KDM3A Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to KDM3A Antibody

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.

Key Findings Using KDM3A Antibody

  • 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:

    • KDM3A is critical for mammary gland epithelial proliferation and ductal morphogenesis. Its ablation in mice disrupts these processes, implicating it in breast cancer progression .

  • Metabolic Regulation:

    • KDM3A regulates genes like PPARA and UCP1, influencing lipid metabolism and obesity resistance .

Validated Applications

ApplicationRecommended DilutionTested Samples
Western Blot (WB)1:1000–1:4000HeLa cells, MCF-7 cells
Immunohistochemistry (IHC)1:20–1:200Human ovary tumor, kidney, and normal tissues
Immunofluorescence (IF)1:50–1:500HepG2 cells

Table: Key Research Findings

Study FocusMethodologyOutcomeCitation
PDAC Tumor GrowthKDM3A knockdown in S2–007 and MiaPaCa2 cellsReduced colony formation, invasion, and tumor growth in mice
Breast CancerKdm3a KO miceImpaired mammary ductal development and epithelial proliferation
Hypoxic Tumor RegulationCRC patient samplesKDM3A linked to poor prognosis and hypoxia adaptation

Protocols and Best Practices

  • 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) .

Limitations and Considerations

  • Species Specificity: While validated for human and mouse, cross-reactivity with other species requires empirical testing.

  • Batch Variability: Optimal dilutions may vary; titration is recommended for new experimental setups .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the purchase method or location. Please consult your local distributor for specific delivery details.
Synonyms
DKFZp686A24246 antibody; DKFZp686P07111 antibody; JHDM2A antibody; JHMD2A antibody; JmjC domain-containing histone demethylation protein 2A antibody; JMJD1 antibody; JMJD1A antibody; Jumonji C domain containing histone demethylase 2A antibody; Jumonji domain containing 1 antibody; Jumonji domain containing 1A antibody; Jumonji domain containing protein 1A antibody; Jumonji domain-containing protein 1A antibody; Kdm3a antibody; KDM3A_HUMAN antibody; KIAA0742 antibody; Lysine (K) specific demethylase 3A antibody; Lysine-specific demethylase 3A antibody; Testis specific protein A antibody; TSGA antibody
Target Names
KDM3A
Uniprot No.

Target Background

Function
KDM3A (Lysine-specific demethylase 3A) is a histone demethylase enzyme that specifically removes methyl groups from lysine 9 of histone H3 (H3K9). This activity plays a crucial role in the histone code, a complex system of epigenetic modifications that regulate gene expression. KDM3A preferentially demethylates mono- and dimethylated H3K9, showing a stronger preference for the dimethylated form. It exhibits minimal or no activity on trimethylated H3K9. The demethylation process generates formaldehyde and succinate. KDM3A is involved in several biological processes, including:

Hormone-dependent transcriptional activation: KDM3A participates in the recruitment of androgen receptors to target genes, leading to H3K9 demethylation and transcriptional activation.

Spermatogenesis: KDM3A regulates the expression of genes like PRM1 and TNP1, essential for packaging and condensing sperm chromatin, contributing to successful spermatogenesis.

Obesity resistance: KDM3A is implicated in regulating metabolic genes like PPARA and UCP1, potentially playing a role in obesity resistance.
Gene References Into Functions
KDM3A's Role in Various Biological Processes and Diseases:

  1. Normal stromal cells restrict cancer cell proliferation by modulating gene expression through KDM3A. PMID: 27488962
  2. KDM3A interacts with and promotes the recruitment of HNRNPF, facilitating the splicing of AR-V7, contributing to prostate cancer development. PMID: 29712835
  3. Elevated KDM3A expression is associated with lymph node metastasis in oral and oropharyngeal squamous cell carcinoma. PMID: 29590186
  4. Hypoxia activates a HIF-KDM3A-MMP12 signaling pathway that promotes trophoblast invasion and uterine spiral artery remodeling. PMID: 27807143
  5. High levels of KDM3A and c-Myc are independent prognostic indicators for cervical cancer patients. PMID: 27835890
  6. Research suggests a critical role for KDM3A in the PI3K/AP-1 oncogenic axis, proposing a novel strategy for inhibiting KDM3A against liver tumor development under PI3K pathway activation. PMID: 28692045
  7. KDM3A promotes anoikis (cell death) through transcriptional activation of BNIP3 and BNIP3L, which encode pro-apoptotic proteins. PMID: 27472901
  8. The KDM3A to PRM1 mRNA expression ratio can serve as a reliable marker for successful testicular sperm extraction in men with obstructive and non-obstructive azoospermia, with 95% sensitivity. PMID: 27027467
  9. KDM3A regulates MCAM expression through both direct and indirect mechanisms. Direct regulation involves modulation of H3K9 methylation at the MCAM promoter, while indirect regulation occurs via the Ets1 transcription factor. PMID: 28319067
  10. KDM3A promotes urinary bladder cancer progression by enhancing glycolysis through coactivation of HIF1alpha. PMID: 28263974
  11. Depletion of KDM3A reactivates mutated p53, inducing pro-apoptotic gene expression in breast cancer with mutant p53. KDM3A knockdown also significantly inhibits the tumorigenic potential of breast cancer stem-like cells and renders them sensitive to apoptosis induced by chemotherapeutic drugs. PMID: 27270439
  12. Research highlights a crucial role for KDM3A in promoting ovarian cancer stem cell proliferation and chemoresistance, suggesting it as a potential therapeutic target for resistant ovarian cancer. PMID: 27694900
  13. KDM3A plays a critical role in regulating prostate cancer cell proliferation and survival by controlling c-Myc expression at both transcriptional and post-translational levels. PMID: 26279298
  14. Deficient expression of KDM3A might reflect and/or contribute to round spermatid maturation arrest. PMID: 27692601
  15. KDM3A could promote non-small cell lung cancer tumorigenesis. PMID: 26945572
  16. Studies have identified KDM3A, an H3K9me2 demethylase, as responsible for H3K9me2 reduction and crucial for breast tumor transformation. PMID: 27034728
  17. The JMJD1A-MALAT1-MAPK signaling pathway might participate in KDM3A-induced cell proliferation in gastric cancer. PMID: 26617828
  18. The KDM3A-KLF2-IRF4 pathway plays a significant role in multiple myeloma cell survival and homing to the bone marrow, suggesting it as a therapeutic target. PMID: 26728187
  19. KDM3A is phosphorylated at S265 by protein kinase A (PKA), which is crucial for activating the beta1-adrenergic receptor gene (Adrb1) and downstream targets including Ucp1 in brown adipocytes. PMID: 25948511
  20. Loss of KDM3A expression is associated with liver fibrosis. PMID: 25609425
  21. Mitogen- and stress-activated protein kinase 1 (MSK1) specifically phosphorylates KDM3A at Ser264 (p-KDM3A), which is enriched in the regulatory regions of gene loci in the human genome. PMID: 25535969
  22. Studies identify the histone demethylase KDM3A as a novel, miR-regulated, tumor promoter in Ewing Sarcoma. PMID: 24362521
  23. ACK1 interacts with KDM3A to regulate the mammary tumor oncogene HOXA1. PMID: 25148682
  24. Research identifies a novel pathway through which N-Myc causes neuroblastoma cell migration and invasion, providing evidence for the development of more potent JMJD1A/MALAT1 inhibitors to prevent tumor metastasis. PMID: 24742640
  25. Studies found that KDM3A was consistently and significantly downregulated at both RNA and protein levels in human germ cell tumors. PMID: 25071150
  26. KDM3A forms a homodimer through its catalytic domains, bringing the two active sites close together. PMID: 24214985
  27. Expression of JHDM2A was significantly increased, while HDAC2, HDAC7, and SUV39H2 were significantly down-regulated in Systemic Sclerosis B cells compared to controls. PMID: 23891737
  28. A single amino acid in KDM3A, T667, affects histone demethylase activity towards H3K9me1 and -me2. PMID: 23593242
  29. Data indicate that JMJD1A gene silencing abrogated the hypoxia-induced adrenomedullin (ADM) expression and inhibited HepG2 and Hep3B cell growth. PMID: 23583388
  30. Exposing cells to either chemical or cellular sources of nitric oxide (NO) resulted in a significant increase in dimethyl Lys-9 on histone 3 (H3K9me2), the preferred substrate for KDM3A. PMID: 23546878
  31. Results suggest that LANA may play a role in regulating epigenetic marks on the KSHV genome, partly through association with the histone demethylase KDM3A. PMID: 23576503
  32. Research indicates that KDM3A is a sensitive recurrence marker, and it can promote malignant transformation via epithelial-mesenchymal transition. PMID: 21607773
  33. KDM3A is recruited to the SLC2A3 locus in an HIF1-dependent manner and demethylates H3K9me2, leading to upregulation of its expression. PMID: 22645302
  34. Findings suggest that both Ni(2+) and ascorbate can regulate the expression of histone demethylase KDM3A, which is significant for cancer development or inhibition. PMID: 22318714
  35. A study demonstrates that KDM3A is overexpressed in various types of cancer and directly activates transcription of HOXA1 through demethylation of histone H3K9 by binding to its promoter region. PMID: 22020899
  36. Up-regulation of miR-155 in nasopharyngeal carcinoma is partly driven by LMP1 and LMP2A, resulting in downregulation of KDM3A. PMID: 21541331
  37. Increased expression of KDM3A might be associated with the progression of kidney cancer. PMID: 21275466
  38. KDM3A is a novel prognostic marker for colorectal cancer. PMID: 20823141
  39. Genetic alterations and expression changes of LSD1, JHDM2A, and GASC1 in prostate cancer (PC) were investigated. As no genetic alterations and only modest expression changes were found, it is unlikely they play a major role in the progression of PC. PMID: 20127736
  40. Data show that the JMJD1A/ABH2 family of dioxygenases is highly sensitive to inhibition by carcinogenic nickel ions. PMID: 20042601
  41. Hypoxic regulation of KDM3A acts as a signal amplifier to facilitate hypoxic gene expression, ultimately enhancing tumor growth. PMID: 19858293
  42. Research demonstrates that KDM3A can be stimulated by hypoxia both in vitro and in vivo, involving binding of HIF-1 to a specific HRE in the KDM3A promoter. PMID: 18538129
  43. Results show that many genes regulated by hypoxia and HIF-1alpha exhibit patterns of induction, with KDM3A and JMJD2B demonstrating robust up-regulation by hypoxia, while JMJD2C shows more modest up-regulation. PMID: 18713068
  44. Histone demethylases KDM3A and JMJD2B are transcriptional targets of hypoxia-inducible factor HIF. PMID: 18984585
  45. Immunohistochemical staining has revealed that KDM3A is widely expressed in tissues, even in cells that are not known to express the androgen receptor, and is significantly increased in smooth muscle cells upon hypoxia treatment. PMID: 19471969
Database Links

HGNC: 20815

OMIM: 611512

KEGG: hsa:55818

STRING: 9606.ENSP00000323659

UniGene: Hs.557425

Protein Families
JHDM2 histone demethylase family
Subcellular Location
Cytoplasm. Nucleus.

Q&A

What is KDM3A and what is its molecular function?

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.

What are the most common applications for KDM3A antibodies?

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 .

What controls should be included when validating a new KDM3A antibody?

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

What are the optimal conditions for Western blot detection of KDM3A?

For optimal Western blot detection of KDM3A:

  • Sample preparation:

    • Use RIPA or NP-40 based lysis buffers containing protease inhibitors

    • Load 20-30 μg of whole cell lysate, or 10-20 μg of nuclear extract for enriched detection

  • Gel electrophoresis conditions:

    • Use 8% SDS-PAGE gels to properly resolve the 150 kDa KDM3A protein

    • Run under reducing conditions using Immunoblot Buffer Group 1

  • Transfer conditions:

    • Transfer to PVDF membrane at low current (250-300 mA) overnight at 4°C for large proteins

  • Antibody dilutions:

    • Primary antibody: 1:1000 to 1:2000 dilution (typically 1-2 μg/mL)

    • Secondary antibody: HRP-conjugated anti-species IgG at 1:5000 dilution

  • Detection system:

    • Enhanced chemiluminescence (ECL) detection is suitable for most applications

    • Note that degradation or processing products may appear at ~70 kDa

The specific conditions should be optimized for each antibody and sample type, as indicated by manufacturer recommendations.

How should ChIP-Seq experiments with KDM3A antibodies be designed and validated?

Designing and validating ChIP-Seq experiments with KDM3A antibodies requires careful consideration:

  • Antibody selection:

    • Choose ChIP-grade antibodies specifically validated for chromatin immunoprecipitation

    • Verify antibody specificity via KDM3A knockdown controls

  • Crosslinking and sonication:

    • Use 1% formaldehyde for 10 minutes at room temperature

    • Optimize sonication to obtain chromatin fragments of 200-500 bp

  • Experimental controls:

    • Input DNA (pre-immunoprecipitation sample)

    • IgG negative control (non-specific antibody)

    • KDM3A-depleted cells as biological negative control

  • Validation approaches:

    • Perform parallel ChIP-qPCR for known KDM3A target regions

    • Include H3K9me1/me2 ChIP to confirm inverse correlation with KDM3A binding

    • Analyze changes in histone marks following KDM3A knockdown

  • Data analysis considerations:

    • For KDM3A binding sites, focus on promoter regions of regulated genes

    • Look for correlation with H3K9me1/2 demethylation events

    • Integrate with transcriptome data to identify functional targets

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 .

What troubleshooting approaches can resolve common issues with KDM3A antibody detection?

IssuePossible CausesTroubleshooting Approaches
No signal in Western blotLow KDM3A expression, antibody degradation, improper dilution1. Use nuclear extracts to enrich KDM3A
2. Increase antibody concentration
3. Extend exposure time
4. Verify protein transfer efficiency
Multiple bands/non-specific bindingCross-reactivity, protein degradation1. Increase blocking time/concentration
2. Use fresh lysates with protease inhibitors
3. Try different KDM3A antibody
4. Note that ~70 kDa band may represent degradation product
Poor ChIP enrichmentInefficient crosslinking, non-optimal sonication, low antibody affinity1. Optimize crosslinking conditions
2. Adjust sonication parameters
3. Increase antibody amount
4. Include positive control regions (known KDM3A targets)
Inconsistent results between experimentsAntibody batch variation, cell culture conditions1. Use the same antibody lot when possible
2. Standardize cell culture conditions
3. Include appropriate controls in each experiment
Discrepancy between protein size (147 kDa calculated vs. observed)Post-translational modifications, isoforms1. KDM3A undergoes phosphorylation and potentially other modifications
2. At least two isoforms of KDM3A are known to exist

How can KDM3A antibodies be used to study its role in cancer progression?

KDM3A antibodies can be strategically employed to investigate its role in cancer progression through several methodological approaches:

  • Expression analysis in cancer tissues:

    • Immunohistochemistry (IHC) to compare KDM3A levels between normal and malignant tissues

    • Correlation of KDM3A expression with clinical outcomes and patient survival

  • Functional studies in cancer models:

    • Combine KDM3A antibodies with knockdown/knockout approaches to validate specificity

    • ChIP-Seq to identify cancer-specific KDM3A targets

    • Analysis of H3K9me1/2 dynamics at oncogene promoters using antibodies against both KDM3A and these histone marks

  • 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:

    • Monitor changes in KDM3A levels and activity following treatment with epigenetic inhibitors

    • Combine with cell proliferation, migration, and invasion assays to correlate KDM3A activity with cancer phenotypes

Research has demonstrated that KDM3A regulates genes involved in cell growth, adhesion, and motility across multiple cancer types, making it a potential therapeutic target .

What approaches can be used to study KDM3A post-translational modifications?

Studying KDM3A post-translational modifications requires specialized approaches:

  • Phosphorylation analysis:

    • Use phospho-specific antibodies like anti-KDM3A-Y1101p to detect tyrosine phosphorylation

    • Combine with phosphatase treatments as controls

    • Immunoprecipitation with anti-KDM3A followed by immunoblotting with pan-phosphotyrosine antibodies

  • Identification of novel modifications:

    • Immunoprecipitate KDM3A followed by mass spectrometry (LC-MS/MS)

    • Compare modification patterns under different cellular conditions

    • Generate and validate site-specific antibodies for newly identified modifications

  • Functional implications:

    • Create point mutations at modification sites (e.g., Y1101A)

    • Compare enzymatic activity of wild-type vs. modification-deficient KDM3A

    • Perform ChIP-Seq with wildtype and mutant forms to identify differentially regulated targets

  • Signaling pathway analysis:

    • Study kinases responsible for KDM3A modification (e.g., JAK2 for Y1101 phosphorylation)

    • Use kinase inhibitors to modulate KDM3A modification status

    • Examine effects on KDM3A localization, stability, and activity

Research has identified that KDM3A is phosphorylated by JAK2 at tyrosine 1101, with the Y1101 residue being well conserved across species, suggesting functional importance .

How can ChIP-Seq and RNA-Seq be integrated to understand KDM3A-mediated gene regulation?

Integration of ChIP-Seq and RNA-Seq provides powerful insights into KDM3A-mediated gene regulation:

  • Experimental design for integration:

    • Perform KDM3A ChIP-Seq, H3K9me1/2 ChIP-Seq, and RNA-Seq in parallel

    • Include KDM3A knockdown/knockout conditions for all assays

    • Consider time-course experiments to capture dynamic regulation

  • Data analysis workflow:

    • Identify KDM3A binding sites genome-wide

    • Map H3K9me1/2 changes associated with KDM3A binding

    • Correlate binding events with transcriptional changes from RNA-Seq

    • Perform pathway enrichment analysis of differentially expressed genes

  • Functional validation strategies:

    • Select candidate target genes for validation by RT-qPCR

    • Perform ChIP-qPCR at specific loci to confirm KDM3A binding

    • Use luciferase reporter assays with promoter regions of regulated genes

    • Examine co-factor recruitment at KDM3A-regulated promoters

  • 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 .

How should experiments be designed to study KDM3A in cell differentiation models?

To effectively study KDM3A in cell differentiation models:

  • Model system selection:

    • Use established differentiation models like F9 embryonal carcinoma cells

    • Consider embryonic stem cells, where KDM3A maintains pluripotency

    • Compare with lineage-committed cell types

  • KDM3A manipulation approaches:

    • Generate stable KDM3A knockdown lines using shRNA

    • Create KDM3A knockout models using CRISPR/Cas9

    • Include wild-type and control vector cells for comparison

  • Differentiation experimental design:

    • Induce differentiation using established protocols (e.g., retinoic acid for F9 cells)

    • Monitor morphological changes throughout differentiation

    • Collect samples at multiple time points for comprehensive analysis

  • 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 .

What strategies can be employed to identify novel KDM3A interaction partners?

To identify novel KDM3A interaction partners:

  • Affinity purification approaches:

    • Immunoprecipitation (IP) with anti-KDM3A antibodies followed by mass spectrometry

    • Tag-based approaches (FLAG, HA) with exogenously expressed KDM3A

    • Proximity-based labeling methods (BioID, APEX)

    • Consider both nuclear and cytoplasmic fractions separately

  • 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.

How can researchers distinguish between catalytic and non-catalytic functions of KDM3A?

Distinguishing between catalytic and non-catalytic functions of KDM3A requires sophisticated experimental approaches:

  • Generation of catalytically inactive mutants:

    • Create point mutations in the JmjC domain (e.g., H1120G/D1122N)

    • Express wild-type and mutant forms at equal levels

    • Verify loss of demethylase activity using in vitro assays

  • Rescue experiments:

    • Deplete endogenous KDM3A using siRNA or shRNA approaches

    • Re-express either wild-type or catalytically inactive KDM3A

    • Compare rescue efficiency for different phenotypes and target genes

  • 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 .

How should researchers interpret apparent discrepancies in KDM3A molecular weight?

When interpreting discrepancies in KDM3A molecular weight:

  • Expected vs. observed molecular weight:

    • KDM3A has a calculated molecular weight of approximately 147.3 kDa

    • In Western blots, it is commonly observed at approximately 150 kDa

    • A band at approximately 70 kDa may represent degradation or processing products

  • Potential explanations for discrepancies:

    • Post-translational modifications (phosphorylation , potential ubiquitination, etc.)

    • Alternative splicing resulting in different isoforms (at least two isoforms are known)

    • Protein processing or degradation during sample preparation

    • Differences in gel systems and molecular weight standards

  • Validation approaches:

    • Use multiple KDM3A antibodies targeting different epitopes

    • Include KDM3A knockdown/knockout controls to confirm specificity

    • Perform immunoprecipitation followed by mass spectrometry for definitive identification

    • Compare detection patterns across different cell lines and tissues

  • 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 .

What criteria should be used to validate KDM3A as a therapeutic target in cancer research?

Validating KDM3A as a therapeutic target in cancer research requires multiple lines of evidence:

  • Expression analysis in clinical samples:

    • Evaluate KDM3A expression in tumor vs. normal tissues across large patient cohorts

    • Correlate expression with clinical outcomes and survival

    • Stratify analysis by cancer subtypes and stages

  • Functional dependency studies:

    • Perform KDM3A knockdown/knockout in multiple cancer cell lines

    • Evaluate effects on proliferation, apoptosis, migration, and invasion

    • Test in both conventional and therapy-resistant models

    • Validate in 3D organoid cultures and xenograft models

  • Mechanistic understanding:

    • Identify KDM3A-regulated oncogenic pathways through integrated genomic analyses

    • Determine if KDM3A regulates established cancer hallmarks

    • Evaluate effects on therapy response genes

    • Assess synthetic lethality interactions

  • 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.

How can researchers resolve contradictory findings about KDM3A function across different studies?

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

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.