KMT2E antibody has been pivotal in elucidating the role of KMT2E in hypoxia-driven pathologies. For example, in pulmonary arterial hypertension (PAH), KMT2E interacts with the lncRNA KMT2E-AS1 to stabilize HIF-2α and drive metabolic reprogramming . Antibodies enabled:
RNA-protein interaction assays: Demonstrated direct binding between KMT2E-AS1 and KMT2E under hypoxic conditions .
H3K4me3 quantification: Showed increased histone trimethylation in hypoxic pulmonary endothelial cells .
Pharmacological interventions: Validated histone methyltransferase inhibitors in reversing PAH pathology .
Mutations in KMT2E are linked to intellectual disability, autism, and macrocephaly . Antibodies facilitate:
Diagnostic screening: Detection of KMT2E protein in patient-derived tissues.
Functional studies: Analysis of KMT2E’s role in cell cycle regulation and genomic stability .
KMT2E-AS1/KMT2E axis exacerbates PAH via:
Epigenetic activation: Enhances H3K4me3 at HIF-2α targets, promoting glycolysis and endothelial proliferation .
Metabolic reprogramming: Increases lactate dehydrogenase (LDH) activity and extracellular acidification rate (ECAR) .
Phenotypic modulation: Antibody-based knockdown reduces endothelin-1 (EDN1) secretion and vascular remodeling .
Heterozygous KMT2E variants correlate with:
Intellectual disability: Disrupted PHD/SET domain function alters chromatin dynamics .
Autism and epilepsy: Dysregulated KMT2E expression impacts synaptic plasticity .
KMT2E binds H3K4me3 via its PHD domain, recruiting histone deacetylase (HDAC) complexes to active chromatin regions . Antibody studies reveal:
NcoR-HDAC3 complex recruitment: Mediates transcriptional repression of cell cycle genes .
HIF-2α stabilization: Antibody knockdown disrupts KMT2E-AS1/KMT2E interaction, reducing HIF-2α protein levels .
KMT2E (lysine methyltransferase 2E) is a protein-coding gene located on chromosome 7 and a member of the myeloid/lymphoid or mixed-lineage leukemia (MLL) family. It encodes a protein with an N-terminal PHD zinc finger and a central SET domain . KMT2E is particularly important in research because it functions as:
A key regulator of hematopoiesis, contributing to terminal myeloid differentiation and hematopoietic stem cell self-renewal through DNA methylation
A crucial cell cycle regulator, influencing the G1/S transition, S phase progression, and mitotic entry
A protein that binds to chromatin regions downstream of active gene start sites, regulating gene transcription through interaction with tri-methylated histone H3 at Lys-4 (H3K4me3)
Additionally, mutations in KMT2E have been associated with neurodevelopmental disorders including intellectual disability, autism, macrocephaly, hypotonia, gastrointestinal abnormalities, and epilepsy , making it relevant for both oncology and neuroscience research.
Current research-grade KMT2E antibodies include:
Most available KMT2E antibodies are raised in rabbits against human KMT2E, with cross-reactivity to mouse and rat in some cases. These antibodies are typically validated for Western blotting, immunohistochemistry, and immunofluorescence applications .
KMT2E is a large protein of 1,858 amino acids with specific functional domains:
Functionally, KMT2E:
Is recruited to E2F1 responsive promoters by HCFC1, promoting tri-methylation of histone H3 at Lys-4 (H3K4me3) and transcriptional activation
During myoblast differentiation, suppresses inappropriate expression of S-phase-promoting genes while maintaining the expression of determination genes in quiescent cells
For optimal Western blotting results with KMT2E antibodies:
Sample preparation:
Electrophoresis conditions:
Use low percentage (6-8%) SDS-PAGE gels to effectively resolve this high molecular weight protein
Run gels at lower voltage (80-100V) to prevent distortion of large proteins
Transfer parameters:
Utilize wet transfer methods with cooled buffers
Extend transfer time (overnight at 30V) for complete transfer of large proteins
Antibody dilutions:
Controls:
Include positive controls from cell lines known to express KMT2E
Consider knockdown samples as negative controls to confirm specificity
Research has shown that KMT2E protein levels can be stabilized under hypoxic conditions, with short-term (4 hours) exposure to transcriptional inhibitor actinomycin D reducing KMT2E transcript but not protein expression , suggesting potential experimental considerations when studying protein stability.
For effective immunohistochemical detection of KMT2E:
Fixation and embedding:
Use 10% neutral buffered formalin for tissue fixation (12-24 hours)
Standard paraffin embedding protocols are suitable
Antigen retrieval:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) is recommended
Pressure cooking for 20 minutes typically yields better results than microwave heating
Antibody dilution and incubation:
Detection systems:
HRP-conjugated secondary antibodies with DAB substrates provide good signal-to-noise ratio
For dual labeling studies, consider fluorescent secondary antibodies
Validation approaches:
Research has revealed increased expression of H3K9me3 (but not H3K27me3) in diseased pulmonary arterioles in human group 1 PAH and group 3 PH , which could be useful as a comparative marker in studies involving KMT2E.
Based on recent research findings, the following experimental approach is recommended:
RNA-protein interaction studies:
Proximity ligation assays:
Knockdown and overexpression experiments:
Design effective siRNAs targeting KMT2E-AS1 to assess effects on KMT2E protein expression
Create lentiviral constructs for forced expression of KMT2E-AS1 to study function
Generate deletion mutants of KMT2E-AS1 missing conserved sequences (~600-bp) to identify functional domains
Histone modification analysis:
Assess H3K4me3 and H3K9me3 marks via immunoblotting after KMT2E-AS1 manipulation
Monitor changes under hypoxic versus normoxic conditions
Research has demonstrated that KMT2E-AS1 complexes with and stabilizes KMT2E protein to increase H3K4me3 histone trimethylation , and that KMT2E deficiency can induce a histone demethylase (LSD1) that specifically reduces H3K9me3 .
KMT2E plays a critical role in hematopoietic regulation through several epigenetic mechanisms:
Chromatin binding and modification:
Hematopoietic stem cell maintenance:
Terminal myeloid differentiation:
Cell cycle regulation in hematopoietic cells:
Recent research has identified a genetic association between rs73184087, a single-nucleotide variant within a KMT2E intron, and disease risk in pulmonary arterial hypertension (PAH), suggesting potential pathogenic roles of KMT2E in vascular diseases beyond its known hematopoietic functions .
KMT2E has been implicated in several neurodevelopmental processes and disorders:
Genetic evidence:
Clinical manifestations:
Developmental delay and/or intellectual disability
Autism spectrum disorder or autistic features
Low muscle tone (hypotonia)
Seizures and speech delay
Head size abnormalities (both macrocephaly and microcephaly reported)
Sleep issues and behavioral problems including self-injury, anxiety, and aggression
Gastrointestinal issues
Molecular mechanisms:
KMT2E is thought to regulate neurodevelopmental genes through histone modifications
Studies suggest roles in neuronal differentiation and maturation
The protein's SET domain, though predicted to be inactive, may still influence other epigenetic regulators
Phenotypic variability:
Research indicates that therapeutic approaches for KMT2E-related syndrome should begin as early as possible, ideally before a child begins school, though currently there are no medicines specifically designed to treat the syndrome .
Recent research has uncovered a significant role for KMT2E in pulmonary hypertension through its interaction with HIF-2α. The following experimental approaches are recommended:
Co-immunoprecipitation studies:
Use KMT2E antibodies to pull down protein complexes from hypoxic pulmonary arterial endothelial cells
Probe for HIF-2α to confirm interaction
Compare normoxic versus hypoxic conditions to assess oxygen-dependent interactions
ChIP-seq analysis:
Perform chromatin immunoprecipitation with KMT2E antibodies followed by sequencing
Identify genomic regions co-occupied by KMT2E and HIF-2α
Analyze enrichment for histone marks such as H3K4me3
Functional assays:
Combine KMT2E antibody-based detection with KMT2E-AS1 manipulation
Research has shown that KMT2E-AS1 stabilizes KMT2E protein to increase H3K4me3, driving HIF-2α-dependent metabolic and pathogenic endothelial activity
Monitor changes in HIF-2α expression and activity across epigenetic, transcriptional, and posttranscriptional contexts
Genetic variant analysis:
Research has demonstrated that KMT2E-AS1 and KMT2E act in a positive feedback loop with HIF-2α to exacerbate pulmonary arterial hypertension through epigenetic and metabolic changes . In vivo studies have shown that KMT2E-AS1 deficiency protected against PAH in mice, as did pharmacologic inhibition of histone methylation in rats, while forced lncRNA expression promoted more severe pulmonary hypertension .
When working with KMT2E antibodies, researchers may encounter several specificity challenges:
High molecular weight detection issues:
Problem: Incomplete transfer of this large protein (204 kDa) during Western blotting
Solution: Extended transfer times, lower percentage gels, and wet transfer methods
Non-specific bands:
Cross-reactivity concerns:
Epitope masking:
Validation approaches:
Some antibodies, like those from Bio-Rad, have been validated for cross-reactivity with mouse KMT2E , which is important for translational studies between human and animal models.
When facing contradictory findings in KMT2E expression analysis:
Consider cellular context and conditions:
Evaluate detection methods:
Assess genetic background:
Examine experimental timepoints:
KMT2E functions in cell cycle regulation, so expression may vary based on cell cycle phase
Compare acute versus chronic manipulation experiments
Consider protein-protein interactions:
Research has shown that proteasomal inhibition with MG132 can reverse the effect of KMT2E-AS1 knockdown on KMT2E protein levels , suggesting that protein stability mechanisms significantly impact observed expression levels and should be considered when interpreting conflicting results.
To address discrepancies between in vitro cell culture and in vivo animal model results:
Microenvironment factors:
Developmental timing:
Compensatory mechanisms:
In vivo systems may activate compensatory pathways absent in vitro
Other MLL family proteins may partially compensate for KMT2E dysfunction in intact organisms
Cell type heterogeneity:
Tissue samples contain multiple cell types with varying KMT2E expression
Compare results from homogeneous cell cultures with heterogeneous tissue samples
Consider single-cell approaches to resolve cell-specific effects
Methodological reconciliation:
Emerging methodologies for investigating KMT2E in blood cancers include:
Single-cell multi-omics:
Combine single-cell RNA-seq with ChIP-seq using KMT2E antibodies
Correlate KMT2E binding patterns with gene expression at single-cell resolution
This approach can identify heterogeneous cell populations with differential KMT2E activity
CRISPR-based epigenome editing:
Target KMT2E to specific genomic loci using dCas9-KMT2E fusions
Assess the direct impact of KMT2E recruitment on gene expression and cell transformation
Compare with effects of other MLL family members to determine functional specificity
Patient-derived xenograft (PDX) models:
Establish PDX models from hematological malignancies
Use KMT2E antibodies to monitor protein expression and localization
Test targeted therapies against the KMT2E pathway in these models
Liquid biopsy approaches:
Develop techniques to detect KMT2E protein or KMT2E-AS1 in circulation
Correlate with disease progression and treatment response
Early research suggests potential for biomarker development
Targeting the KMT2E-AS1/KMT2E interaction:
KMT2E has been implicated in myeloid/lymphoid or mixed-lineage leukemia, and aberrant levels have been shown to inhibit cell cycle progression , making it a promising target for therapeutic intervention in hematological malignancies.
Innovative applications of KMT2E antibodies in neurodevelopmental research include:
Brain organoid studies:
Use KMT2E antibodies for immunostaining of human brain organoids
Compare organoids derived from patients with KMT2E-related syndrome versus controls
Track KMT2E expression during organoid development to identify critical windows
Genome-wide binding profiling:
Perform ChIP-seq with KMT2E antibodies in neural progenitor cells and mature neurons
Identify neuronal genes regulated by KMT2E
Compare binding profiles between cells with wild-type and mutant KMT2E
Protein interaction networks:
Use KMT2E antibodies for co-immunoprecipitation followed by mass spectrometry
Map KMT2E interaction partners in neural tissues
Compare interactomes between normal and KMT2E-mutant samples
Post-mortem tissue analysis:
Apply KMT2E antibodies for immunohistochemistry on brain sections
Examine expression patterns in individuals with neurodevelopmental disorders
Compare with genetic data on KMT2E mutations when available
Therapeutic monitoring:
Research has shown that KMT2E mutations are associated with a range of neurological disorders including epilepsy, autism, and abnormalities in gastrointestinal function , making KMT2E antibodies valuable tools for studying these conditions.
Recent research has revealed promising avenues for targeting KMT2E in pulmonary hypertension treatment:
Inhibition of the KMT2E-AS1/KMT2E axis:
Histone methylation inhibition:
Targeting the rs73184087 variant:
Biomarker development:
KMT2E antibodies could be used to develop assays for monitoring disease progression
Changes in KMT2E protein levels or post-translational modifications might correlate with disease severity
Combined with genetic screening for the rs73184087 variant, this could enable personalized treatment approaches
Combination therapies:
Targeting the KMT2E pathway alongside standard PAH therapies
KMT2E antibodies can be used to monitor pathway modulation in response to treatment
Synergistic approaches targeting both KMT2E and HIF-2α pathways may provide enhanced efficacy