The KDM5C antibody is a polyclonal rabbit immunoglobulin (IgG) designed to detect the lysine-specific demethylase 5C (KDM5C) protein. KDM5C belongs to the JARID1 family of histone demethylases, which removes methyl groups from lysine residues on histone H3, specifically targeting H3K4me3 (trimethylation of lysine 4 on histone H3) . This enzymatic activity regulates chromatin structure and gene transcription, playing critical roles in development, cell differentiation, and disease states such as cancer and intellectual disability .
The KDM5C antibody is validated for use in:
Citations: (Proteintech product specifications).
Mutations in the KDM5C gene, detected using this antibody, have been linked to X-linked intellectual disability (XLID). Truncated KDM5C proteins were observed in patient fibroblasts, with reduced mRNA stability due to nonsense-mediated decay (NMD) . The antibody confirmed altered protein expression in XLID cases, highlighting its utility in diagnostic and mechanistic studies .
In oncology, KDM5C alterations correlate with enhanced tumor immunogenicity and anti-tumor immune signatures. The antibody has been employed to study KDM5C’s role in modulating immune checkpoints (e.g., PD-1, PD-L1) and its association with tumor mutational burden (TMB) . KDM5C-altered tumors show increased CD8+ T-cell infiltration, suggesting potential as a biomarker for immune checkpoint inhibitor (ICI) therapy .
KDM5C exhibits context-dependent oncogenic/tumor-suppressive functions. In breast and renal cancers, it suppresses enhancer activation via chromatin remodeling, functioning as a tumor suppressor . Conversely, it promotes oncogenesis in other contexts by recruiting chromatin modifiers (e.g., p300) to enhancers .
Wikipedia contributors. (2007). KDM5C. Retrieved from https://en.wikipedia.org/wiki/KDM5C
Schenck et al. (2015). Mutations in the intellectual disability gene KDM5C reduce protein stability. Scientific Reports. https://doi.org/10.1038/srep4406297
Zhang et al. (2021). Predictive Value of KDM5C Alterations for Immune Checkpoint Inhibitors. Frontiers in Immunology. https://doi.org/10.3389/fimmu.2021.664847
Li et al. (2022). Regulation of KDM5C stability and enhancer reprogramming in breast cancer. Cell Death & Disease. https://doi.org/10.1038/s41419-022-05296-5
Proteintech. (2025). KDM5C Antibody (14426-1-AP). Retrieved from https://www.ptglab.com/products/KDM5C-Antibody-14426-1-AP.htm
KDM5C antibodies are instrumental in multiple research applications crucial for epigenetic studies:
Western Blot (WB): For detecting KDM5C protein, typically observed at 171-180 kDa molecular weight . Most validated KDM5C antibodies are recommended at dilutions of 1:1000-1:5000 for WB applications .
Immunofluorescence (IF)/Immunocytochemistry (ICC): For visualizing KDM5C localization primarily in cell nuclei, with recommended dilutions of 1:50-1:500 .
Chromatin Immunoprecipitation (ChIP): For analyzing KDM5C binding sites throughout the genome, which has revealed that KDM5C demonstrates a strong preference for promoter regions (55% of all KDM5C-enriched sequences) .
Immunoprecipitation (IP): For studying KDM5C protein interactions and complex formation .
The choice of application should be guided by the specific research question and experimental system being investigated.
Proper storage and handling are critical for maintaining antibody functionality:
| Storage Condition | Duration | Special Considerations |
|---|---|---|
| -20°C | One year | Primary recommendation for long-term storage |
| 4°C | Up to one month | For frequent use and short-term storage |
Handling recommendations:
Avoid repeated freeze-thaw cycles that can degrade antibody quality
Store in aliquots if frequent use is anticipated
Most KDM5C antibodies are supplied in buffer containing PBS with 0.02% sodium azide and 50% glycerol at pH 7.3
Allow antibody to equilibrate to room temperature before opening the vial
Briefly centrifuge before use to collect all solution at the bottom of the tube
For 20μl sizes, note that some products contain 0.1% BSA in their formulation , which may impact certain applications.
Rigorous validation is essential for reliable research outcomes:
Reactivity verification: Confirm reactivity with your species of interest. Many KDM5C antibodies show reactivity with human, mouse, and rat samples , but cross-reactivity should be experimentally verified.
Application-specific validation: Ensure the antibody has been validated for your specific application through:
Clonality consideration: Choose between:
Immunogen mapping: Select antibodies with immunogens matching your region of interest, particularly important when studying KDM5C mutations or truncated variants .
Detection of mutant KDM5C proteins requires careful consideration of antibody epitope location:
Key methodology considerations:
Epitope mapping relative to mutations: When studying KDM5C mutations, verify the antibody epitope location relative to the mutation site. For instance, in cases like the c.2T>C mutation affecting the translation start codon, antibodies raised against the C-terminus can still detect the N-terminally truncated protein (p.M1_E165del) .
Multi-antibody approach: Using multiple antibodies targeting different regions of KDM5C provides more comprehensive detection. This is particularly important when analyzing patient samples with potential KDM5C mutations:
N-terminal antibodies for C-terminal mutations
C-terminal antibodies for N-terminal mutations
Domain-specific antibodies when studying particular functional domains
Western blot optimization: For mutant proteins with altered molecular weights:
Validation in relevant models: Validate antibody performance using:
Research has demonstrated that some KDM5C mutations (e.g., c.3223delG) can lead to complete absence of detectable protein, while others (e.g., c.2T>C) produce truncated proteins that lack functional domains like JmjN and ARID , affecting both detection and function.
ChIP-seq with KDM5C antibodies requires specific technical considerations:
Antibody ChIP-suitability: Not all KDM5C antibodies perform equally in ChIP applications. Select antibodies specifically validated for ChIP-seq , as the fixation process can mask epitopes.
Fixation optimization:
Standard 1% formaldehyde for 10 minutes is often insufficient
Test fixation times (5-15 minutes) and formaldehyde concentrations (0.5-2%)
Dual crosslinking with DSG (disuccinimidyl glutarate) prior to formaldehyde can improve chromatin binding protein detection
Sonication parameters:
Aim for 200-500 bp fragments
Optimize sonication conditions for each cell type
Monitor sonication efficiency by agarose gel electrophoresis
Controls and peak calling strategies:
Biological interpretation:
KDM5C expression varies significantly across tissue and cell types, necessitating experimental adjustments:
Expression patterns:
Nuclear localization in cytotrophoblasts (CTBs) and extravillous trophoblasts (EVTs)
Variable expression in cancer cell lines including HeLa, Jurkat, and MCF-7
Sex-specific differences in expression related to X-chromosome inactivation in females versus single X-chromosome in males
Antibody dilution optimization strategy:
Pilot titration experiment:
| Tissue/Cell Type | Recommended Starting WB Dilution | Recommended Starting IF Dilution |
|---|---|---|
| High expression (e.g., HeLa) | 1:5000 | 1:500 |
| Medium expression | 1:2000 | 1:200 |
| Low expression | 1:1000 | 1:50 |
Signal enhancement techniques for low expression systems:
Extended primary antibody incubation (overnight at 4°C)
Signal amplification systems (e.g., TSA)
More sensitive detection methods (ECL Plus for WB)
Protein concentration methods before analysis
Quantification and normalization:
Always include loading controls appropriate for subcellular fraction
Consider spike-in standards for absolute quantification
Use recombinant KDM5C standards when absolute quantification is needed
Distinguishing between KDM5C and KDM5D is particularly important for sex-specific studies:
Paralog-specific considerations:
Antibody selection strategy:
Verify antibody specificity against both KDM5C and KDM5D
Use epitopes targeting non-conserved regions between paralogs
Perform validation in male and female cell lines
Include appropriate controls (male cells lacking Y chromosome for KDM5C specificity)
Experimental design for sex difference studies:
The loss of Y chromosome, harboring KDM5D, occurs in most male KDM5C mutant clear cell renal cell carcinomas (ccRCCs)
Mutations in KDM5D prevented xenograft tumor formation in male 786-O cells, which was rescued by co-mutation of KDM5C
Transcriptional analyses show that KDM5C and KDM5D regulate both overlapping and distinct sets of genes
Methodological approach:
Use combination of antibody-based detection and genetic approaches
siRNA/shRNA knockdown validations with paralog-specific reagents
CRISPR-Cas9 knockout confirmations when possible
Quantitative PCR with paralog-specific primers to confirm target specificity
Research demonstrates that KDM5D in male cells does not function equivalently to the second KDM5C allele in female cells, challenging previous assumptions about these paralogs .
KDM5C mutations are implicated in X-linked intellectual disability (ID) and autism spectrum disorder, requiring specialized research approaches:
Patient-derived cell models:
Functional assays:
Histone demethylase activity assays to assess enzymatic function of mutant KDM5C
Transiently transfected mutant KDM5C constructs have shown reduced protein expression, stability, and decreased histone demethylase activities in cells
Layer-specific neuronal marker analysis (CTIP2, TBR1) shows reduction in KDM5C knockout models
Developmental timing considerations:
KDM5C's role is critical during specific neurodevelopmental windows
Conditional knockout models allow time-specific deletion
The canonical WNT signaling pathway has been identified as a potential therapeutic target for intellectual disability, with an unexpected role in cognition during specific developmental windows
Genotype-phenotype correlations:
Patients with KDM5C variations near the C-terminus tend to exhibit autism spectrum disorder in addition to intellectual disability
Different mutations produce varying phenotypic severity
Novel variations c.2233C>G and c.3392_3393delAG are associated with severe ID, short stature, and facial dysmorphism
Combined genomic and epigenomic analyses:
Rigorous quality control is essential for quantitative applications:
Assay validation parameters:
Reference standards and normalization:
Include recombinant KDM5C standards when possible
For relative quantification, select stable reference proteins
For ChIP-qPCR, normalize to input and include positive and negative genomic regions
Consider spike-in standards for absolute quantification
Troubleshooting inconsistent results:
Verify antibody lot consistency if results change over time
Test multiple antibodies targeting different epitopes
Consider protein degradation during sample preparation
Ensure complete protein extraction from nuclear fractions
Validation across research platforms:
Cross-validate results using orthogonal methods (e.g., MS-based proteomics)
Confirm protein-level changes with mRNA expression analysis
Use genetic approaches (siRNA, CRISPR) to confirm specificity
KDM5C exhibits context-dependent dual functions in gene activation and repression, requiring careful experimental design:
ChIP-seq and transcriptome integration:
Perform parallel H3K4me3 ChIP-seq and RNA-seq in KDM5C wildtype vs. knockout/knockdown systems
Categorize genes into KDM5C-activated and KDM5C-repressed groups
Identify genomic features associated with each category
Research has revealed that KDM5C can regulate both canonical and non-canonical WNT signaling pathways
Protein complex identification:
Co-immunoprecipitation with KDM5C antibodies followed by mass spectrometry
Identify context-specific interaction partners in different cell types
Compare complex formation in activation versus repression contexts
Proximity labeling approaches (BioID/TurboID) to identify transient interactions
Domain-specific functions:
Generate domain deletion constructs (JmjC, ARID, PHD, C5HC2 zinc-finger domains)
Rescue experiments with domain mutants in KDM5C-depleted cells
Domain-specific ChIP to identify regional binding patterns
Catalytic-dead mutants to distinguish enzymatic from scaffolding functions
Temporal dynamics analysis:
Time-course experiments after stimulation/differentiation
Compare early versus late KDM5C genomic occupancy
Correlate with dynamic changes in histone modifications
Inducible systems for temporal control of KDM5C expression
Research in breast cancer has shown that KDM5C can function in both gene transcriptional activation and repression to promote cancer cell growth , highlighting the importance of context in determining KDM5C function.
Different disease contexts require tailored experimental approaches:
Cancer research approaches:
KDM5C is commonly mutated in clear cell renal cell carcinomas (ccRCC) in men but rarely in women
Mutation of KDM5D in male 786-O cells prevented xenograft tumor formation and was rescued by co-mutation of KDM5C
Design experiments accounting for sex differences and Y chromosome loss
Test KDM5 inhibitors in preclinical models for therapeutic potential
Reproductive biology studies:
Elevated KDM5C increases recurrent miscarriage risk by preventing trophoblast proliferation and invasion
KDM5C is mainly expressed within the nuclei of cytotrophoblasts (CTBs) and extravillous trophoblasts (EVTs)
Combine tissue immunohistochemistry with functional trophoblast assays
Design in vivo models using systemically delivered KDM5C adenovirus vectors (Ad-KDM5C) which have been shown to promote embryo resorption in mice
Bone metabolism research:
Neurodevelopmental disorder investigations:
Tissue-specific optimization is critical for successful KDM5C immunohistochemistry:
Sample preparation considerations:
Fresh frozen versus FFPE tissue requires different antibody dilutions
Antigen retrieval methods: heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) is often optimal
Section thickness: 4-5 μm sections typically work best
Fixation time affects epitope accessibility and should be standardized
Protocol optimization strategy:
Tissue-specific considerations:
Chorionic villus samples: KDM5C is mainly expressed within the nuclei of CTBs and EVTs
Brain tissue: Nuclear staining in neurons with some regional variability
Cancer samples: Expression may be heterogeneous within the sample
Always include positive control tissues (e.g., HeLa cells embedded in paraffin)
Validation approaches:
Use double immunofluorescence staining with established markers (e.g., CK-7 for trophoblasts)
Include tissue from KDM5C knockout models as negative controls
Compare multiple antibodies targeting different epitopes
Quantify staining intensity using standardized scoring systems or digital image analysis
Investigating the demethylase activity of KDM5C requires specialized approaches:
Enzymatic activity assays:
In vitro histone demethylase assays with recombinant KDM5C
KDM5C specifically demethylates trimethylated and dimethylated but not monomethylated H3K4
KDM5C does not demethylate H3K9, H3K27, H3K36, H3K79, or H4K20
Use KDM5 inhibitors as controls - they increase H3K4me3 levels in CD14+ monocytes of human peripheral blood mononuclear cells
ChIP-seq approaches:
Parallel KDM5C and H3K4me3 ChIP-seq experiments
Analyze genomic regions where KDM5C binding correlates with reduced H3K4me3
Time-course experiments to capture demethylation dynamics
Combined analysis of RNA-seq, ChIP-seq, and CUT&Tag assays shows that KDM5C overexpression leads to reduction of H3K4me3 on promoters and corresponding downregulation of gene expression
Advanced epigenomic methods:
CUT&Tag for higher resolution profiling of histone modifications
Single-cell approaches to capture cellular heterogeneity
Mass spectrometry-based histone modification analysis for global quantification
Genomic engineering of histone H3 to test modification-specific effects
Inhibitor-based approaches:
KDM5 inhibitors dose-dependently affect biological processes
Median inhibitory concentration (IC50) of 5.6 μM has been reported for inhibition of RANKL-induced osteoclastogenesis
Monitor dose-dependent increases in H3K4me3 levels as confirmation of target engagement
Combine with genetic approaches to confirm specificity
Troubleshooting strategies for common KDM5C antibody issues:
Weak or no signal in Western blots:
Potential causes and solutions:
| Issue | Solution |
|---|---|
| Insufficient protein | Increase loading amount (start with 50-75 μg total protein) |
| Inefficient extraction | Use nuclear extraction protocols with detergents |
| Protein degradation | Add protease inhibitors freshly; keep samples cold |
| Inefficient transfer | Optimize transfer conditions for large proteins (>170 kDa) |
| Antibody dilution too high | Try more concentrated primary antibody (1:1000 instead of 1:5000) |
| Inadequate detection | Use more sensitive detection systems (ECL Plus) |
Multiple bands or unexpected molecular weight:
Potential causes and solutions:
Inconsistent immunostaining:
Standardize fixation methods and times
Optimize permeabilization for nuclear antigens
Include positive control samples in each experiment
Use automated staining systems when available for consistency
Standardize image acquisition parameters
Batch-to-batch variability:
Test new antibody lots against reference samples
Maintain reference lysates as standards
Consider monoclonal antibodies for better reproducibility
Document lot numbers and maintain consistent suppliers
Comprehensive validation approaches ensure reliable results:
Genetic validation approaches:
siRNA/shRNA knockdown followed by Western blot (expected reduction in signal)
CRISPR/Cas9 knockout (complete absence of specific signal)
Rescue experiments with ectopic expression
Quantitative PCR correlation with protein levels
Cross-reactivity assessment:
Test in multiple species if cross-reactivity is claimed
Test for cross-reactivity with other KDM5 family members (particularly KDM5D)
Peptide competition assays to confirm epitope specificity
Pre-adsorption tests with recombinant antigens
Orthogonal method confirmation:
Mass spectrometry validation of immunoprecipitated proteins
Correlation with mRNA expression data
Independent antibodies targeting different epitopes
Multiple application validation (WB, IP, IF, ChIP)
Context-specific validation:
KDM5C protein is homologous to three other proteins of the human JARID1 family (JARID1A, JARID1B and JARID1D/KDM5D), sharing several evolutionarily conserved domains that may cause cross-reactivity if not properly validated .
Investigating non-canonical functions of KDM5C:
Co-immunoprecipitation strategies:
Use validated KDM5C antibodies for pull-down experiments
Analyze by mass spectrometry to identify novel interacting proteins
Reciprocal co-IP to confirm interactions
Proximity labeling approaches (BioID/TurboID) to identify adjacent proteins
Functional domain mapping:
Generate domain-specific antibodies or tagged constructs
Determine which domains mediate non-histone interactions
Create domain deletion mutants to test functional consequences
Investigate proteins containing lysine methylation sites
Non-histone methylation targets:
Develop methylation-specific antibodies for candidate non-histone targets
Use recombinant KDM5C in in vitro demethylation assays
Test methylation status of candidates after KDM5C manipulation
Apply proteomic approaches to identify global changes in protein methylation
Subcellular localization studies:
Investigate potential cytoplasmic roles using fractionation
Co-localization studies with candidate interaction partners
Live-cell imaging with fluorescently tagged KDM5C
Test localization changes under different cellular conditions
KDM5 inhibitors are emerging therapeutic agents requiring specialized research approaches:
Target engagement confirmation:
Functional consequence assessment:
KDM5 inhibitors dose-dependently suppress RANKL-induced osteoclastogenesis with an IC50 of 5.6 μM
KDM5 inhibition down-regulates key mitochondrial OXPHOS complex proteins and mRNAs
Measure cellular processes impacted by inhibition (OCR, ECAR, ATP production)
Compare phenotypic effects to genetic KDM5C depletion
Experimental design considerations:
Include dose-response analyses (typically 0.1-10 μM range)
Determine time-dependency of effects
Address isoform specificity through selective knockdown experiments
Test effects across multiple cell types (effects may be context-dependent)
Combination approaches:
Test synergy with other epigenetic modulators
Investigate combinations relevant to disease contexts
Measure combined effects on transcriptional outputs
Develop biomarkers for response prediction