The MLLT10 antibody is a polyclonal reagent designed to detect the endogenous MLLT10 (myeloid/lymphoid or mixed-lineage leukemia translocated to 10) protein, also known as AF-10. This antibody is primarily used in research to study the role of MLLT10 in chromatin remodeling, leukemia pathogenesis, and developmental biology . MLLT10 is a critical component of chromosomal translocations observed in acute leukemias, particularly those involving the MLL1 (KMT2A) gene .
The antibody is utilized in:
Leukemia Studies: Detects MLLT10 in translocation-driven malignancies (e.g., t(10;11)(p12;q23) in AML and ALL) .
Developmental Biology: Investigates MLLT10’s role in craniofacial development via H3K79 methylation regulation .
Mechanistic Insights: Identifies interactions with DOT1L, a histone methyltransferase critical for leukemogenesis .
Leukemia Diagnostics: MLLT10 translocations are biomarkers for high-risk AML/ALL subtypes .
Therapeutic Targeting: DOT1L inhibitors (e.g., pinometostat) show promise in MLLT10-rearranged leukemias .
Further studies are needed to:
Explore MLLT10’s role in non-hematopoietic cancers.
Validate the antibody’s utility in detecting novel fusion proteins.
Develop standardized protocols for clinical research applications.
MLLT10, also known as AF10 (ALL1-fused gene from chromosome 10 protein), is a transcriptional regulator protein encoded by the MLLT10 gene. It contains several key structural domains:
N-terminal zinc finger domain
C-terminal leucine zipper domain (OM-LZ)
Octapeptide motif
PHD finger-like domain
The leucine zipper motif is particularly significant as it mediates interactions with GAS41, which in turn interacts with integrase interactor-1 (INI1), a component of the SWI/SNF chromatin remodeling complex. This suggests MLLT10's involvement in chromatin modification and transcriptional regulation .
MLLT10 functions as a transcriptional regulator with several identified roles:
Acts as a cofactor for DOT1L-mediated histone H3K79 methylation
Regulates cell identity maintenance in somatic cells
Functions in chromatin remodeling through interactions with the SWI/SNF complex
May play a role in transcriptional initiation
Mechanistically, MLLT10 facilitates higher-order (di- and tri-) methylation of H3K79 through direct interaction with the histone methyltransferase DOT1L. This epigenetic modification is crucial for maintaining cell identity and preventing cellular reprogramming .
MLLT10 is implicated in several hematological malignancies through chromosomal translocations:
| Fusion Gene | Translocation | Associated Malignancies | Clinical Features |
|---|---|---|---|
| PICALM-MLLT10 (formerly CALM-AF10) | t(10;11)(p12;q14) | AML, ALL, MPAL | Poor prognosis, mixed clinical manifestations |
| MLL-MLLT10 | t(10;11)(p12;q23) | Various leukemias | Often aggressive disease course |
The resulting fusion proteins consistently retain the leucine zipper motif of MLLT10, which is believed to interfere with normal gene regulation through interactions with chromatin remodeling complexes. In a recent Chinese cohort study, patients with PICALM-MLLT10 fusion gene (median age 25 years) demonstrated poor survival, with three out of six patients dying within one year despite intensive treatment .
Beyond hematological malignancies, MLLT10 has been implicated in several solid tumors:
Colorectal cancer (CRC): MLLT10 promotes tumor migration, invasion, and metastasis via epithelial-mesenchymal transition (EMT). Higher expression of MLLT10 is associated with increased CRC cell migration and invasion both in vitro and in vivo. At metastatic sites, MLLT10 expression correlates with increased Vimentin and decreased E-Cadherin expression, indicating its role in EMT regulation .
Non-small cell lung cancer (NSCLC): MLLT10 is targeted by miR-331-3p, which acts as a tumor suppressor. Overexpression of MLLT10 promotes tumor cell proliferation, EMT-mediated metastasis, and invasion in NSCLC. The miR-331-3p/MLLT10 axis has been proposed as a potential therapeutic target for NSCLC .
MLLT10 antibodies have been validated for multiple experimental applications:
| Application | Recommended Dilution | Expected Results | Considerations |
|---|---|---|---|
| Western Blotting (WB) | 1:500-1:1000 | Detection of endogenous MLLT10 protein (~110-120 kDa) | Use GAPDH or other housekeeping proteins as loading controls |
| Immunohistochemistry (IHC) | 1:200-1:500 | Nuclear localization in positive tissues | Antigen retrieval may be necessary |
| Immunofluorescence (IF) | 0.25-2 μg/mL | Nuclear distribution pattern | Wild-type shows specific nuclear localization while mutants may show aberrant patterns |
These applications allow researchers to detect endogenous levels of MLLT10/AF-10 protein in various experimental settings .
Detecting MLLT10 fusion proteins (such as PICALM-MLLT10 or MLL-MLLT10) requires special considerations:
Epitope selection: Choose antibodies raised against regions preserved in the fusion protein. For MLL-MLLT10 fusions, antibodies targeting the C-terminal region of MLLT10 are preferable since this region is typically retained in the fusion.
Size verification: Fusion proteins will have different molecular weights than wild-type MLLT10. For example:
Wild-type MLLT10: ~110-120 kDa
MLL-MLLT10: ~200-230 kDa (depending on breakpoints)
PICALM-MLLT10: ~140-170 kDa
Dual antibody approach: For definitive identification, use antibodies against both fusion partners simultaneously (e.g., anti-MLL and anti-MLLT10) in co-immunoprecipitation or co-localization studies .
MLLT10 has been identified as a barrier to cellular reprogramming through its regulation of H3K79 methylation. To investigate this function:
Chromatin immunoprecipitation (ChIP): Use MLLT10 antibodies in conjunction with H3K79me2/me3 antibodies to identify genomic regions where MLLT10 regulates H3K79 methylation during reprogramming.
Co-immunoprecipitation (Co-IP): Implement MLLT10 antibodies to pull down associated proteins like DOT1L to understand the dynamic changes in complex formation during reprogramming.
Loss-of-function/rescue experiments: After MLLT10 knockdown, perform rescue experiments using wild-type or mutant MLLT10 (particularly the DOT1L binding-impaired OM-LZ deletion mutant), and use antibodies to verify:
MLLT10 expression levels
H3K79 methylation status
Expression of pluripotency markers (OCT4, NANOG, SSEA4)
A key finding from research is that while wild-type MLLT10, when reintroduced, can reduce reprogramming efficiency in MLLT10 knockout cells, the DOT1L binding-impaired mutant cannot, suggesting that the MLLT10-DOT1L interaction is critical for maintaining somatic cell identity .
Validating antibody specificity is crucial for reliable results:
Positive and negative controls:
Positive: Cell lines with known MLLT10 expression (e.g., leukemia cell lines)
Negative: MLLT10 knockout cells generated via CRISPR/Cas9 (targeting splice site exon 2 or exon 3)
Multiple detection methods:
Western blot should show a band at the expected molecular weight (~110-120 kDa)
Immunofluorescence should demonstrate proper nuclear localization
Signal should diminish with siRNA or CRISPR/Cas9-mediated MLLT10 knockdown
Cross-reactivity assessment:
Test against related proteins (e.g., other MLL fusion partners)
Validate across multiple species if working with non-human models
Epitope verification:
Successful immunoprecipitation of MLLT10 and its protein complexes requires specific conditions:
Lysis buffer composition:
RIPA buffer with reduced detergent concentration to preserve protein-protein interactions
Include protease inhibitors, phosphatase inhibitors, and HDAC inhibitors
For chromatin-associated complexes, include DNase I treatment
Antibody selection and protocol:
Use 2-5 μg of antibody per sample
Pre-clear lysates with protein A/G beads
Incubate antibody with lysate overnight at 4°C
For weaker interactions (like with DOT1L), consider crosslinking with DSP or formaldehyde
Verification of interacting partners:
MLLT10's role in epigenetic regulation can be studied using several antibody-dependent approaches:
Sequential ChIP (ChIP-reChIP):
First ChIP with anti-MLLT10 followed by second ChIP with anti-H3K79me2/me3
This identifies genomic regions where MLLT10 and H3K79 methylation co-occur
Proximity-based labeling:
BioID approach using MLLT10 fusion with biotin ligase
Identify proximal proteins using streptavidin pulldown
Verify interactions with MLLT10 antibodies
Functional domain analysis:
Compare wild-type MLLT10 with mutants (L107A histone-binding mutant or OM-LZ DOT1L-binding deletion mutant)
Use antibodies to confirm expression and localization
Assess effects on H3K79 methylation levels
When analyzing patient samples that may contain MLLT10 fusion proteins:
Use multiple antibodies targeting different regions:
Antibodies against N-terminal regions will detect only wild-type MLLT10
Antibodies against C-terminal regions will detect both wild-type and most fusion proteins
Size discrimination:
Run gradient gels (4-15%) to better separate proteins of different molecular weights
Use size markers that span the range of both wild-type and fusion proteins
Fusion-specific detection:
Design PCR primers spanning the fusion breakpoint
Use fluorescence in situ hybridization (FISH) as a complementary approach
Consider RNA-seq to identify fusion transcripts
Controls for validation:
Researchers should be aware of several challenges:
Expression level variations:
MLLT10 expression varies significantly across tissue types
Low expression may require signal amplification techniques
Consider enrichment of nuclear fraction before analysis
Isoform diversity:
Multiple MLLT10 isoforms exist (including types I, III, and IV)
Antibodies may have differential affinity for specific isoforms
Western blots may show multiple bands representing different isoforms
Subcellular localization:
MLLT10 primarily shows nuclear localization
Mutant forms may show aberrant localization patterns
Perform proper nuclear/cytoplasmic fractionation for accurate assessment
Context-dependent interactions:
MLLT10 antibodies can support therapeutic development through several approaches:
Target validation in preclinical models:
Immunohistochemistry to confirm MLLT10 expression in patient-derived xenografts
Correlation of MLLT10 levels with response to DOT1L inhibitors
Analysis of MLLT10-dependent pathways in drug-resistant samples
Biomarker development:
Quantitative assessment of MLLT10 levels in patient samples
Correlation with clinical outcomes and treatment response
Development of companion diagnostics for targeted therapies
Mechanism-of-action studies:
Several cutting-edge technologies offer promising avenues for MLLT10 research:
Single-cell protein analysis:
Mass cytometry (CyTOF) with MLLT10 antibodies
Single-cell Western blotting
Co-detection of MLLT10 with other markers at single-cell resolution
Advanced imaging approaches:
Super-resolution microscopy to visualize MLLT10 chromatin interactions
Live-cell imaging with fluorescently labeled antibody fragments
Multiplexed ion beam imaging (MIBI) for spatial analysis in tissues
Proteomics integration:
Combine MLLT10 antibody-based purification with mass spectrometry
Phospho-proteomics to identify signaling pathways affected by MLLT10
Thermal proteome profiling to identify drugs targeting MLLT10 complexes
CRISPR screens with antibody readouts: