The term "ML4 Antibody" is not directly defined in available literature, but contextual analysis suggests two potential interpretations based on search results:
Anti-ID4 Antibody (clone DML07): Cataloged as MABE164 by Merck Millipore, this monoclonal antibody targets the human ID4 protein, a transcriptional regulator involved in cellular differentiation and cancer progression .
MLLT4/AF-6 Antibodies: Antibodies targeting the MLLT4 gene product (AF-6), a cell adhesion molecule linked to leukemia and epithelial cancers .
This article focuses on Anti-ID4 Antibody (MABE164) due to its well-documented properties and research applications.
Function: ID4 (Inhibitor of DNA Binding 4) regulates gene expression by binding to transcription factors, impacting cell cycle progression and tumor suppression .
Disease Relevance: Overexpression correlates with glioblastoma, breast cancer, and leukemia .
Glioblastoma: Anti-ID4 antibodies detect ID4 overexpression in tumor tissues, aiding in prognosis .
Breast Cancer: ID4 silencing via antibody-mediated targeting reduces tumor growth in vitro .
ID4 modulates NOTCH and WNT signaling pathways, making it a therapeutic target for hormone-resistant cancers .
Therapeutic Development: Engineered variants of Anti-ID4 antibodies for targeted drug delivery .
Diagnostic Tools: Integration with single-cell Western platforms (e.g., Milo Antibody Database) for high-resolution tumor profiling .
Validation typically involves:
Knockout controls: Compare protein detection in wild-type vs. ML4-deficient cell lines (e.g., CRISPR-Cas9-generated Mll4 −/− mouse embryonic stem cells) .
Multi-method confirmation: Combine Western blot (1:500 dilution in PBS-Tween) and ELISA (1:1,000–1:5,000 dilution) to confirm target engagement .
Epitope mapping: Use synthetic peptides matching immunogen sequences to verify antibody-antigen binding (Figure 1 in ).
Chromatin remodeling studies: Investigate ML4’s role in H3K4 methylation using ChIP-seq in hematopoietic malignancies .
Disease modeling: Assess ML4 expression in mouse models of mixed-lineage leukemia and immune dysregulation .
Protein interaction assays: Co-immunoprecipitation to identify binding partners like histone-modifying enzymes .
Phylogenetic alignment: Compare ML4 ortholog sequences (e.g., human vs. chimpanzee) to predict conserved epitopes .
Empirical testing: Validate in non-model species (e.g., primates) using flow cytometry or immunohistochemistry (IHC) .
Structural modeling: Predict antibody-paratope compatibility using AlphaFold2 for untested species .
Signal amplification: Use tyramide-based systems (e.g., Opal™) for IHC in archival tissue .
Pre-clearing: Reduce background via pre-absorption with Mll4 −/− lysates in Western blot .
Multiplex assays: Pair with fluorescence-conjugated secondary antibodies for dual-target visualization .
Isotype-matched IgG: Rule out nonspecific binding in ChIP-seq .
Peptide competition: Pre-incubate antibody with immunogen peptides to confirm signal loss .
Biological replicates: Account for batch effects in histone modification studies (n ≥ 3) .
Conditional knockout models: Use Cre-lox systems to delete Mll4 in specific cell lineages (e.g., B cells vs. myeloid cells) .
Pathway enrichment analysis: Integrate RNA-seq data with KEGG pathways to identify context-dependent interactors .
In vivo inhibition: Test small-molecule blockers of ML4’s SET domain in xenograft models .
ELISA titer stability: Maintain absorbance ≥2.0 at 1:1,000 dilution across batches .
Knockout validation: Ensure no signal in Mll4 −/− lysates via Western blot (Figure 2 in ).
Inter-lab benchmarking: Share aliquots with collaborating labs for cross-validation (e.g., SickKids Research Institute protocols ).