Experimental evidence demonstrates KMT5A's tumor-promoting effects through multiple pathways:
Silencing KMT5A induces G1/S phase arrest in papillary thyroid cancer cells (P < 0.05)
Maintains DNA replication integrity through H4K20me1 deposition
Metabolic Reprogramming
In thyroid cancer models, KMT5A knockdown:
Downregulates lipid metabolism genes:
Metastatic Promotion
KMT5A enhances epithelial-mesenchymal transition (EMT) in clear cell renal carcinoma:
Recent studies identify KMT5A as a promising intervention point:
Pharmacological inhibition decreases cell viability in prostate cancer models (IC50 = 3.2 μM)
Combined targeting with DNA-damaging agents shows synergistic effects
Current research focuses on developing small-molecule inhibitors targeting the SET domain while preserving physiological H4K20 methylation patterns. Challenges remain in achieving tumor-specific activity due to KMT5A's essential role in normal cell cycle progression .
Lysine Methyltransferase 5A (KMT5A), also known as SETD8 or SET8, is an enzyme that plays a crucial role in regulating various cellular processes. It catalyzes the addition of a methyl group to lysine residues on both histone and non-histone proteins. This methylation activity is essential for maintaining the structural integrity of DNA, especially during cell division. KMT5A is involved in controlling gene expression, particularly in silencing genes during specific cell cycle phases and mitosis. It acts as a safeguard against premature cellular aging by regulating metabolic processes through chromatin modifications. KMT5A directly influences the tumor suppressor protein p53 by mediating its monomethylation, which in turn suppresses the expression of genes regulated by p53. Loss of KMT5A can disrupt normal cellular functions, leading to increased nucleolar activity and alterations in mitochondrial metabolism regulated by the retinoblastoma protein.
This product consists of the recombinant human KMT5A protein, specifically amino acids 195 to 352, produced in Sf9 insect cells using baculovirus expression system. It is a single polypeptide chain with a 7 amino acid Histidine tag at the C-terminus to facilitate purification. The protein has a molecular weight of 18.9 kDa and appears as a band between 18-28 kDa on SDS-PAGE under reducing conditions due to glycosylation. The protein has been purified using proprietary chromatographic techniques to ensure high purity.
The KMT5A protein is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0, 5mM DTT (reducing agent), 0.2M NaCl (salt), 1mM EDTA (chelating agent), and 50% glycerol (cryoprotectant). The protein concentration is 0.25mg/ml.
The purity of the KMT5A protein is greater than 85%, as determined by SDS-PAGE analysis.
KMT5A, PR-Set7, SET07, SET8, SETD8, H4-K20-HMTase KMT5A.
MKAELQSEER KRIDELIESG KEEGMKIDLI DGKGRGVIAT KQFSRGDFVV EYHGDLIEIT DAKKREALYA QDPSTGCYMY YFQYLSKTYC VDATRETNRL GRLINHSKCG NCQTKLHDID GVPHLILIAS RDIAAGEELL YDYGDRSKAS IEAHPWLKHH HHHHH.
Human KMT5A is primarily a lysine methyltransferase that catalyzes mono-methylation of histone H4 at lysine 20 (H4K20me1). This epigenetic modification plays critical roles in chromatin organization, DNA replication, cell cycle progression, and genome stability. The enzyme contains a highly conserved catalytic SET domain that binds to S-adenosyl methionine (SAM) as a methyl donor and transfers the methyl group to the ε-amino group of lysine residues. Structural studies have demonstrated that the SET domain forms a distinct binding pocket that accommodates both the substrate and cofactor in a specific orientation to facilitate methyl transfer. Mutations disrupting SAM binding, as shown through molecular dynamics simulations, can eliminate methyltransferase activity while minimally impacting H4 peptide binding .
Human KMT5A consists of distinct functional domains with specific roles:
The C-terminal SET domain (catalytic domain) - Responsible for methyltransferase activity and is 57% identical to the Drosophila Set8 SET domain
The N-terminal domain - Contains regulatory regions involved in protein-protein interactions and cellular localization
Research using chimeric proteins between Drosophila Set8 and human KMT5A has demonstrated that while the SET domains are functionally interchangeable across species, the N-terminal region of Set8 has specific functions in eye development that cannot be complemented by the human KMT5A N-terminus . This suggests that while the catalytic function is evolutionarily conserved, regulatory domains may have species-specific roles. Additionally, the N-terminal region may be responsible for targeting KMT5A to specific genomic loci through interactions with various transcription factors and chromatin-associated proteins.
KMT5A demonstrates significant evolutionary conservation across various species, particularly in the catalytic SET domain. Functional studies have shown that human KMT5A can rescue developmental defects in Drosophila Set8 null mutants, indicating the fundamental conservation of its enzymatic function . The SET domains of human KMT5A and Drosophila Set8 share 57% sequence identity, and experimental evidence demonstrates they are functionally interchangeable .
Several experimental systems have proven valuable for investigating KMT5A function:
Drosophila models: Drosophila has been used effectively to study KMT5A/Set8 function through the creation of null mutants and transgenic rescue experiments. This model allows assessment of developmental functions and provides a well-characterized system for studying eye development, which appears particularly sensitive to KMT5A/Set8 perturbations .
Mammalian cell lines: Human and mouse cell lines with KMT5A knockdown, knockout, or overexpression provide insights into cell cycle regulation, DNA damage responses, and chromatin structure alterations.
Mouse models: Conditional knockout mice allow tissue-specific analysis of KMT5A function during development and in adult tissues.
Biochemical assays: In vitro methyltransferase assays using recombinant KMT5A protein and synthetic or native substrates enable detailed mechanistic studies of enzyme kinetics and substrate specificity.
Structural biology approaches: X-ray crystallography and cryo-EM have been used to determine the three-dimensional structure of KMT5A in complex with substrates and inhibitors, facilitating structure-based drug design.
When selecting an experimental system, researchers should consider that while the catalytic function of KMT5A is conserved, certain regulatory aspects may differ between species, particularly those involving the N-terminal domain .
Distinguishing between H4K20 methylation-dependent and -independent functions of KMT5A requires sophisticated experimental designs:
Catalytically dead mutants: Generate KMT5A variants with point mutations in the SET domain that abolish catalytic activity but preserve protein structure. The R265G mutation in KMT5A has been shown to eliminate catalytic activity and fails to support embryonic development in mice, suggesting that catalytic activity is essential for proper development .
Substrate-specific mutations: Create H4K20 mutants (K20A or K20R) that cannot be methylated and assess whether specific phenotypes can be rescued by wild-type H4K20 but not the mutant versions.
Non-histone substrate identification: Use immunoprecipitation followed by mass spectrometry to identify non-histone KMT5A substrates, then confirm methylation sites using in vitro methyltransferase assays.
Domain-specific rescue experiments: As demonstrated in Drosophila, chimeric constructs with swapped domains between species can reveal domain-specific functions that are independent of catalytic activity .
Temporal analysis: Use inducible systems to modulate KMT5A activity at different developmental stages or cell cycle phases to distinguish between immediate and delayed effects, which may indicate direct versus indirect mechanisms.
Interestingly, research in Drosophila suggests that the essential developmental function of Set8 may be either non-catalytic or largely independent of H4K20 methylation activity , highlighting the importance of these experimental approaches.
Developing specific inhibitors for KMT5A faces several significant challenges:
Selectivity over related methyltransferases: The SET domain structure shares similarities with other lysine methyltransferases, making it difficult to achieve selective inhibition of KMT5A without affecting related enzymes like KMT5B or other SET domain-containing proteins.
Substrate binding site complexity: The substrate binding pocket accommodates both the peptide substrate and the cofactor SAM, creating a complex binding interface that is challenging to target selectively.
Protein-protein interaction surfaces: KMT5A functions within multi-protein complexes, and disrupting specific protein-protein interactions might offer greater selectivity than targeting the catalytic site but presents additional design challenges.
Cellular permeability: Many potential inhibitors that bind to the SAM pocket are highly polar and struggle to cross cell membranes, limiting their cellular efficacy.
Functional redundancy: Potential compensatory mechanisms involving related enzymes may limit the efficacy of KMT5A-specific inhibitors.
Structural studies and molecular dynamics simulations examining SAM binding and H4 peptide interactions have provided valuable insights that can guide inhibitor development . Researchers are currently exploring allosteric sites and unique structural features of KMT5A to overcome these challenges and develop more selective inhibitors.
KMT5A plays essential roles in embryonic development across species:
Developmental necessity: KMT5A/Set8 null mutations are lethal in both mice and Drosophila, demonstrating its fundamental requirement for organismal development .
Cell cycle regulation: KMT5A regulates cell cycle progression, particularly during G1/S transition and mitosis, which is critical for proper tissue development.
Tissue-specific functions: In Drosophila, Set8 has a specific role in eye development mediated by its N-terminal domain . This suggests KMT5A may have tissue-specific functions during development.
Growth regulation: In both mice and Drosophila, loss of Set8/KMT5A leads to smaller larval tissues and growth defects .
Genomic stability: KMT5A helps maintain genomic integrity during development, with mutant cells accumulating DNA damage that can impair proper development .
The observation that human KMT5A can rescue most but not all developmental defects in Drosophila Set8 null mutants indicates evolutionary conservation of core developmental functions with some species-specific adaptations . The catalytic activity of KMT5A appears essential for proper development, as demonstrated by the failure of a catalytically inactive KMT5A R265G mutant to support mouse embryonic development .
KMT5A and KMT5B are related lysine methyltransferases with distinct developmental roles:
Substrate specificity: While both enzymes can catalyze H4K20 methylation, they may have different preferences for non-histone substrates.
Developmental consequences: Pathogenic variants in KMT5B are associated with global developmental delay, macrocephaly, autism, and congenital anomalies (OMIM #617788) , while KMT5A mutations appear to have different developmental consequences.
Expression patterns: KMT5B shows peak human brain mRNA expression before 20 weeks after conception, falling after birth to a steady state , whereas KMT5A may have different temporal expression patterns.
Neurological impact: KMT5B haploinsufficiency significantly affects neurodevelopment, with RNA sequencing of patient lymphoblasts and KMT5B haploinsufficient mouse brains showing differential expression of genes involved in nervous system development and axon guidance signaling . KMT5A's neurological functions may be distinct.
Genetic variation tolerance: Population-level genetic data shows differing tolerance to loss-of-function variations between these related enzymes.
Understanding the distinct roles of these related enzymes provides important context for interpreting experimental results and developing targeted therapeutic approaches for associated developmental disorders.
Research indicates several cellular processes are particularly sensitive to KMT5A dysregulation:
DNA replication: KMT5A is required for proper DNA replication timing and origin firing; its disruption leads to replication stress and S-phase defects.
Cell cycle progression: KMT5A levels fluctuate throughout the cell cycle, with dysregulation causing cell cycle checkpoint abnormalities and proliferation defects .
DNA damage response: Loss of KMT5A results in increased DNA damage and impaired DNA repair mechanisms .
Chromatin compaction: KMT5A-mediated H4K20 methylation influences higher-order chromatin structure; its disruption affects nuclear architecture and chromosome condensation .
Transcriptional regulation: KMT5A influences gene expression patterns, with its dysregulation potentially affecting developmental gene expression programs.
In Drosophila Set8 mutants, defects in eye development suggest that neuronal differentiation and organized tissue formation are particularly sensitive to proper KMT5A function . The accumulation of DNA damage in KMT5A-deficient cells indicates that genome stability mechanisms are especially vulnerable to KMT5A dysregulation .
Several complementary approaches can effectively detect and quantify KMT5A-mediated methylation:
Antibody-based detection:
Western blotting with methylation-specific antibodies (anti-H4K20me1)
Immunofluorescence for cellular localization of methylation marks
Chromatin immunoprecipitation (ChIP) followed by sequencing (ChIP-seq) to map genomic distribution of H4K20me1
Mass spectrometry-based approaches:
Targeted mass spectrometry for absolute quantification of methylation levels
Proteome-wide analysis to identify non-histone substrates
SILAC (Stable Isotope Labeling with Amino acids in Cell culture) for comparative quantification across conditions
In vitro methyltransferase assays:
Radiometric assays using ³H-SAM to measure methyl transfer
Fluorescence-based assays for high-throughput screening
Enzyme kinetics studies to determine substrate preferences
Genetic approaches:
Structural analysis:
When selecting detection methods, researchers should consider the specificity of antibodies for different methylation states (mono-, di-, or tri-methylation) and the potential cross-reactivity with related modifications.
Effective modeling of KMT5A function across experimental systems requires tailored approaches:
Cell culture models:
Inducible knockdown/knockout systems (shRNA, CRISPR-Cas9)
Rescue experiments with wild-type or mutant KMT5A
Cell cycle synchronization to study phase-specific functions
Primary cell cultures to examine tissue-specific effects
Drosophila models:
Mouse models:
Conditional knockout approaches using tissue-specific Cre recombinase
Knock-in of catalytically inactive mutants
Embryonic analysis at different developmental stages
Behavioral assessment for neurological phenotypes
In vitro biochemical systems:
Reconstituted nucleosome substrates
Recombinant KMT5A protein with defined mutations
High-throughput enzyme assays for inhibitor screening
Computational approaches:
The complementary use of these systems has revealed that human KMT5A can functionally substitute for Drosophila Set8 in most developmental contexts, with the exception of certain eye development functions . This approach of cross-species complementation provides valuable insights into both conserved and divergent aspects of KMT5A function.
Accurate measurement of KMT5A enzymatic activity faces several technical challenges:
Substrate complexity:
Nucleosomal versus free histone substrates produce different activity profiles
Physiologically relevant non-histone substrates are often unknown or difficult to produce
Post-translational modifications on substrates can influence KMT5A activity
Enzyme stability and purity:
Full-length KMT5A can be difficult to express and purify in active form
Truncated constructs may not recapitulate all regulatory features
Co-factors or interacting proteins may be required for optimal activity
Assay limitations:
Radiometric assays offer high sensitivity but have safety and disposal concerns
Antibody-based detection depends on specificity and can be affected by epitope masking
Coupling enzyme assays may be influenced by the coupling reaction
Physiological conditions:
In vitro conditions may not replicate the cellular environment (pH, salt, crowding)
Chromatin context and higher-order structure affect accessibility to substrates
Cell cycle-dependent regulation is difficult to model in vitro
Detection specificity:
Distinguishing KMT5A activity from other H4K20 methyltransferases
Differentiating between mono-, di-, and tri-methylation states
Identifying specific non-histone methylation events
To address these challenges, researchers often employ multiple complementary approaches, including in vitro assays with defined components, cellular assays with genetic manipulations (such as the chimeric constructs used in Drosophila studies ), and advanced analytical techniques like mass spectrometry for unambiguous identification of methylation sites.
KMT5A and KMT5B have distinct but related roles in human development:
Developmental disorders:
Neurological functions:
Growth regulation:
Substrate specificity:
Both enzymes catalyze H4K20 methylation but may have different preferences for non-histone substrates
Different genomic distributions may lead to regulation of distinct gene sets
Cellular functions:
This comparative understanding is essential for interpreting phenotypes in model systems and for developing targeted therapeutic approaches for developmental disorders associated with these enzymes.
Distinguishing between the activities of KMT5A and other H4K20 methyltransferases requires specialized methodological approaches:
Genetic manipulation strategies:
Single and combined knockouts/knockdowns to identify unique and redundant functions
Rescue experiments with enzyme-specific mutations
CRISPR-Cas9 base editing to introduce catalytic mutations at endogenous loci
Biochemical discrimination:
In vitro assays with recombinant enzymes and defined substrates
Enzyme kinetics analysis to identify differences in substrate preferences
Development of selective inhibitors based on structural differences
Temporal and spatial analysis:
Cell cycle-specific activity measurements (KMT5A peaks in G2/M)
Tissue-specific expression analysis
Developmental stage-specific requirements
Methylation state specificity:
KMT5A primarily catalyzes mono-methylation of H4K20
KMT5B and KMT5C can catalyze di- and tri-methylation
Mass spectrometry to quantify different methylation states
Substrate profiling:
Proteome-wide identification of non-histone substrates
ChIP-seq to map genomic distributions of each enzyme
Motif analysis to identify sequence preferences
Chimeric protein approaches, such as those used in Drosophila studies with human KMT5A and Drosophila Set8 , can also help identify which domains contribute to enzyme-specific functions and interactions.
Analyzing the interplay between KMT5A and other epigenetic regulators requires integrated approaches:
Protein-protein interaction studies:
Affinity purification-mass spectrometry to identify interacting partners
Proximity labeling techniques (BioID, APEX) to capture transient interactions
Co-immunoprecipitation to confirm direct interactions
Yeast two-hybrid or mammalian two-hybrid screening for binary interactions
Chromatin landscape analysis:
Sequential ChIP (Re-ChIP) to identify co-occupancy of multiple factors
ChIP-seq combined with ATAC-seq to correlate with chromatin accessibility
Integration of multiple histone modification datasets to identify patterns
CUT&RUN or CUT&Tag for higher resolution mapping
Functional genomics approaches:
CRISPR screens targeting multiple epigenetic regulators
Synthetic lethal/viable screens to identify genetic interactions
Combinatorial knockdown/knockout experiments
Epistasis analysis to determine hierarchical relationships
Multi-omics integration:
Correlation of H4K20me1 profiles with other epigenetic marks
Integration of methylation data with transcriptomics
Pathway analysis to identify coordinated regulation
Systems biology modeling of epigenetic networks
Developmental transitions:
Analysis during cellular differentiation
Embryonic development timepoints
Cell cycle progression studies
Stress response dynamics
RNA-seq analyses from KMT5B patient lymphoblasts have revealed differential regulation of genes important for neurodevelopment , and similar approaches can be applied to study KMT5A's regulatory network and its intersection with other epigenetic modifiers.
KMT5A is unique in its ability to monomethylate lysine 20 on histone H4 (H4K20me1), a modification that is essential for various cellular processes . The enzyme uses S-adenosyl methionine (SAM) as a methyl group donor to transfer a methyl group to the ε-amine group of the lysine residue . This methylation increases the hydrophobic and basic nature of the lysine residue, allowing other proteins to recognize and bind to the methylated lysine .
The activity of KMT5A is implicated in several critical cellular functions, including:
Given its central role in essential cellular processes, dysregulation of KMT5A activity has been linked to various diseases, including cancer. Overexpression or mutations in the KMT5A gene can lead to aberrant cell proliferation and tumorigenesis . As a result, KMT5A is considered a potential target for therapeutic intervention in cancer treatment .