MYD88 Human

Myeloid Differentiation Primary Response 88 Human Recombinant
Shipped with Ice Packs
In Stock

Description

MYD88 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 345 amino acids (1-309 a.a) and having a molecular mass of 38.7kDa.
MYD88 is fused to a 36 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
The protein known as Myeloid differentiation primary response gene 88 (MYD88) plays a crucial role in the immune system's response to infections. It acts as an adaptor protein, facilitating the transmission of signals from receptors like Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs) that recognize pathogens. This signaling cascade ultimately activates NF-kappa-B, a protein complex that regulates immune and inflammatory responses by controlling the expression of genes involved in inflammation and immune cell activation. MYD88 is essential for the production of cytokines, molecules that mediate communication between cells and are vital for coordinating immune responses. Defects in the MYD88 gene can lead to increased vulnerability to bacterial infections, highlighting its critical role in immune defense.
Description
This product consists of the human MYD88 protein, recombinantly produced in E. coli. It is engineered with a His-tag on the N-terminus to facilitate purification and is provided in a solution free from glycosylation. The protein encompasses amino acids 1 to 309 of the MYD88 sequence, with an additional 36 amino acids from the His-tag, resulting in a molecular weight of 38.7 kDa. The purity of this recombinant MYD88 protein exceeds 90%, as verified by SDS-PAGE analysis.
Physical Appearance
A clear and colorless solution, sterile-filtered.
Formulation
The MYD88 protein is supplied in a solution at a concentration of 1 mg/ml. The solution is buffered with 20mM Tris-HCl at a pH of 8.0 and contains 0.4M urea and 10% glycerol.
Stability
For optimal storage, the product should be kept refrigerated at 4°C if it will be used within 2-4 weeks. For longer-term storage, freezing at -20°C is recommended. To enhance stability during prolonged storage, consider adding a carrier protein like HSA or BSA (0.1%). Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the MYD88 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Myeloid differentiation primary response protein MyD88, MYD88, MYD88D.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMRPD RAEAPGPPAM AAGGPGAGSA APVSSTSSLP LAALNMRVRR RLSLFLNVRT QVAADWTALA EEMDFEYLEI RQLETQADPT GRLLDAWQGR PGASVGRLLE LLTKLGRDDV LLELGPSIEE DCQKYILKQQ QEEAEKPLQV AAVDSSVPRT AELAGITTLD DPLGHMPERF DAFICYCPSD IQFVQEMIRQ LEQTNYRLKL CVSDRDVLPG TCVWSIASEL IEKRCRRMVV VVSDDYLQSK ECDFQTKFAL SLSPGAHQKR LIPIKYKAMK KEFPSILRFI TVCDYTNPCT KSWFWTRLAK ALSLP.

Q&A

What is MYD88 and what is its function in human immune signaling?

MYD88 (Myeloid differentiation primary response 88) is an adapter protein involved in innate immune signaling pathways. Originally discovered as a myeloid differentiation primary response gene, this 296-amino acid protein (Met1-Pro296) acts as a critical connector between cell surface receptors and intracellular signaling components . In humans, MYD88 functions primarily as an adapter protein that links Toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R) family members to downstream signaling machinery, ultimately activating transcription factors like NF-κB that drive inflammatory gene expression .

Biochemically, MYD88 contains a C-terminal Toll/IL-1R (TIR) domain that interacts with the cytoplasmic portions of TLRs and IL-1R family members, and an N-terminal death domain (DD) that facilitates recruitment of IL-1R-associated kinases (IRAKs). When studying MYD88 expression, researchers typically observe cytoplasmic localization, as demonstrated by immunocytochemistry in various cell lines including Raji Burkitt's lymphoma cells, where specific staining localizes to the cytoplasm .

MYD88 is widely expressed in immune and non-immune tissues, including heart, kidney, liver, and in multiple immune cell types such as monocytes, T cells, B cells, NK cells, and dendritic cells . This broad expression pattern reflects its fundamental role in host defense and inflammatory responses.

How does MYD88 participate in TLR signaling cascades?

MYD88 serves as the primary adaptor protein for most Toll-like receptors (all except TLR3) and plays a central role in transducing signals from pattern recognition to inflammatory gene activation. The signaling cascade follows a well-characterized sequence:

  • Ligand recognition by TLRs causes receptor dimerization and conformational changes

  • MYD88 is recruited to activated TLRs through TIR-TIR domain interactions

  • MYD88 oligomerizes to form the "Myddosome" signaling complex

  • The Myddosome recruits and activates IRAK family kinases (primarily IRAK4 and IRAK1/2)

  • Activated IRAKs recruit TRAF6 (TNF receptor-associated factor 6)

  • TRAF6 facilitates activation of TAK1 kinase and the IKK complex

  • IKK phosphorylates IκB, leading to its degradation

  • Released NF-κB translocates to the nucleus to induce inflammatory gene expression

Importantly, MYD88 participates differently in signaling from cell surface TLRs (TLR1, TLR2, TLR4, TLR5, TLR6) versus endosomal TLRs (TLR7, TLR8, TLR9). Cell surface TLRs recognize primarily microbial membrane components, while endosomal TLRs detect nucleic acids . This compartmentalization provides spatial regulation of immune responses to different classes of pathogens.

Experimental studies of TLR-MYD88 signaling typically employ detection methods such as Western blotting with specific antibodies (e.g., Anti-Human MYD88 at 0.5 μg/mL concentration) to visualize the approximately 35 kDa MYD88 protein in cell lysates , complemented by phosphorylation analysis of downstream signaling components.

What experimental models are most suitable for studying MYD88 function?

Multiple experimental systems have been developed to study MYD88 function, each with specific advantages for addressing different research questions:

  • Cell line models:

    • Human cell lines including Raji (Burkitt's lymphoma), Jurkat (T cell leukemia), and HT-29 (colon adenocarcinoma) express MYD88 and are useful for signaling studies

    • DLBCL cell lines such as OCI-LY3 (homozygous for MYD88 L265P mutation) and OCI-LY19 (wild-type MYD88) provide models for studying oncogenic MYD88 mutations

  • Primary cell systems:

    • CD19+-selected bone marrow cells from healthy donors or patients

    • Peripheral blood mononuclear cells for studying MYD88 function in multiple immune cell types

    • Techniques include flow cytometry with fixation/permeabilization to detect intracellular MYD88

  • Mouse models:

    • Myd88−/− mice provide a complete knockout system

    • Conditional knockout mice (tissue-specific or inducible) allow temporal and spatial control

    • Disease models such as the azoxymethane (AOM)/dextran sodium sulfate (DSS) colitis-associated cancer model reveal context-specific functions

    • Receptor-specific knockout models (Tlr2−/−, Il1r−/−, Il18−/−) help dissect specific upstream pathways

  • Molecular tools:

    • Expression vectors for wild-type or mutant MYD88

    • siRNA/shRNA for transient knockdown

    • CRISPR/Cas9 for precise gene editing

For detection and analysis, researchers commonly employ antibody-based methods including Western blotting, which can detect MYD88 as a specific band at approximately 35 kDa in various cell lines , immunofluorescence at concentrations of 3-15 μg/mL , and flow cytometry with appropriate fixation and permeabilization protocols to access intracellular MYD88 .

How do MYD88-dependent and MYD88-independent pathways differ mechanistically?

MYD88-dependent and MYD88-independent pathways represent distinct arms of pattern recognition receptor signaling with different kinetics, downstream components, and functional outcomes:

MYD88-dependent pathway:

  • Utilized by all TLRs except TLR3

  • Results in rapid NF-κB activation (typically within 10-30 minutes)

  • Employs IRAK kinases (IRAK4, IRAK1, IRAK2) and TRAF6

  • Strongly induces pro-inflammatory cytokines (TNF-α, IL-6, IL-1β, IL-12)

  • Essential for early inflammatory responses to bacterial and fungal pathogens

MYD88-independent pathway:

  • Primarily mediated by TRIF (TIR domain-containing adapter-inducing IFN-β)

  • Activated by TLR3 (exclusively) and TLR4 (in addition to MYD88)

  • Leads to activation of IRF3 and delayed NF-κB activation

  • Induces type I interferons and interferon-stimulated genes

  • Critical for antiviral responses

Experimental discrimination between these pathways typically involves comparative studies in wild-type versus Myd88−/− systems. Interestingly, research has shown that these pathways can have distinct biological consequences even in the same disease context. For example, Tlr4-deficient mice (affected in both pathways) are similar to Myd88-deficient mice in colitis susceptibility but differ dramatically in cancer susceptibility, with Tlr4-deficient mice being resistant to colitis-associated cancer while Myd88-deficient mice show increased tumor development .

This differential outcome reveals complex interactions between signaling pathways that regulate inflammation versus tissue homeostasis and carcinogenesis. Methodologically, distinguishing these pathways requires careful experimental design, including pathway-specific stimulation, time-course studies, and analysis of distinct downstream targets.

What is the relationship between MYD88 and IL-1R/IL-18R signaling pathways?

Beyond its role in TLR signaling, MYD88 is essential for signal transduction downstream of IL-1 family receptors, including IL-1R and IL-18R. This functional overlap creates important biological redundancies and specificities in inflammatory responses:

  • Shared signaling architecture:

    • IL-1R and IL-18R contain cytoplasmic TIR domains similar to TLRs

    • Both receptors recruit MYD88 following ligand binding

    • Downstream signaling proceeds through IRAK kinases and TRAF6 to NF-κB activation

  • Distinct biological functions:

    • IL-1 signaling primarily induces broad inflammatory responses

    • IL-18 signaling potently stimulates IFN-γ production, particularly in conjunction with IL-12

Research using gene knockout models has revealed important insights into these relationships. In the context of colitis and colitis-associated cancer, Il1r-deficient mice show no differential susceptibility compared to wild-type mice, suggesting IL-1R signaling is not critical in this context . In contrast, Il18−/− and Il18r1−/− mice demonstrate increased susceptibility to colitis and polyp formation upon AOM/DSS treatment, closely resembling the phenotype of Myd88−/− mice .

This important finding indicates that the inability to signal through IL-18R is a significant contributor to the increased cancer susceptibility observed in Myd88-deficient mice. Methodologically, these relationships are best studied through comparative analysis of receptor-specific knockout models, signaling assays with selective receptor agonists, and functional readouts such as cytokine production profiles.

How does MYD88 signaling affect tissue homeostasis versus inflammation?

MYD88 signaling exhibits remarkable context-dependency in regulating the balance between tissue homeostasis and inflammation. This dual functionality is particularly evident in epithelial tissues exposed to microbiota, such as the intestine:

  • In tissue homeostasis:

    • MYD88 signaling promotes epithelial regeneration and wound healing

    • It facilitates recognition of commensal microbiota without triggering excessive inflammation

    • Maintains barrier integrity through regulation of antimicrobial peptides and mucin production

  • In inflammatory contexts:

    • MYD88 orchestrates inflammatory cytokine and chemokine production

    • Recruits and activates innate immune cells

    • Shapes subsequent adaptive immune responses

The critical role of MYD88 in maintaining this balance is dramatically illustrated in colitis-associated cancer models. While MYD88 signaling promotes carcinogenesis in several cancer models, it actually has a protective role in azoxymethane (AOM)/dextran sodium sulfate (DSS) colitis-associated cancer . Myd88−/− mice demonstrate an inability to heal ulcers generated upon injury, creating an altered inflammatory environment that leads to dysregulation of pathways controlling cell proliferation, apoptosis, and DNA repair .

This dysregulation results in increased adenoma formation and progression to infiltrating adenocarcinomas with frequent clonal mutations in the β-catenin gene . The complex role of MYD88 is further evidenced by comparing various knockout models. For instance, Tlr4-deficient mice are susceptible to colitis (like Myd88−/− mice) but resistant to cancer development, while Il18−/− and Il18r1−/− mice more closely phenocopy Myd88−/− mice .

These findings reveal "a previously unknown level of complexity surrounding MyD88 activities downstream of different receptors that impact tissue homeostasis and carcinogenesis" . When studying these relationships, researchers should consider tissue-specific knockout models, acute versus chronic inflammatory settings, and comprehensive analysis of both inflammatory mediators and tissue regeneration markers.

What is the significance of the MYD88 L265P mutation in B-cell malignancies?

The MYD88 L265P mutation represents one of the most clinically significant alterations in the MYD88 gene, serving as both a diagnostic marker and potential therapeutic target in certain B-cell malignancies:

  • Prevalence and diagnostic value:

    • Present in approximately 90% of Waldenström macroglobulinemia (WM) patients, based on CD19+-selected bone marrow samples

    • Also detected in a subset of IgM monoclonal gammopathy of undetermined significance (MGUS) cases

    • Less frequently found in other B-cell malignancies, including certain diffuse large B-cell lymphoma subtypes

  • Functional consequences:

    • The L265P mutation occurs in the TIR domain of MYD88

    • Creates constitutive MYD88 signaling independent of receptor engagement

    • Activates downstream pathways including NF-κB, JAK-STAT, and BTK signaling

    • Promotes B-cell survival, proliferation, and malignant transformation

  • Detection methodologies:

    • Conventional allele-specific PCR (AS-PCR) can detect MYD88 L265P with sensitivity to 0.1% mutation burden

    • SYBR green-based real-time AS-PCR quantifies MYD88 L265P allele burden with sensitivity to 0.08%

    • PCR assays utilize primers where AS primers are located in exon 5 while the common forward primer is in intron 4

    • Enhanced specificity is achieved with an additional mismatch (T>A) at the third position from the 3' end of AS primers

    • The MYD88 L265P mutant-specific amplicon has a characteristic melting temperature of 84°C

  • Clinical applications:

    • Aids in differential diagnosis between WM and other lymphoproliferative disorders

    • Can be used to monitor disease progression and treatment response

    • Quantitative assessment using real-time PCR shows high correlation (coefficient of 0.998) and precision

    • Potential biomarker for targeted therapy selection

This mutation exemplifies how a single amino acid change in a signaling adaptor can drive oncogenesis, highlighting the critical nature of regulated innate immune signaling pathways. For researchers studying this mutation, precision studies have demonstrated complete reproducibility among replicates both within and between runs, making it a reliable biomarker for both research and clinical applications .

What roles does MYD88 play in inflammatory bowel diseases and colitis-associated cancer?

MYD88 functions as a critical regulator at the intersection of inflammation, tissue repair, and carcinogenesis in the gastrointestinal tract, with particularly nuanced roles in inflammatory bowel diseases (IBDs) and associated malignancies:

  • Intestinal homeostasis and IBD:

    • Intestinal microbiota represent key pathogenic triggers in IBD development

    • TLRs function as primary sensors of microbial colonization, with MYD88 transducing signals from most TLRs

    • MYD88 signaling maintains epithelial barrier integrity under normal conditions

    • In IBD contexts, MYD88 mediates both protective and pathogenic inflammatory responses

  • Colitis-associated cancer (CAC):

    • In the azoxymethane (AOM)/dextran sodium sulfate (DSS) model of CAC, MYD88 signaling unexpectedly shows a protective role, contrary to its pro-carcinogenic effect in other cancer models

    • Myd88−/− mice exhibit impaired ulcer healing after injury, creating a permissive environment for tumor development

    • This results in early alterations in expression of genes regulating inflammation, cell proliferation, apoptosis, and DNA repair

    • The consequence is dramatic increases in adenoma formation and progression to infiltrating adenocarcinomas with frequent β-catenin mutations

  • Receptor-specific contributions:

    • Comparative studies using various knockout models reveal receptor-specific effects:

    • Tlr4−/− mice: Similar susceptibility to colitis as Myd88−/− mice but resistant to CAC

    • Tlr2−/− and Il1r−/− mice: No differential susceptibility to colitis or CAC

    • Il18−/− and Il18r1−/− mice: Increased susceptibility to colitis and polyp formation, similar to Myd88−/− mice

  • Mechanistic implications:

    • These findings suggest the protective effect of MYD88 in CAC is significantly mediated through IL-18R signaling

    • They reveal complex interactions between inflammation, tissue repair, and carcinogenesis

    • The data highlights the importance of receptor-specific signaling outcomes rather than global MYD88 effects

These insights demonstrate how MYD88 signaling can have contextually different roles in disease processes, depending on the tissue microenvironment, upstream receptors engaged, and temporal context of inflammation. This complexity must be carefully considered when targeting innate immune pathways therapeutically in inflammatory and neoplastic conditions.

What are the most sensitive and specific methods for detecting MYD88 L265P mutations?

Detection of the MYD88 L265P mutation requires high sensitivity and specificity, particularly in clinical samples where mutant allele burden may be low. Several methodologies have been developed with distinct advantages for different research and clinical applications:

  • Conventional allele-specific PCR (AS-PCR):

    • Sensitivity: Detects MYD88 L265P to a dilution of 0.1%

    • Design: AS primers located in exon 5 with common forward primer in intron 4

    • Specificity enhancement: Additional mismatch (T>A) at third position from 3' end of AS primers

    • Validation: Complete precision (100% agreement) among replicate aliquots, both within and between runs

    • Advantages: Simple equipment requirements, rapid turnaround time

  • SYBR green-based real-time AS-PCR:

    • Sensitivity: Detects MYD88 L265P to a dilution of 0.08% with >2 cycle differences from wild-type

    • Quantification: Enables allele burden quantification with high correlation coefficient (0.998)

    • Melting characteristics: MYD88 L265P mutant-specific amplicon melts at 84°C

    • Specificity: Minor nonspecific amplification only at dilutions of 0.4% or lower (melting peak at 80°C)

    • Applications: Useful for monitoring disease progression and treatment response

  • Next-generation sequencing approaches:

    • Whole genome sequencing (WGS) and targeted sequencing panels

    • Advantages: Comprehensive mutation profiling beyond MYD88

    • Limitations: More costly, time-consuming, and require greater DNA input than PCR methods

  • Sanger sequencing:

    • Conventional approach that can detect MYD88 L265P in 90% of WM patients

    • Limitations: Lower sensitivity (typically 10-20% mutant allele frequency required)

    • Not ideal for minimal residual disease monitoring

For assay validation and quality control, researchers should:

  • Create standard curves using serial dilutions of DNA from MYD88 L265P homozygous cells (e.g., OCI-LY3) with wild-type MYD88 DNA (e.g., OCI-LY19)

  • Include appropriate positive and negative controls in each run

  • Perform melting curve analysis for real-time PCR assays

  • Conduct precision studies with replicate samples to establish reproducibility

These methodologies provide researchers with a toolkit for addressing different experimental questions, from high-throughput screening to sensitive quantification of mutation burden in response to therapeutic interventions.

What antibodies and detection systems are optimal for human MYD88 protein analysis?

Selecting appropriate antibodies and detection systems is critical for successful analysis of human MYD88 protein. Based on validated research applications, the following options have demonstrated reliability across different experimental platforms:

  • Primary antibodies for MYD88 detection:

    AntibodyTypeCatalog #ApplicationsWorking ConcentrationsTarget
    Goat Anti-Human MYD88Polyclonal, Affinity-purifiedAF2928WB, ICC, FCWB: 0.5 μg/mL, ICC: 15 μg/mLFull-length (Met1-Pro296)
    Rat Anti-Human MYD88Monoclonal (Clone 316603)MAB2928ICC3 μg/mLFull-length (Met1-Pro296)
  • Secondary detection systems:

    • For Western blotting: HRP-conjugated Anti-Goat IgG (HAF017)

    • For immunofluorescence with polyclonal antibody: NorthernLights™ 557-conjugated Anti-Goat IgG (NL001)

    • For immunofluorescence with monoclonal antibody: NorthernLights™ 557-conjugated Anti-Rat IgG (NL013)

    • For flow cytometry: Allophycocyanin-conjugated Anti-Goat IgG (F0108)

  • Experimental conditions for optimal results:

    • Western blotting: Use reducing conditions with Immunoblot Buffer Group 3

    • Expected band size: approximately 35 kDa

    • Immunocytochemistry: Fix cells by immersion, incubate with primary antibody for 3 hours at room temperature, counterstain with DAPI

    • Flow cytometry: Fix cells with paraformaldehyde and permeabilize with saponin to access intracellular MYD88

  • Quality control measures:

    • Include recombinant human MYD88 (1 ng) as positive control for Western blots

    • Use isotype control antibody (e.g., AB-108-C) for specificity assessment in flow cytometry

    • Test across multiple cell lines: Raji, Jurkat, HT-29 demonstrate consistent MYD88 expression

    • For immunofluorescence, specific staining should localize to the cytoplasm

  • Storage and handling:

    • Antibody shelf-life: 12 months from date of receipt when stored at -20 to -70°C

    • After reconstitution: 1 month at 2-8°C or 6 months at -20 to -70°C under sterile conditions

    • Avoid repeated freeze-thaw cycles

When selecting antibodies for specific applications, researchers should first validate antibody performance in their experimental system, as optimal dilutions may vary between laboratories and applications. General protocols for various applications are typically available from antibody manufacturers to facilitate optimization .

How can MYD88-dependent and MYD88-independent pathways be experimentally distinguished?

Discriminating between MYD88-dependent and MYD88-independent signaling pathways requires strategic experimental design and multiple complementary approaches. The following methodological strategies enable researchers to dissect these distinct signaling mechanisms:

  • Genetic approaches:

    • Compare wild-type versus Myd88−/− cells or tissues for pathway-specific responses

    • Utilize Trif−/− models to isolate MYD88-dependent signaling

    • Employ receptor-specific knockouts (e.g., Tlr3−/−, Tlr4−/−) to examine pathway contributions

    • Create combinatorial knockouts (e.g., Myd88−/−Trif−/−) to eliminate both pathways

  • Selective pathway activation:

    • TLR3 ligands (poly(I:C)) specifically activate MYD88-independent/TRIF-dependent signaling

    • TLR9 ligands (CpG DNA) activate exclusively MYD88-dependent pathways

    • TLR4 ligands (LPS) activate both pathways, allowing temporal discrimination

    • IL-1 or IL-18 stimulation activates MYD88-dependent pathways without engaging TRIF

  • Temporal analysis:

    • MYD88-dependent pathways: Rapid NF-κB activation (15-30 minutes post-stimulation)

    • TRIF-dependent pathways: Delayed NF-κB activation but rapid IRF3 activation

    • Time-course experiments with 15-30 minute intervals effectively distinguish these kinetics

  • Biochemical discrimination:

    • MYD88-dependent: Monitor IRAK1/4 phosphorylation and TRAF6 recruitment

    • TRIF-dependent: Assess TBK1 and IKKε phosphorylation and IRF3 activation

    • Western blotting with phospho-specific antibodies can track these distinct events

  • Transcriptional profiling:

    • MYD88-dependent: Primarily inflammatory cytokine gene expression (IL-6, TNF, IL-1β)

    • TRIF-dependent: Strong type I interferon and interferon-stimulated gene induction

    • qRT-PCR or RNA-seq can identify pathway-specific gene signatures

  • Functional readouts:

    • Compare cytokine production profiles (inflammatory cytokines versus interferons)

    • Assess antimicrobial responses against different classes of pathogens

    • Measure cell type-specific activation markers

A comprehensive approach often combines these methods, as illustrated in research on colitis and colitis-associated cancer. Investigators used multiple knockout models (Myd88−/−, Tlr4−/−, Tlr2−/−, Il1r−/−, Il18−/−, Il18r1−/−) to systematically dissect pathway contributions, revealing that while Tlr4−/− and Myd88−/− mice showed similar colitis susceptibility, they displayed opposite cancer phenotypes . This methodological approach demonstrated that different receptors signaling through the same adaptor can produce distinct biological outcomes.

What are the key considerations when using MYD88 knockout or knockdown approaches?

When employing MYD88 knockout or knockdown strategies, researchers must carefully consider several methodological aspects to ensure valid interpretation of results. These considerations span experimental design, biological complexity, and technical limitations:

  • Genetic background effects:

    • Different mouse strains can show variable phenotypes with the same MYD88 deficiency

    • Backcross knockout mice to appropriate background strain (minimum 10 generations)

    • Include proper wild-type littermate controls with identical genetic background

    • Consider using multiple independently derived knockout/knockdown lines

  • Developmental versus acute effects:

    • Constitutional Myd88 knockout may cause developmental compensation

    • Consider inducible systems (e.g., tamoxifen-inducible Cre-loxP) for temporal control

    • Compare acute knockdown (siRNA/shRNA) with stable knockout phenotypes

    • Evaluate developmental milestones in constitutional knockout models

  • Cell/tissue specificity:

    • MYD88 functions differently across cell types (epithelial cells versus immune cells)

    • Use tissue-specific promoters to drive Cre recombinase expression

    • Consider cell type-specific transduction with viral vectors

    • Employ bone marrow chimeras to distinguish hematopoietic versus non-hematopoietic effects

  • Functional redundancy and pathway complexity:

    • MYD88 mediates signaling from multiple receptors (TLRs, IL-1R, IL-18R)

    • Phenotypes may result from combined deficiency in multiple pathways

    • Compare receptor-specific knockouts to isolate individual contributions

    • Research has shown complex phenotypic differences between Myd88−/− and receptor-specific knockouts (e.g., Tlr4−/−, Il18r1−/−) in disease models

  • Validation approaches:

    • Confirm knockout/knockdown at DNA, RNA, and protein levels

    • For MYD88 protein detection, use validated antibodies like AF2928 at appropriate concentrations (0.5 μg/mL for Western blot)

    • Assess functional consequences through pathway-specific stimulation

    • Perform rescue experiments with wild-type MYD88 expression

  • Context-dependent interpretations:

    • MYD88's role varies by disease context (protective in colitis-associated cancer but promoting other cancers)

    • Acute versus chronic inflammation may reveal different MYD88 functions

    • Environmental factors (e.g., microbiota) strongly influence MYD88-dependent phenotypes

    • Consider housing conditions, microbiome status, and pathogen exposure

The complexity of MYD88 biology is exemplified in colitis-associated cancer research, where Myd88−/− mice show an inability to heal ulcers after injury, creating an altered inflammatory environment that leads to increased cancer susceptibility. Comparison with receptor-specific knockouts revealed that IL-18R signaling, rather than TLR or IL-1R pathways, most closely accounts for this phenotype . Such systematic approaches are essential for untangling the multifaceted roles of MYD88 in different physiological and pathological contexts.

What are the most significant challenges in MYD88 research today?

MYD88 research faces several critical challenges that limit both our fundamental understanding and therapeutic applications. Key among these are:

  • Pathway specificity discrimination:

    • MYD88 signaling occurs downstream of multiple receptors (TLRs, IL-1R, IL-18R)

    • Experimental systems often cannot distinguish receptor-specific effects

    • Context-dependent outcomes make generalizations difficult

    • Research shows unexpected complexity, as exemplified by differential cancer susceptibility in Myd88−/− versus Tlr4−/− mice despite similar colitis phenotypes

  • Cell type-specific functions:

    • MYD88 functions differently in epithelial cells, myeloid cells, and lymphocytes

    • Effects on homeostasis versus inflammation vary by cell type

    • Technical challenges in achieving cell type-specific modulation

    • Interpreting mixed cell population data in complex tissues

  • Temporal regulation:

    • Acute versus chronic MYD88 signaling produces different outcomes

    • Dynamic regulation during disease progression remains poorly understood

    • Technical limitations in capturing signaling dynamics in vivo

    • Challenges in developing systems for temporal control of signaling

  • Therapeutic targeting:

    • Broad involvement in immunity makes selective targeting difficult

    • Identifying context-specific inhibition strategies

    • Developing approaches that target oncogenic MYD88 mutations (e.g., L265P) without compromising normal function

    • Balancing antimicrobial defense with inflammatory pathology

  • Methodological limitations:

    • Sensitivity concerns in detecting low-abundance MYD88 L265P mutations

    • Developing standardized quantification methods for mutation burden

    • Technical challenges in monitoring pathway activation in clinical samples

    • Translating research findings to practical diagnostic applications

Addressing these challenges requires innovative experimental approaches and conceptual frameworks that account for the multifaceted roles of MYD88 in health and disease.

What emerging technologies are advancing human MYD88 research?

Recent technological advances are transforming our ability to investigate MYD88 biology and its role in human disease:

  • Enhanced mutation detection methods:

    • Highly sensitive allele-specific PCR assays detect MYD88 L265P mutation to 0.1% dilution

    • Real-time quantitative approaches measure mutation burden with sensitivity to 0.08%

    • Digital PCR platforms enable absolute quantification of mutant alleles

    • These advances facilitate minimal residual disease monitoring and earlier detection

  • Single-cell technologies:

    • Single-cell RNA sequencing reveals cell type-specific MYD88-dependent gene expression

    • Mass cytometry (CyTOF) enables high-dimensional analysis of signaling states

    • Spatial transcriptomics preserves tissue context while assessing signaling outcomes

    • These approaches help resolve heterogeneity in complex tissues and tumors

  • Sophisticated genetic models:

    • Conditional and inducible knockout systems provide temporal and spatial control

    • CRISPR/Cas9 gene editing enables precise mutation introduction (e.g., L265P)

    • Humanized mouse models better recapitulate human disease biology

    • Organoid systems bridge the gap between cell lines and in vivo models

  • Structural and computational approaches:

    • Cryo-electron microscopy reveals molecular details of Myddosome assembly

    • Molecular dynamics simulations predict effects of mutations on protein-protein interactions

    • Systems biology approaches model complex signaling networks

    • Artificial intelligence algorithms identify novel patterns in large datasets

  • Therapeutic development platforms:

    • Selective MYD88 pathway inhibitors with improved pharmacokinetics

    • Strategies targeting specific MYD88 functions or mutations

    • Combination approaches addressing multiple nodes in signaling networks

    • Biomarker-guided patient selection for clinical trials

These technological advances enable researchers to address previously intractable questions about MYD88 biology, from molecular mechanisms to clinical applications. As these methods mature and become more accessible, we can expect accelerated progress in understanding MYD88's complex roles in immunity, inflammation, and cancer.

Product Science Overview

Structure and Function

MYD88 is a cytoplasmic adaptor protein that plays a central role in both the innate and adaptive immune responses. It functions as a vital signal transducer in the interleukin-1 (IL-1) and Toll-like receptor (TLR) signaling pathways . The protein is composed of 296 amino acids and has a modular structure with three main domains encoded by five exons .

Role in Immune Signaling

MYD88 acts as an adapter, connecting proteins that receive signals from outside the cell to those that relay signals inside the cell. In the context of innate immunity, MYD88 is pivotal for immune cell activation through TLRs, which are pattern recognition receptors (PRRs) that detect pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) .

Upon ligand binding, all TLRs, except TLR3, interact with MYD88. This interaction leads to the activation of transcription factors such as NF-κB, which is responsible for the expression of various inflammatory cytokines, chemokines, and adhesion molecules, triggering acute inflammation and stimulating adaptive immunity .

Recombinant MYD88

Recombinant human MYD88 protein is often used in research to study its role in immune signaling pathways. This recombinant protein is typically expressed in E. coli and can be fused to tags such as His-tag at the N-terminus for purification purposes . It has been shown to increase IL-8 transcription and is involved in the IL-18-mediated signaling pathway .

Therapeutic Potential

Given its central role in immune signaling, MYD88 has been recognized as a potential drug target. Dysregulation of MYD88 can lead to a wide range of inflammation-associated syndromes and diseases . Small molecule inhibitors targeting the TIR domain of MYD88 have shown promising therapeutic efficacy in experimental models by blocking TIR–TIR domain homo-dimerization, thereby attenuating MYD88-mediated inflammatory responses and enhancing antiviral type I IFN responses .

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.