88 roles:
Experimental strategies:
Subcellular fractionation followed by immunoblotting to identify unexpected locations
Immunoprecipitation combined with mass spectrometry to identify novel interaction partners
ChIP assays to explore potential nuclear functions
CRISPR-edited cells expressing tagged MyD88 for comprehensive interaction mapping
Context-specific analyses:
Cell type-specific expression and function studies
Investigation of tissue-specific roles beyond immune cells
Developmental timing of MyD88 expression and function
Myeloid Differentiation Primary Response 88 (MYD88) is a crucial adaptor protein in the innate immune system. It was originally discovered in the laboratory of Dan A. Liebermann as a primary response gene involved in myeloid differentiation . The MYD88 gene encodes a protein that plays a pivotal role in the signaling pathways of the immune system, particularly in the activation of immune cells through Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs) .
Function and Mechanism:
Clinical Significance:
Mouse anti-Human antibodies are secondary antibodies generated by immunizing mice with human antigens. These antibodies are widely used in research and diagnostic applications due to their specificity and versatility .
Production and Purification:
Applications:
Challenges: