KDM3A Antibody (e.g., Proteintech 12835-1-AP) is a polyclonal rabbit IgG antibody targeting the N-terminal region of human KDM3A, also known as JMJD1A. This antibody is widely used to investigate KDM3A's role in epigenetic processes, including its enzymatic activity of demethylating mono- and di-methylated lysine 9 on histone H3 (H3K9me1/me2) . Its applications span cancer biology, developmental studies, and metabolic research.
Cancer Biology:
KDM3A is upregulated in pancreatic ductal adenocarcinoma (PDAC) and promotes tumor growth by enhancing stemness and hypoxia-related pathways. Knockdown experiments using KDM3A-targeting shRNAs reduced PDAC cell invasion and migration .
In colorectal cancer (CRC), KDM3A serves as a prognostic biomarker and therapeutic target due to its hypoxic regulation .
Developmental Biology:
Metabolic Regulation:
Western Blot: Use RIPA lysates with protease inhibitors. Block with 5% non-fat milk .
IHC: Antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) .
KDM3A (Lysine Demethylase 3A, also known as JMJD1A or JHDM2A) is a histone demethylase that specifically removes mono- and dimethyl marks from lysine 9 of histone H3 (H3K9me1/2), thereby playing a central role in histone code regulation . It is a member of the JHDM2 histone demethylase family and has a calculated molecular weight of approximately 147.3 kDa, though it is commonly observed at around 150 kDa in Western blot analyses . KDM3A is primarily localized in the nucleus but can also be found in the cytoplasm . Its demethylase activity contributes to transcriptional activation by removing repressive H3K9 methyl marks from gene promoters.
KDM3A antibodies are primarily used in the following applications:
Western blotting (WB) - For detecting KDM3A protein expression levels in cell and tissue lysates
Immunoprecipitation (IP) - For isolating KDM3A protein complexes
Chromatin Immunoprecipitation (ChIP) - For identifying genomic regions bound by KDM3A
Immunofluorescence (IF) - For visualizing subcellular localization of KDM3A
Enzyme-linked immunosorbent assay (ELISA) - For quantitative detection of KDM3A
Most commercially available antibodies recognize human KDM3A, with some also cross-reacting with mouse and rat orthologs .
When validating a new KDM3A antibody, the following controls should be included:
Positive control: Use cell lines known to express KDM3A (e.g., 293T human embryonic kidney cells, MOLT-4 human acute lymphoblastic leukemia cells)
Negative control: Include KDM3A knockdown samples using siRNA or shRNA approaches
Specificity control: Test cross-reactivity with related proteins (KDM3B, KDM3C/JMJD1C) to confirm specificity
Loading control: Include antibodies against housekeeping proteins (e.g., β-actin) to normalize protein loading
Subcellular fractionation: Compare detection in nuclear vs. cytoplasmic fractions
Molecular weight verification: Confirm band appears at expected size (~150 kDa)
Peptide blocking: If available, perform peptide competition assay to confirm specificity
For optimal Western blot detection of KDM3A:
Sample preparation:
Gel electrophoresis conditions:
Transfer conditions:
Transfer to PVDF membrane at low current (250-300 mA) overnight at 4°C for large proteins
Antibody dilutions:
Detection system:
The specific conditions should be optimized for each antibody and sample type, as indicated by manufacturer recommendations.
Designing and validating ChIP-Seq experiments with KDM3A antibodies requires careful consideration:
Antibody selection:
Crosslinking and sonication:
Use 1% formaldehyde for 10 minutes at room temperature
Optimize sonication to obtain chromatin fragments of 200-500 bp
Experimental controls:
Validation approaches:
Data analysis considerations:
Studies have shown that KDM3A often co-localizes with transcription factors such as estrogen receptor (ER) and androgen receptor (AR) at their target gene promoters .
KDM3A antibodies can be strategically employed to investigate its role in cancer progression through several methodological approaches:
Expression analysis in cancer tissues:
Functional studies in cancer models:
Mechanistic studies in specific cancer types:
Breast cancer: Investigate KDM3A interaction with estrogen receptor (ER) at target gene promoters using sequential ChIP or co-immunoprecipitation
Ewing Sarcoma: Examine KDM3A regulation of metastasis-associated genes like MCAM
Prostate cancer: Study co-localization with androgen receptor at target loci
Therapeutic target validation:
Research has demonstrated that KDM3A regulates genes involved in cell growth, adhesion, and motility across multiple cancer types, making it a potential therapeutic target .
Studying KDM3A post-translational modifications requires specialized approaches:
Phosphorylation analysis:
Identification of novel modifications:
Functional implications:
Signaling pathway analysis:
Research has identified that KDM3A is phosphorylated by JAK2 at tyrosine 1101, with the Y1101 residue being well conserved across species, suggesting functional importance .
Integration of ChIP-Seq and RNA-Seq provides powerful insights into KDM3A-mediated gene regulation:
Experimental design for integration:
Data analysis workflow:
Functional validation strategies:
Advanced insights from integrated analysis:
Identify direct vs. indirect KDM3A targets
Uncover cell-type specific regulation patterns
Map KDM3A cooperation with lineage-specific transcription factors
Understand temporal dynamics of gene regulation
Studies combining these approaches have revealed that KDM3A regulates distinct gene clusters in different cancers - estrogen-responsive genes in breast cancer , migration-related genes in Ewing Sarcoma , and androgen-regulated genes in prostate cancer .
To effectively study KDM3A in cell differentiation models:
Model system selection:
KDM3A manipulation approaches:
Differentiation experimental design:
Analysis methods:
Assess KDM3A expression changes during differentiation by RT-qPCR and Western blot
Examine differentiation markers (e.g., TROMA-1 for primitive endoderm)
Analyze expression of key differentiation genes by qRT-PCR
Perform ChIP-Seq to identify KDM3A binding sites during differentiation
Consider rescue experiments by re-expressing KDM3A or downstream targets
Research has shown that KDM3A is essential for proper differentiation of F9 cells into primitive endoderm, with knockdown cells failing to properly express differentiation markers despite morphological changes .
To identify novel KDM3A interaction partners:
Affinity purification approaches:
Validation of interactions:
Reciprocal co-immunoprecipitation with antibodies against identified partners
GST pull-down assays to test direct interactions
Proximity ligation assay (PLA) for visualizing interactions in situ
Co-localization studies using immunofluorescence
Functional characterization:
Knockdown of interaction partners to assess effects on KDM3A activity
Domain mapping to identify interaction interfaces
ChIP-seq for co-localization at genomic loci
Transcriptome analysis following partner depletion
Context-specific interactions:
Compare interactomes under different conditions (e.g., hormone stimulation)
Analyze cell-type specific interactions
Investigate interactions during different cell cycle phases
Studies have identified several important interaction partners for KDM3A including transcription factors like estrogen receptor , androgen receptor , and Ets1 , suggesting context-dependent regulatory mechanisms.
Distinguishing between catalytic and non-catalytic functions of KDM3A requires sophisticated experimental approaches:
Generation of catalytically inactive mutants:
Rescue experiments:
Chromatin analysis:
Perform ChIP for H3K9me1/2 marks at KDM3A target genes
Compare histone modification patterns with wild-type vs. mutant KDM3A
Conduct sequential ChIP to identify factors recruited by KDM3A regardless of its catalytic activity
Transcriptome analysis:
Perform RNA-seq in cells expressing wild-type or catalytically inactive KDM3A
Identify genes regulated by KDM3A independent of its demethylase activity
Compare with direct binding sites from ChIP-seq data
Protein domain analysis:
Create truncation mutants to isolate functions of different KDM3A domains
Examine protein-protein interactions of each domain
Assess localization patterns of domain mutants
Research has demonstrated that the catalytic activity of KDM3A is required for estrogen receptor target gene expression and cell growth in breast cancer, indicating that its enzymatic function is essential for at least some of its biological roles .
When interpreting discrepancies in KDM3A molecular weight:
Expected vs. observed molecular weight:
Potential explanations for discrepancies:
Validation approaches:
Experimental considerations:
Use gradient gels for better resolution of high molecular weight proteins
Include appropriate molecular weight markers covering the expected range
Consider using phosphatase treatment to eliminate phosphorylation-based shifts
The observation of both full-length (~150 kDa) and partial (~70 kDa) KDM3A bands is documented in the literature and should be interpreted in the context of the specific experimental conditions .
Validating KDM3A as a therapeutic target in cancer research requires multiple lines of evidence:
Expression analysis in clinical samples:
Functional dependency studies:
Mechanistic understanding:
Therapeutic potential assessment:
Develop and test small molecule inhibitors of KDM3A catalytic activity
Compare genetic vs. pharmacological inhibition phenotypes
Evaluate potential synergies with established therapies
Assess possible toxicities through studies in normal cells
Research has shown that KDM3A regulates estrogen receptor signaling in breast cancer , promotes metastasis in Ewing Sarcoma through MCAM regulation , and modulates androgen receptor activity in prostate cancer , supporting its potential as a therapeutic target across multiple cancer types.
When encountering contradictory findings about KDM3A function:
Systematic comparison of experimental conditions:
Cell type and tissue context differences (e.g., embryonic vs. differentiated, normal vs. cancer)
Knockdown/knockout methodology (transient vs. stable, partial vs. complete)
Antibody specificity and epitope locations
Experimental timing and dynamic processes
Molecular context considerations:
Expression levels of KDM3A cofactors and interaction partners
Presence of compensatory mechanisms (e.g., other KDM family members)
Cell-type specific transcription factor landscapes
Epigenetic background and chromatin accessibility
Technical validation approaches:
Reproduce key experiments using multiple methodologies
Use complementary gain- and loss-of-function approaches
Validate antibody specificity through knockout controls
Test critical findings across multiple cell lines/models
Integrative interpretation strategies:
Develop context-specific models of KDM3A function
Consider distinct roles in different biological processes
Integrate data across multiple omics platforms
Acknowledge limitations and biological complexity