MLLT4 (Myeloid/Lymphoid or Mixed-Lineage Leukemia; Translocated to, 4), also known as AF6 or Afadin, is a multi-domain protein involved in signaling and organization of cell junctions during embryogenesis. It has received significant attention due to its identification as a fusion partner of the ALL-1 gene in acute myeloid leukemias with t(6;11)(q27;q23) translocation .
MLLT4 has diverse cellular functions, including:
Maintenance of adherens junctions between radial glial cells (RGCs) in cortical development
Involvement in chromosomal translocations associated with leukemogenesis
Role as a transcriptional coregulator in hematopoietic development
Its complex role in both normal development and disease states makes it an important target for antibody-based detection and characterization in various research contexts .
Based on the technical specifications of available antibodies, MLLT4 antibodies are most widely utilized in the following applications:
| Application | Common Dilutions | Typical Sample Types | Detection Method |
|---|---|---|---|
| Western Blot (WB) | 1:500-1:1000 | Cell lysates | ~200-207 kDa band |
| Immunohistochemistry (IHC-P) | 1:50-1:200 | Paraffin-embedded tissues | Nuclear/cytoplasmic staining |
| Immunofluorescence (IF) | 1:50-1:200 | Fixed cells | Subcellular localization |
| ELISA | 1:2000-1:5000 | Protein samples | Quantitative detection |
Researchers should note that MLLT4 antibodies show reactivity with human, mouse, and rat samples, making them versatile for comparative studies across species . The choice of application should be guided by the specific research question, with Western blotting being particularly useful for characterizing fusion proteins in leukemia research .
Robust validation is essential to ensure experimental reliability when working with MLLT4 antibodies. A comprehensive validation protocol should include:
Positive and negative control samples: Utilize cell lines known to express MLLT4 (e.g., HeLa cells) as positive controls and MLLT4-knockout or knockdown samples as negative controls .
Multiple detection methods: Confirm antibody specificity using at least two independent techniques (e.g., Western blot combined with immunofluorescence).
Peptide competition assay: Pre-incubate the antibody with immunizing peptide to confirm binding specificity through signal abolishment .
Cross-reactivity assessment: Test against related proteins or in tissues where MLLT4 expression is well-characterized.
Molecular weight verification: For Western blotting, confirm detection at the expected molecular weight (~208 kDa for full-length protein; different for fusion proteins) .
Research by multiple groups indicates that epitope-affinity purified antibodies offer superior specificity compared to conventional purification methods when detecting MLLT4 and its fusion variants .
When studying MLLT4 in leukemia contexts, especially MLL-rearranged leukemias, researchers should consider these critical factors:
Epitope location: Select antibodies targeting the appropriate domain based on your research focus. For MLL-AF4 fusion protein detection, antibodies recognizing the C-terminal domain of MLLT4 are preferable .
Fusion protein specificity: Some antibodies may detect wild-type MLLT4 but fail to recognize fusion proteins or vice versa. Validation with positive controls expressing the specific fusion protein of interest is essential .
Sensitivity requirements: For minimal residual disease (MRD) monitoring, highly sensitive antibodies are required, often necessitating supplementation with molecular techniques like RT-PCR for MLL-AF4 fusion detection .
Cross-species reactivity: For translational research using mouse models of MLL leukemia, antibodies with confirmed reactivity in both human and mouse samples are advantageous .
Application compatibility: Ensure the antibody is validated for your intended application, particularly for flow cytometry applications in leukemia phenotyping .
The literature suggests combining antibody detection with molecular methods for comprehensive characterization of MLLT4 fusion proteins in leukemia research settings .
MLLT4/Afadin plays a critical role in cell adhesion and junction formation. To effectively study these interactions:
Co-immunoprecipitation protocols: For MLLT4 co-IP studies, use a buffer system containing 50mM Tris-HCl (pH 7.5), 150mM NaCl, 1% NP-40, and appropriate protease inhibitors. Pre-clear lysates with protein A/G beads before antibody incubation to reduce non-specific binding .
Proximity ligation assays: When studying MLLT4 interactions with other adhesion molecules like N-cadherin, combine primary antibodies from different species with species-specific secondary antibodies for in situ proximity detection .
Immunofluorescence co-localization: Use confocal microscopy with validated MLLT4 antibodies (typically at 1:100 dilution) in combination with antibodies against known interaction partners such as N-cadherin to visualize co-localization at cell junctions .
Domain-specific antibodies: For mapping specific interaction domains, select antibodies targeting different regions of MLLT4, particularly those recognizing the PDZ domain which mediates many protein-protein interactions .
Research has demonstrated that MLLT4's interaction with RGCs is critical for controlling progenitor cell proliferation during neocortical development, making these methodological approaches particularly valuable for developmental neurobiology research .
When utilizing mouse models to study MLLT4-related leukemias, several methodological considerations are critical:
Model selection and validation: Different MLL-fusion protein mouse models recapitulate distinct aspects of human disease. For MLL-AF4 studies, conditional knockout models using Cre-lox systems (such as Mllt4-flox mice crossed with Emx1-Cre mice) allow tissue-specific expression .
Antibody validation in mouse tissues: Confirm cross-reactivity and specificity of MLLT4 antibodies in mouse samples before experimental use. Western blotting of mouse tissues alongside human positive controls is recommended .
Immunophenotyping protocols: For flow cytometry of mouse leukemia cells, use the following antibody panel alongside anti-MLLT4:
Transplantation studies: When transplanting MLLT4 fusion-expressing cells into recipient mice (NSG or NSGS), confirm engraftment through peripheral blood sampling at 4-week intervals using species-specific MLLT4 antibodies .
Research has demonstrated that the cellular context significantly affects MLLT4 fusion protein activity, with MLL-AF4 showing stronger leukemogenic potential in lymphoid versus myeloid contexts .
MLLT4-AS1 is a long non-coding RNA associated with MLLT4 that has emerged as an important regulatory molecule in autophagy and cancer. Research methodologies for investigating this relationship include:
RNA-protein interaction studies: RNA fluorescence in situ hybridization (RNA-FISH) combined with MLLT4 immunofluorescence can visualize co-localization. This requires careful fixation protocols (typically 4% paraformaldehyde for 10 minutes) and RNase-free conditions .
RNA immunoprecipitation (RIP): To detect physical interactions between MLLT4-AS1 and proteins:
Expression correlation analysis: Quantify both MLLT4-AS1 and MLLT4 levels across tissue samples using RT-qPCR and Western blotting, respectively, to establish expression pattern relationships .
Functional studies: The impact of MLLT4-AS1 on MLLT4 function can be assessed through:
Research has revealed that MLLT4-AS1 is upregulated by the MTORC inhibitor PP242 and rapamycin in cervical cells, promoting autophagy through interaction with myosin-9 protein and ATG14 transcription regulation rather than directly with MLLT4 protein itself .
Detection of MLLT4 fusion proteins presents several technical challenges that require specific methodological approaches:
Protein size considerations: The MLL-AF4 fusion protein is large (~376 kDa), necessitating:
Epitope accessibility: Fusion proteins may have altered epitope exposure. Recommendations include:
Sample heterogeneity: Leukemia samples often contain mixed populations. Address by:
Low abundance detection: For minimal residual disease monitoring:
Research indicates that MLL-AF4 is predominantly associated with pro-B-ALL and much more rarely with AML, making proper selection of control samples critical for antibody validation in different leukemia subtypes .
MLLT4/Afadin's critical role in cell adhesion and migration requires specific experimental approaches:
Cell junction visualization: For optimal immunofluorescence of MLLT4 at cell junctions:
Migration assay protocols: For studying MLLT4's impact on cell migration in cancer models:
Live-cell imaging: To track dynamic MLLT4 behavior:
Wound healing assays: For studying collective cell migration:
Research has demonstrated that MLLT4-AS1 overexpression significantly inhibits the invasive and migratory abilities of cervical cancer cells, suggesting important roles for the MLLT4 regulatory network in cancer progression .
MLLT4/Afadin plays a crucial role in neurodevelopment, particularly in cortical formation. When studying these processes:
Brain section preparation: For optimal MLLT4 detection in brain tissue:
Neuronal culture immunostaining: For primary neurons or neural progenitors:
Cortical development analysis: When studying double cortex formation:
Neurogenesis assays: For proliferation studies:
Research has shown that conditional inactivation of MLLT4 using Emx1-Cre mice results in widespread disruption of adherens junctions between radial glial cells and dramatically increased progenitor proliferation, leading to a double cortex-like phenotype in adult mice. This makes MLLT4 antibodies valuable tools for studying cortical malformation disorders .
The emerging connection between MLLT4-AS1 and autophagy regulation necessitates specific methodological approaches:
Autophagy flux assessment: When studying MLLT4-AS1's impact on autophagy:
Co-localization studies: For assessing MLLT4-AS1's interaction with autophagy machinery:
Immunoprecipitation protocol: For protein-RNA interactions:
In vivo autophagy visualization: For tissue samples from xenograft models:
Research has shown that MLLT4-AS1 was upregulated by H3K27ac modification with PP242 treatment, and knockdown of MLLT4-AS1 reversed autophagy by modulating ATG14 expression. This suggests a regulatory pathway where MLLT4-AS1 associates with myosin-9 protein to promote ATG14 transcription and induce autophagy .
While MLLT4 itself has not been widely targeted for antibody-drug conjugate (ADC) development, related research on leukocyte immunoglobulin-like receptor B4 (LILRB4) in AML provides valuable methodological insights for researchers interested in developing MLLT4-targeting therapies:
Antibody selection criteria: When selecting antibodies for potential ADC development:
Conjugation technologies: For creating homogeneous antibody-drug conjugates:
Payload selection considerations: When selecting cytotoxic payloads:
Pharmacokinetic assessment: For evaluating ADC behavior in vivo:
Research has demonstrated that homogeneous antibody-drug conjugates can achieve remarkable therapeutic effects with minimal toxicity in xenograft mouse models of disseminated human AML, suggesting similar approaches could be valuable for MLLT4-targeted therapies in leukemias with MLL rearrangements .