Biotin-conjugated KMT5C antibodies are utilized in diverse experimental workflows:
ELISA: Used to quantify KMT5C protein levels in lysates or serum, paired with streptavidin-HRP for colorimetric detection .
Western Blotting: Identifies KMT5C expression in cancer cell lines (e.g., ccRCC, NSCLC) or clinical samples, validated via SDS-PAGE and chemiluminescence .
IHC: Localizes KMT5C in tissue sections, critical for studying its role in tumor progression (e.g., ccRCC) .
IF: Visualizes subcellular localization (nuclear) and interactions with chromatin modifiers .
Knockdown/Overexpression: Combined with biotin-conjugated antibodies to validate KMT5C depletion or ectopic expression in cell lines .
Protein-Protein Interactions: Used in co-immunoprecipitation (Co-IP) to study KMT5C’s binding partners (e.g., RB1 family proteins) .
Clear Cell Renal Cell Carcinoma (ccRCC):
Non-Small Cell Lung Cancer (NSCLC):
H4K20me3 and Chromatin Dynamics:
Targeted Therapy:
The following table summarizes key features of commercially available antibodies:
KMT5C (also known as SUV420H2) is a histone-lysine methyltransferase that specifically methylates monomethylated 'Lys-20' (H4K20me1) and dimethylated 'Lys-20' (H4K20me2) of histone H4 to produce dimethylated and trimethylated 'Lys-20' (H4K20me3) respectively. This enzyme plays a critical role in transcriptional regulation and genome integrity maintenance. Its importance in epigenetic research stems from its involvement in chromatin organization and gene expression control, making it a valuable target for studies on cellular differentiation, cancer biology, and response to therapy .
Biotin-conjugated KMT5C antibodies offer several methodological advantages:
Enhanced sensitivity through signal amplification via the strong biotin-streptavidin interaction
Greater flexibility in experimental design with compatibility across multiple detection systems
Reduced background in multi-color immunofluorescence experiments
Improved stability during long-term storage compared to directly labeled fluorescent antibodies
Versatility in applications including ELISA, immunofluorescence, and chromatin immunoprecipitation followed by sequencing (ChIP-seq)
For optimal results with biotin-conjugated KMT5C antibodies in immunofluorescence:
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.3% Triton X-100 in PBS for 10 minutes
Block with 5% normal serum (from the same species as the secondary antibody) with 0.1% Triton X-100 for 1 hour
Incubate with biotin-conjugated KMT5C antibody at 1:50 to 1:100 dilution overnight at 4°C
Detect using streptavidin-conjugated fluorophores (FITC, Texas Red, etc.)
Use DAPI for nuclear counterstaining
This protocol has been validated in multiple cell types including embryonic cells as demonstrated in published immunofluorescence analyses .
Quantifying H4K20me3 levels requires a rigorous approach:
Western Blot Quantification:
Use biotin-conjugated KMT5C antibody followed by streptavidin-HRP
Include β-actin or total H4 as loading controls
Perform densitometry analysis using ImageJ or similar software
Normalize H4K20me3 signal to loading control
Immunofluorescence Quantification:
Implement parallel staining of WT and mutant samples
Use identical acquisition parameters (exposure time, gain)
Quantify nuclear signal intensity using CellProfiler or similar software
Normalize to nuclear area or DAPI intensity
Analyze ≥100 cells per condition
In-cell Western Analysis:
Plate equal cell numbers in 96-well format
Perform fixation and antibody incubation in-plate
Use GAPDH as endogenous control
Scan with appropriate imaging system
Calculate relative H4K20me3 levels
This multi-method approach enables robust quantification across different genetic backgrounds, as demonstrated in studies comparing H4K20me3 levels in KMT5C wild type and mutant cell lines .
For successful ChIP-qPCR with biotin-conjugated KMT5C antibodies:
Chromatin Preparation:
Crosslink cells with 1% formaldehyde for exactly 10 minutes
Quench with 125mM glycine
Sonicate to achieve fragments of 200-500bp (verify by gel electrophoresis)
Immunoprecipitation:
Pre-clear chromatin with protein G beads
Incubate chromatin with biotin-conjugated KMT5C antibody overnight at 4°C
Capture antibody-chromatin complexes with streptavidin-coated magnetic beads
Include IgG control for background determination
qPCR Design:
Design primers targeting regions of interest, such as promoters or enhancers
Include primers for known H4K20me3-enriched regions as positive controls
Include primers for regions devoid of H4K20me3 as negative controls
Calculate fold enrichment as the ratio of immunoprecipitated DNA to input DNA, relative to IgG control
Data Analysis:
Present data as fold enrichment of regions pulled down by H4K20me3 antibody relative to IgG
Compare enrichment between experimental conditions (e.g., WT vs. mutant)
Apply appropriate statistical tests (one-way ANOVA with Dunnett's multiple comparison test)
This approach has been successfully applied to identify H4K20me3-enriched regions in regulatory elements, such as those upstream of MET gene .
Loss of KMT5C function has been demonstrated to confer resistance to EGFR tyrosine kinase inhibitors (TKIs) including erlotinib, gefitinib, afatinib, and osimertinib in non-small cell lung cancer (NSCLC) cell lines. Biotin-conjugated KMT5C antibodies can be instrumental in investigating this mechanism through:
Monitoring H4K20me3 Levels:
Compare H4K20me3 levels in sensitive versus resistant cell lines using Western blot and immunofluorescence
Correlate H4K20me3 levels with GI50 values for various EGFR inhibitors
Track changes in H4K20me3 during acquired resistance development
ChIP-seq Analysis:
Identify genome-wide changes in H4K20me3 distribution following KMT5C loss
Uncover potential regulatory elements affected by KMT5C deficiency
Correlate with expression changes in resistance-associated genes
Target Gene Identification:
Studies have shown that loss of KMT5C leads to upregulation of MET and MKK3, contributing to EGFR-TKI resistance
H4K20me3 marks on regulatory elements like LINC01510 can be detected using biotin-conjugated KMT5C antibodies
The methylation status of these regions correlates with gene expression changes
This approach provides insights into the epigenetic mechanisms of drug resistance, as exemplified by studies showing that KMT5C transcript levels are downregulated in tumors post-treatment with osimertinib in NSCLC patients .
An effective experimental design would include:
Baseline Analysis:
Measure KMT5C expression and H4K20me3 levels across a panel of cancer cell lines with varying drug sensitivities
Use biotin-conjugated KMT5C antibody for immunofluorescence and Western blot analysis
Correlate findings with drug sensitivity data (GI50 values)
Genetic Manipulation:
Generate KMT5C knockout or knockdown models using CRISPR-Cas9 or siRNA
Create KMT5C overexpression models in sensitive cell lines
Validate alterations in H4K20me3 levels using biotin-conjugated antibodies
Assess changes in drug sensitivity using dose-response assays
Pharmacological Inhibition:
Treat cells with KMT5B/C inhibitors (e.g., A-196)
Confirm reduction in H4K20me3 levels
Evaluate changes in drug sensitivity
Compare effects with genetic manipulation
Mechanistic Investigation:
Perform RNA-seq to identify differentially expressed genes
Use ChIP-seq with biotin-conjugated KMT5C antibodies to map H4K20me3 distribution
Identify candidate genes regulated by KMT5C
Validate candidates through targeted knockdown/overexpression
Clinical Correlation:
Analyze KMT5C expression and H4K20me3 levels in patient samples pre- and post-treatment
Correlate findings with treatment response and survival outcomes
This comprehensive approach has been successfully employed to demonstrate that KMT5C loss induces MET and MKK3 overexpression in EGFR-mutant cell lines, contributing to therapy resistance .
Source of Background | Mitigation Strategy | Implementation Details |
---|---|---|
Endogenous biotin | Pre-block with avidin/streptavidin | Incubate samples with avidin (10 μg/ml) followed by biotin (50 μg/ml) before antibody addition |
Non-specific binding | Optimize blocking conditions | Use 5% BSA or 10% normal serum from secondary antibody species; add 0.1-0.3% Triton X-100 |
Inadequate washing | Increase wash stringency | Perform 5-6 washes with PBS containing 0.1% Tween-20; extend wash duration to 10 minutes each |
Cross-reactivity | Use proper controls | Include no-primary antibody control; use isotype control; test on KMT5C knockout samples if available |
Excessive antibody concentration | Optimize antibody dilution | Perform titration experiments (1:25, 1:50, 1:100, 1:200); select concentration with optimal signal-to-noise ratio |
Fixation artifacts | Adjust fixation protocol | Try different fixatives (4% PFA, methanol, acetone) and fixation times (10, 15, 20 minutes) |
Autofluorescence | Add quenching step | Treat samples with 50 mM NH₄Cl for 10 minutes after fixation to reduce autofluorescence |
These approaches have been validated across multiple cellular systems and applications including immunofluorescence in embryonic cells and Chlamydomonas cells .
Optimization strategies for detecting low-abundance H4K20me3 marks include:
Increase Cell Input:
Start with at least 10⁷ cells for each immunoprecipitation
Scale buffer volumes accordingly while maintaining antibody concentration
Crosslinking Optimization:
Dual crosslinking approach: 2 mM disuccinimidyl glutarate (DSG) for 45 minutes followed by 1% formaldehyde for 10 minutes
This preserves protein-protein interactions more effectively than formaldehyde alone
Sonication Refinement:
Optimize sonication conditions to achieve consistent 200-300bp fragments
Verify fragment size by agarose gel electrophoresis
Consider enzymatic fragmentation (MNase) as an alternative
Immunoprecipitation Enhancement:
Extend incubation time with antibody to 16-20 hours at 4°C with gentle rotation
Use a sequential ChIP approach for highly specific enrichment
Add carrier proteins (e.g., sheared salmon sperm DNA) to reduce non-specific binding
Signal Amplification:
Implement biotin-streptavidin signal amplification systems
Consider using specialized ChIP-grade streptavidin beads
Reduce bead volume to concentrate the signal
qPCR Sensitivity:
Design highly efficient primers (90-110% efficiency)
Increase PCR cycles (up to 45) for low-abundance targets
Use nested PCR approach for extremely low signals
This optimized approach has been successfully applied to detect H4K20me3 marks on chromatin regions with varying enrichment levels, as demonstrated in studies examining the regulation of genes like MET by KMT5C .
KMT5C has distinct functions compared to other histone methyltransferases:
Feature | KMT5C (SUV420H2) | KMT5B (SUV420H1) | Other HMTs (e.g., EZH2, G9a) |
---|---|---|---|
Substrate Specificity | H4K20me1/me2 → H4K20me2/me3 | H4K20me0/me1 → H4K20me1/me2 | Different lysine residues (H3K27, H3K9, etc.) |
Genomic Distribution | Mainly heterochromatin | Broader distribution | Variable patterns |
Functional Role | Genome stability, silencing | Cell cycle regulation | Context-dependent |
Protein Size | 462 aa, 52.1 kDa | Larger | Variable |
Cellular Location | Nucleus | Nucleus | Mostly nuclear |
Experimental approaches to distinguish these differences using biotin-conjugated antibodies:
Sequential ChIP (Re-ChIP):
First IP with biotin-conjugated KMT5C antibody
Elute complexes and perform second IP with antibodies against other HMTs
Analyze co-occupancy of different HMTs at specific genomic loci
Co-Immunoprecipitation:
Use biotin-conjugated KMT5C antibodies for pull-down
Probe for interaction partners
Compare interactome with other HMTs
Histone Modification Profiling:
Perform Western blots with antibodies against various histone marks (H4K20me1/2/3, H3K9me3, H3K27me3)
Compare modification patterns in WT, KMT5C-mutant, and other HMT-mutant cells
Assess cross-talk between different histone modifications
Inhibitor Studies:
Compare effects of KMT5B/C-specific inhibitors (e.g., A-196) with inhibitors of other HMTs
Measure changes in global histone modification levels
Assess differential effects on cellular phenotypes and gene expression
These approaches have been instrumental in distinguishing the specific roles of KMT5C in processes like EGFR inhibitor resistance, which appears to be uniquely regulated by this particular HMT through specific target genes .
For effective multiplexing of biotin-conjugated KMT5C antibodies with other epigenetic markers:
Panel Design for Mass Cytometry (CyTOF):
Use metal-conjugated streptavidin (e.g., Sm-149-streptavidin) to detect biotin-conjugated KMT5C antibodies
Select metals with minimal signal overlap for other epigenetic markers
Include both activating (H3K4me3, H3K27ac) and repressive (H3K9me3, H3K27me3) marks
Add cell cycle markers (e.g., Ki-67) for cell state context
Example panel design:
Target | Metal Tag | Antibody Type | Function |
---|---|---|---|
KMT5C | Sm-149 (via streptavidin) | Biotin-conjugated primary | H4K20 methyltransferase |
H4K20me3 | Nd-142 | Direct metal-conjugated | KMT5C product |
H3K9me3 | Eu-151 | Direct metal-conjugated | Heterochromatin mark |
H3K27me3 | Gd-160 | Direct metal-conjugated | Polycomb-mediated silencing |
H3K4me3 | Yb-172 | Direct metal-conjugated | Active promoters |
Ki-67 | Ir-191 | Direct metal-conjugated | Proliferation marker |
Multiplex Immunofluorescence:
Use far-red fluorophore-conjugated streptavidin to detect biotin-conjugated KMT5C antibody
Apply tyramide signal amplification (TSA) for low-abundance targets
Implement sequential antibody labeling and stripping protocol:
a. Apply first primary antibody
b. Detect with appropriate secondary
c. Fix signal with 4% PFA
d. Strip remaining antibodies with glycine-SDS buffer (pH 2.5)
e. Repeat for each target
Use spectral unmixing algorithms to separate overlapping fluorophores
Imaging Mass Cytometry:
Prepare FFPE or frozen tissue sections
Apply biotin-conjugated KMT5C antibody followed by metal-tagged streptavidin
Include antibodies against cell type-specific markers
Analyze spatial relationships between KMT5C, its histone mark, and other epigenetic features
Implement neighborhood analysis to identify cellular microenvironments
CODEX (CO-Detection by indEXing) Approach:
Conjugate biotin-KMT5C antibody with unique DNA barcodes
Apply all barcoded antibodies simultaneously
Detect sequentially through repeated cycles of complementary fluorescent oligo hybridization
This enables visualization of 40+ targets on the same sample
These multiplexing strategies have been applied successfully in epigenetic research to understand the complex interplay between different histone modifications and their regulatory enzymes .
Biotin-conjugated KMT5C antibodies offer valuable tools for investigating H4K20 methylation in metabolic regulation:
Adipocyte Differentiation Studies:
Track changes in H4K20me3 levels during adipocyte differentiation
Perform ChIP-seq with biotin-conjugated KMT5C antibodies at different time points
Correlate H4K20me3 distribution with expression of adipogenic genes
Compare patterns in brown versus white adipose tissue
Metabolic Phenotyping:
Research has shown that Suv420h histone methyltransferases (including KMT5C/SUV420H2) regulate PPAR-γ and energy metabolism
Use biotin-conjugated KMT5C antibodies to investigate H4K20me3 marks on metabolic gene promoters
Correlate with physiological parameters in metabolic disease models
Tissue-Specific Analysis:
Perform immunohistochemistry on adipose tissue sections using biotin-conjugated KMT5C antibodies
Quantify nuclear H4K20me3 levels in different cell populations
Compare normal versus metabolically challenged tissues (high-fat diet, diabetes models)
Response to Environmental Stimuli:
Studies have demonstrated that Suv420h enzymes respond to environmental stimuli
Investigate H4K20me3 pattern changes following cold exposure, caloric restriction, or exercise
Correlate with mitochondrial function and brown adipose tissue activation
Intervention Studies:
Use A-196 inhibitor to modulate KMT5B/C activity in metabolic tissues
Monitor subsequent changes in H4K20me3 patterns and gene expression
Assess metabolic parameters (oxygen consumption, energy expenditure)
This research approach can reveal how KMT5C-mediated H4K20 methylation contributes to metabolic regulation, as suggested by studies showing that Suv420h histone methyltransferases regulate PPAR-γ and energy metabolism .
When studying H4K20me3 dynamics during cell cycle progression with biotin-conjugated KMT5C antibodies, researchers should consider:
Cell Synchronization:
Implement precise synchronization methods (double thymidine block, nocodazole arrest, etc.)
Verify synchronization efficiency by flow cytometry
Collect cells at defined time points throughout the cell cycle
Fixation Timing and Method:
Rapid fixation is critical to capture transient states
Use formaldehyde fixation (1-4%) for 10 minutes at room temperature
Avoid methanol fixation which can extract histones
Dual Labeling Strategy:
Combine biotin-conjugated KMT5C antibody with cell cycle markers:
G1: Cyclin D1 or CDT1
S: EdU incorporation or PCNA
G2: Cyclin B1
M: Phospho-histone H3 (Ser10)
Use different fluorophores to distinguish cell cycle stages
Quantitative Analysis:
Implement high-content imaging to analyze thousands of cells
Classify cells by cycle phase based on marker expression
Quantify nuclear H4K20me3 intensity for each phase
Calculate relative changes in H4K20me3 throughout the cycle
Dynamic ChIP Studies:
Perform ChIP-seq with biotin-conjugated KMT5C antibodies at different cell cycle phases
Compare H4K20me3 distribution patterns across the genome
Identify cell cycle-regulated H4K20me3 domains
Live-Cell Imaging Considerations:
For real-time dynamics, use cell lines expressing fluorescently tagged histones
Complement with fixed-cell time course using biotin-conjugated KMT5C antibodies
Correct for potential cell cycle variability through single-cell analysis
Technical Controls:
Include IgG controls for each cell cycle phase
Use KMT5C knockout cells as negative controls
Include total H4 staining to normalize for histone content changes
This methodological approach enables precise characterization of H4K20me3 dynamics throughout the cell cycle, providing insights into the temporal regulation of chromatin structure and its impact on genome stability and gene expression .