KMT2D Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
-20°C, pH 7.4 PBS, 0.05% NaN3, 40% Glycerol
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery details.
Synonyms
AAD10 antibody; ALL1 related gene antibody; ALL1-related protein antibody; ALR antibody; CAGL114 antibody; Histone-lysine N-methyltransferase MLL2 antibody; KABUK1 antibody; Kabuki make up syndrome antibody; Kabuki mental retardation syndrome antibody; KMS antibody; KMT2B antibody; KMT2D antibody; Lysine N methyltransferase 2D antibody; Lysine N-methyltransferase 2B antibody; MLL2 antibody; MLL2_HUMAN antibody; MLL4 antibody; Myeloid/lymphoid or mixed lineage leukemia 2 antibody; Myeloid/lymphoid or mixed-lineage leukemia protein 2 antibody; TNRC21 antibody; Trinucleotide repeat containing 21 antibody
Target Names
Uniprot No.

Target Background

Function
KMT2D is a histone methyltransferase that catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to the epsilon-amino group of Lysine 4 on histone H3 (H3K4). As a component of the chromatin remodeling machinery, KMT2D predominantly forms H3K4me1 methylation marks at active chromatin sites, where transcription and DNA repair processes take place. KMT2D functions as a coactivator for the estrogen receptor by being recruited by ESR1, thereby activating transcription.
Gene References Into Functions
  • Clinical and neurobehavioral features of three novel Kabuki Syndrome unrelated patients with mosaic KMT2D mutations have been described. PMID: 29283410
  • Research suggests that overexpression of MLL2 predicts poor clinical outcomes and facilitates ESCC tumor progression, and it may exert an oncogenic role via activation of EMT. PMID: 29532228
  • Findings reveal a critical role for KMT2D in the control of epithelial enhancers and p63 target gene expression. This includes its requirement for the maintenance of epithelial progenitor gene expression and the coordination of proper terminal differentiation. PMID: 29440247
  • Congenital heart defects (CHD) are detected in 70% of patients with KMT2D (MLL2) pathogenic variants, most commonly left-sided obstructive lesions, including multiple left-sided obstructions similar to those observed in the Shone complex spectrum, and septal defects. PMID: 28884922
  • Analysis of highly recurrent genetic lesions in components of the NF-kappaB pathway, of NOTCH1 and NOTCH2 as well as KMT2D, suggests a potential role in ocular adnexal MALT-type marginal zone lymphomas. PMID: 27566587
  • Results highlight the emerging role of mutations in epigenetic regulators, particularly MLL2, in cervical carcinogenesis, suggesting a potential disruption of histone modifications. PMID: 28390392
  • Mutations of ARID1A, GPRC5A, and MLL2 grant bladder cancer non-stem cells the capability of self-renewal. PMID: 27387124
  • This study reports mutation screening in patients with Kabuki Syndrome Subtype 2 (KS2), identifying 12 novel KDM6A mutations. These findings confirm that female patients with KS2 may exhibit a milder manifestation of KS and may even develop normally in terms of cognitive function. PMID: 27302555
  • This case uniquely presents a sporadic co-occurrence of two genetic disorders: a de novo frameshift variant in the KMT2D gene and a de novo 3.2 Mbp 10q22.3q23.1 deletion. PMID: 28590022
  • KMT2D mutation is associated with small Cell Lung Cancer. PMID: 28007623
  • Two patients with Kabuki syndrome have been described, each presenting with different KMT2D mutations. Both cases include an interrupted/bipartite clavicle. PMID: 28256057
  • Data supports a key role for KMT2D in modulating the chromatin competence necessary for the assembly of the ER-FOXA1-PBX1 transcriptional regulatory network in breast cancer. PMID: 28336670
  • A possible correlation between the position of the KMT2D premature termination codon caused by the mutation and height SDS was assessed, but no significant difference could be observed for the Kabuki syndrome patients. PMID: 27530205
  • KMT2D p.Gln3575His segregated with disease status in the family and is associated with a unique and conserved phenotype in the affected family members, with features overlapping with Kabuki and CHARGE syndromes. These findings further support the potential etiological link between these two classically distinct conditions. PMID: 27991736
  • Three out of four cases of histiocytic sarcoma had alterations in the KMT2D gene. PMID: 28805986
  • This study shows the contribution of MLL2's methyltransferase and CXXC domain in the trimethylation of H3K4 in embryonic stem cells. While it trimethylates H3K4 at both bivalent gene promoters and non-TSS elements, it regulates transcription at a limited number of genes, including those required for primordial germ cell specification. PMID: 28157506
  • KMT2D Mutation is associated with esophageal squamous cell carcinoma. PMID: 27749841
  • MLL1 and MLL2 collaborate to regulate gene expression and leukemia maintenance, not through redundancy, but through distinct pathways. PMID: 28609655
  • These findings provide evidence that CHARGE and Kabuki syndromes result from dysregulation of CHD7 and KMT2D genes involved in embryonic development and expressed in a tissue-specific manner. PMID: 28475860
  • Low KMT2C and KMT2D expression in biopsies defines better outcome groups in pancreatic ductal adenocarcinoma. PMID: 27280393
  • Three mosaic missense and likely-gene disrupting mutations in genes previously implicated in ASD (KMT2C, NCKAP1, and MYH10) were identified in probands, but none in siblings. There's a strong ascertainment bias for mosaic mutations in probands relative to their unaffected siblings. PMID: 27632392
  • The enzymatic activity of H3K4 methyltransferase MLL4 is required for its protein stability. PMID: 28013028
  • A female Kabuki syndrome patient with typical dysmorphic features and developmental delay and a novel KMT2D mutation has been reported. PMID: 25944076
  • Pygo2 functions as a prognostic factor for glioma due to its up-regulation of H3K4me3 and promotion of MLL1/MLL2 complex recruitment. PMID: 26902498
  • Mutation in the MLL2 gene is associated with Kabuki Syndrome. PMID: 26757828
  • Data suggest that lysine methyltransferase 2D (KMT2D) mutations are rare in abdominal paraganglioma (PGLs). PMID: 26303934
  • A high proportion of recurrent somatic DICER1 and KMT2D mutations in this series of sporadic IO-MEPL points to their likely important roles in the molecular pathogenesis of these rare embryonal tumors. PMID: 26841698
  • Whereas Set1 targets are largely associated with the maintenance of the stem cell population, MLL1/2 targets are specifically enriched for genes involved in ciliogenesis. PMID: 26711341
  • Although enhancer priming by MLL4/KMT2D is dispensable for cell-identity maintenance in mice, it controls cell fate transition by orchestrating p300-mediated enhancer activation. PMID: 27698142
  • Our data suggest that MLL2 protein is overexpressed in primary gastrointestinal diffuse large B cell lymphoma and appears as a prognostic factor. PMID: 26722499
  • The results do not support the hypothesis that common germline genetic variants in the MLL2 genes are associated with the risk of developing medulloblastoma. PMID: 26290144
  • Mutations in the KMT2D gene are associated with cutaneous T cell lymphoma and Sezary syndrome. PMID: 26551667
  • In patients with Kabuki Syndrome, autosomal dominant KMT2D mutations are associated with dysregulation of terminal B-cell differentiation, leading to humoral immune deficiency and, in some cases, autoimmunity. PMID: 26194542
  • KMT2D represents a recurrently mutated gene with potential implications for pheochromocytoma development. PMID: 26032282
  • MLL4 is identified as a major mammalian H3K4 mono- and di-methyltransferase essential for enhancer activation during cell differentiation. PMID: 24368734
  • Similar to KMT2D, CHD7 interacts with members of the WAR complex, namely WDR5, ASH2L, and RbBP5. Therefore, it is proposed that CHD7 and KMT2D function in the same chromatin modification machinery. PMID: 24705355
  • KMT2D mutations may promote malignant outgrowth by perturbing the expression of tumor suppressor genes that control B cell-activating pathways. PMID: 26366710
  • Findings suggest that KMT2D acts as a tumor suppressor gene whose early loss facilitates lymphomagenesis by remodeling the epigenetic landscape of the cancer precursor cells. PMID: 26366712
  • Reduced or lost expression of MLL2 was commonly observed in tumor tissues compared to paired adjacent non-tumor tissues regardless of mutation status. PMID: 25112956
  • Mutations in MLL2 are present in approximately 27% of urothelial carcinoma cases. PMID: 26138514
  • Targeted sequencing in patients with acute lymphoblastic leukemia identified KMT2D and KIF1B as novel putative driver genes and a putative regulatory non-coding variant that coincided with overexpression of the growth factor MDK. PMID: 25355294
  • Ten out of the 11 mutations found in patients with Kabuki syndrome were novel. KMT2D mutations included four small deletions or insertions and four nonsense and two missense mutations. PMID: 24739679
  • Data identified mutations in epigenetic modifiers such as KMT2D as potential early driving events in lymphomagenesis and immune escape alterations as relapse-associated events in diffuse large B-cell lymphoma. PMID: 25123191
  • MLL2 mutation-positive patients have a more severe and typical Kabuki phenotype than the MLL2 mutation-negative group. PMID: 23320472
  • This study supports that KMT2D has distinct roles in neoplastic cells compared to normal cells. PMID: 24240169
  • MLL3 and MLL4 function in the regulation of enhancer activity. PMID: 24081332
  • The identification of novel MLL2 mutations in patients with Kabuki syndrome. PMID: 23913813
  • These results indicate that the suppression of MLL genes, especially MLL2 and MLL5, participates in modulating breast carcinogenesis. PMID: 23754336
  • Data indicate that seven genes showed statistical enrichment for mutation: TP53, RB1, PTEN, NFE2L2, KEAP1, MLL2, and PIK3CA. PMID: 24323028
  • MLL4 (KMT2D) is a major H3K4 mono- and di-methyltransferase with partial functional redundancy with MLL3 (KMT2C) in mouse and human cells. MLL4 is enriched on enhancers and is required for enhancer activation, cell-type-specific gene expression, and cell differentiation. PMID: 24368734

Show More

Hide All

Database Links

HGNC: 7133

OMIM: 147920

KEGG: hsa:8085

STRING: 9606.ENSP00000301067

UniGene: Hs.731384

Involvement In Disease
Kabuki syndrome 1 (KABUK1)
Protein Families
Class V-like SAM-binding methyltransferase superfamily, Histone-lysine methyltransferase family, TRX/MLL subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in most adult tissues, including a variety of hematoipoietic cells, with the exception of the liver.

Q&A

What is KMT2D and why is it significant in cancer research?

KMT2D is the largest H3K4 methyltransferase in the COMPASS/Set1 family and represents one of the most frequently mutated genes across multiple cancer types. The enzymatic function of KMT2D depends on several conserved C-terminal domains, including a PHD domain, two FY-rich motifs (FYRC and FYRN), and a catalytic SET domain . It plays a crucial role as a bona fide tumor suppressor gene in follicular lymphoma and diffuse large B-cell lymphoma .

In recent studies, KMT2D mutations have been identified in over 20% of lung squamous cell carcinoma (LUSC) cases, where it functions as a key regulator of tumorigenesis . When KMT2D is deleted in experimental models, it can transform lung basal cell organoids to LUSC and promote overgrowth and squamous differentiation, consistent with early malignant transformation . Additionally, KMT2D loss increases activation of receptor tyrosine kinases (RTKs) EGFR and ERBB2, partly through reprogramming the chromatin landscape .

What applications are validated for KMT2D antibodies?

KMT2D antibodies have been validated for multiple experimental applications with specific dilution recommendations:

ApplicationValidated DilutionPositive Controls
Western Blot (WB)1:2000-1:16000HEK-293 cells
Immunohistochemistry (IHC)1:50-1:500Human breast cancer tissue, human lymphoma tissue, human colon cancer tissue
Immunofluorescence (IF)Reported in multiple publicationsVarious as noted in literature
Chromatin Immunoprecipitation (ChIP)Reported in multiple publicationsDocumented in relevant studies
ELISAValidated, specific dilutions sample-dependentAs noted in validation data

For IHC applications, it is recommended to perform antigen retrieval with TE buffer pH 9.0, with citrate buffer pH 6.0 as an alternative . Researchers should titrate the antibody in each testing system to obtain optimal results as outcomes can be sample-dependent .

How can researchers validate the specificity of KMT2D antibodies?

Antibody specificity can be validated through several approaches:

First, verify antibody reactivity using positive controls such as HEK-293 cells for Western blot applications, which show the expected 593 kDa molecular weight for the full-length KMT2D protein . Observe caution that both calculated and observed molecular weights of KMT2D are approximately 593 kDa, making it a challenging protein to resolve on standard gels .

Second, employ genetic approaches using KMT2D knockdown or knockout models. Evidence from the literature confirms the specificity of certain antibodies, as demonstrated in studies where KMT2D protein loss was verified by Western blot after CRISPR/Cas9-mediated gene targeting . Similarly, samples carrying biallelic truncating mutations that eliminate the epitope recognized by C-terminal targeted antibodies showed absence of full-length KMT2D protein expression .

Third, consider epitope location when selecting antibodies. For instance, antibodies targeting the C-terminal portion of KMT2D can identify full-length protein but may not detect truncated versions resulting from genetic alterations .

How do KMT2D mutations affect antibody reactivity and experimental design?

KMT2D undergoes two primary types of genomic alterations: truncation and missense mutations . These mutations can significantly impact antibody recognition and experimental outcomes.

For truncation mutations, C-terminal-directed antibodies will fail to detect the protein as the epitope is typically lost. Research has shown that in samples with biallelic truncating mutations, immunoblot analysis using C-terminal antibodies revealed complete absence of intact KMT2D protein . Interestingly, even when using antibodies directed against the N-terminal half of KMT2D, detection of truncated proteins remains challenging despite expression of the mutant cDNA .

For missense mutations, most maintain protein stability levels comparable to wild-type. In a systematic study testing 16 DLBCL-derived mutant alleles alongside three germline variants as controls, all but one (S5404F) produced similar amounts of both mRNA and protein as the wild-type allele, indicating that missense mutations generally do not affect KMT2D protein stability .

When designing experiments involving KMT2D detection in cancer samples, researchers should employ multiple antibodies targeting different epitopes and combine this approach with genetic sequencing to fully characterize KMT2D status. Additionally, functional assays measuring H3K4 methyltransferase activity should be considered, as mutations can impair enzymatic function without affecting protein expression .

What methodological approaches are recommended for studying KMT2D's role in epigenetic regulation?

To effectively study KMT2D's role in epigenetic regulation, a multi-faceted approach is recommended:

For ChIP applications, antibodies targeting KMT2D can identify genomic binding sites. Studies have shown that over 95% of KMT2D-bound chromatin in germinal center B cells is decorated by H3K4me1 (36% of peaks) and/or H3K4me3 (62% of peaks) . This finding underscores KMT2D's role as a non-redundant methyltransferase that controls the methylation state of numerous regions in mature B cell compartments.

For epigenomic profiling, researchers should combine KMT2D ChIP with histone modification mapping (particularly H3K4me1/me3) and assessment of chromatin accessibility. This integrated approach can reveal how KMT2D loss affects enhancer activation and gene expression programs, as demonstrated in studies where KMT2D deficiency led to diminished global H3K4 methylation in germinal-center B-cells and DLBCL cells .

For functional validation, genetic manipulation of KMT2D (through conditional deletion, CRISPR-mediated knockout, or expression of mutant forms) combined with transcriptomic and epigenomic profiling provides comprehensive insights. For instance, conditional deletion of Kmt2d early during B cell development resulted in increased germinal center B-cells and enhanced B cell proliferation in mice , demonstrating that timing of KMT2D loss can significantly impact phenotypic outcomes.

How does combined haploinsufficiency of KMT2D with other epigenetic regulators affect experimental outcomes?

Recent research indicates that KMT2D and CREBBP (another epigenetic regulator) are paradoxically co-mutated in lymphomas despite having similar enhancer regulatory functions . This suggests important mechanistic implications for researchers studying epigenetic regulators.

Studies demonstrate that combined haploinsufficiency of Crebbp and Kmt2d (referred to as C+K) accelerates lymphomagenesis . Specifically, this combined loss causes disruption of super-enhancers driving immune synapse genes, leading to reduction of CD8 cells in lymphomas—directly linking super-enhancer function to immune surveillance and potentially contributing to immunotherapy resistance .

When designing experiments investigating KMT2D function, researchers should consider potential cooperative effects with other epigenetic modifiers. Transcriptional analysis revealed that dual CREBBP and KMT2D haploinsufficiency results in cooperative repression of genes related to germinal center exit/immune synapse response and DNA repair, while simultaneously inducing biosynthetic programs normally restricted to centrocytes undergoing T cell help .

For experimental design, this suggests that single-gene manipulation studies may not fully capture the complexity of KMT2D function in disease contexts where multiple epigenetic regulators are altered. Integration of multi-omics approaches (including trajectory analysis of gene expression changes) becomes essential to elucidate the full spectrum of biological effects resulting from combinatorial epigenetic dysregulation .

How can KMT2D antibodies be utilized to investigate its role in lung cancer development?

KMT2D has emerged as a critical player in lung squamous cell carcinoma (LUSC), with mutations present in over 20% of cases . Researchers can employ KMT2D antibodies in multiple applications to investigate its role in lung cancer:

For immunohistochemical analysis, KMT2D antibodies can be used to assess protein expression in LUSC patient samples compared to normal lung tissues, as studies have shown significantly lower KMT2D expression in LUSC compared to normal lung tissue . The recommended dilution range of 1:50-1:500 for IHC applications allows for optimization depending on tissue type and fixation methods .

For mechanistic studies, Western blot analysis using KMT2D antibodies can verify knockout efficiency in experimental models, as demonstrated in studies utilizing CRISPR/Cas9 sgRNAs targeting Kmt2d in organoid models . This approach helps establish critical causative relationships between KMT2D loss and phenotypic changes in lung cancer progression.

For investigation of downstream effects, researchers can combine KMT2D expression analysis with assessment of receptor tyrosine kinase (RTK) activity, particularly EGFR and ERBB2, as KMT2D loss increases activation of these pathways . This multi-parametric approach helps identify therapeutic vulnerabilities in KMT2D-deficient tumors.

What considerations are important when using KMT2D antibodies to study B-cell development and lymphoma?

When investigating KMT2D in B-cell development and lymphoma, several important considerations should guide experimental design:

First, understand the expression pattern of KMT2D across B-cell developmental stages. Studies indicate KMT2D is expressed in all mature B cell compartments, including naïve, germinal center, and memory B cells . Co-immunofluorescence analysis using KMT2D and germinal center-specific markers like BCL6 in reactive human tonsils has confirmed KMT2D staining in nuclei across all mature B cell compartments, including germinal centers .

Second, consider the timing of KMT2D loss in experimental models. Research has demonstrated that conditional deletion of Kmt2d early during B cell development results in increased germinal center B-cells and enhanced B cell proliferation, while deletion after initiation of the germinal center reaction has different outcomes . This temporal aspect is critical when designing studies to recapitulate human disease pathogenesis.

Third, interpret results in the context of genetic cooperation. In BCL2-overexpressing mouse models that develop germinal center-derived lymphomas resembling human tumors, genetic ablation of Kmt2d leads to a further increase in tumor incidence . This indicates that KMT2D loss synergizes with other oncogenic events, which should be considered when analyzing human lymphoma samples with multiple genetic alterations.

How should researchers troubleshoot inconsistent KMT2D detection in different experimental systems?

Inconsistent detection of KMT2D across experimental systems can arise from several factors that require specific troubleshooting approaches:

For Western blot applications, KMT2D's large size (593 kDa) presents significant technical challenges . Researchers should employ low percentage (3-5%) SDS-PAGE gels or gradient gels optimized for high molecular weight proteins. Transfer efficiency should be monitored using appropriate size markers, and extended transfer times may be necessary. The recommended dilution range of 1:2000-1:16000 for WB applications allows flexibility for optimization based on specific sample types .

For detection of endogenous KMT2D in cell lines with potential mutations, consider the epitope location of your antibody. If using a C-terminal-directed antibody, samples with truncation mutations will not show signal despite expressing truncated protein . Using antibodies targeting different regions of KMT2D can help resolve such discrepancies.

For immunohistochemistry applications, antigen retrieval conditions are critical. The recommended protocol suggests TE buffer at pH 9.0, with citrate buffer pH 6.0 as an alternative . Systematic optimization of antigen retrieval methods, antibody concentration, and incubation conditions may be necessary for each tissue type.

When encountering negative results, validate antibody performance using known positive controls (HEK-293 cells for WB; human breast cancer, lymphoma, or colon cancer tissues for IHC) . Additionally, genetic confirmation through sequencing can help interpret negative immunostaining results, particularly in tumor samples where KMT2D mutations are common.

What strategies can help distinguish between wild-type and mutant KMT2D in research samples?

Distinguishing between wild-type and mutant KMT2D forms presents unique challenges that require integrated approaches:

For missense mutations, most mutant KMT2D proteins show similar expression levels to wild-type . In these cases, functional assays measuring methyltransferase activity provide more informative readouts than expression analysis alone. Researchers can assess H3K4 methylation status at KMT2D target loci as a surrogate for KMT2D function.

Genetic confirmation through DNA sequencing remains essential, particularly in primary tumor samples where multiple genetic alterations may coexist. This can be complemented with transcriptomic analysis to assess expression levels of KMT2D target genes, providing functional insights even when protein detection is challenging.

For complex samples containing mixed populations, consider single-cell approaches combining protein detection (through immunofluorescence or mass cytometry) with genetic analysis to correlate KMT2D protein status with mutational profile at the single-cell level.

How are KMT2D antibodies advancing our understanding of epigenetic dysregulation in cancer?

KMT2D antibodies have been instrumental in elucidating the epigenetic mechanisms underlying cancer development and progression. Through immunoblotting, immunohistochemistry, immunofluorescence, and chromatin immunoprecipitation approaches, these antibodies have helped establish KMT2D as a bona fide tumor suppressor in multiple cancer types, including follicular lymphoma, diffuse large B-cell lymphoma, and lung squamous cell carcinoma .

Key insights facilitated by KMT2D antibody-based research include the discovery that over 95% of KMT2D-bound chromatin in germinal center B cells is decorated by H3K4me1 and/or H3K4me3, establishing KMT2D as a non-redundant methyltransferase controlling the methylation state of numerous genomic regions in mature B cells . Furthermore, antibody-based approaches have demonstrated that KMT2D deletion can transform lung basal cell organoids to squamous cell carcinoma, highlighting its critical role in maintaining epithelial cell identity .

The integration of antibody-based detection methods with genetic, transcriptomic, and epigenomic analyses has revealed that KMT2D cooperates with other epigenetic regulators like CREBBP, and their combined haploinsufficiency accelerates lymphomagenesis through disruption of super-enhancers driving immune synapse genes . This mechanistic insight provides a potential explanation for immunotherapy resistance in certain cancers.

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.