ZNF423 antibody targets the ZNF423 protein, a nuclear transcription factor with 30 C2H2 zinc finger domains that regulate DNA binding and protein interactions . ZNF423 modulates signaling pathways such as BMP/SMAD, retinoic acid receptor (RAR), and EBF1, affecting neurogenesis, adipogenesis, and cancer progression .
Prognostic Marker: In ETV6-RUNX1-negative ALL, high ZNF423 expression predicts poor survival and chemotherapy resistance .
Therapeutic Target: ZNF423 interacts with SMAD1/4 and RARα, suggesting potential for pathway inhibition in cancers .
Epigenetic Regulation: DNA hypomethylation upregulates ZNF423 in leukemia, linking epigenetic therapies to ZNF423 modulation .
Research priorities include:
ZNF423 antibodies are predominantly used in Western blot (WB), immunohistochemistry (IHC), and chromatin immunoprecipitation (ChIP). For WB, lysates from tissues/cells expressing ZNF423 (e.g., brain, heart, or neuroblastoma cell lines) are recommended, with validation via siRNA knockdown to confirm band specificity at ~144 kDa . In IHC, antigen retrieval protocols using citrate buffer (pH 6.0) improve epitope detection in nuclear compartments . ChIP-grade antibodies require crosslinking optimization (e.g., 1% formaldehyde for 10 min) and validation using positive controls like SMAD1/5-binding regions in BMP-responsive genes .
Specificity validation involves three complementary approaches:
Genetic knockout controls: CRISPR-Cas9-mediated ablation of ZNF423 in cell lines (e.g., SEM leukemia cells) confirms loss of signal in WB/IHC .
Isoform discrimination: ZNF423 has two major isoforms (α and β). Antibodies targeting the C-terminal region (e.g., ab216970) detect both isoforms, while N-terminal-specific antibodies distinguish them .
Cross-reactivity screens: Testing across species (e.g., mouse, rat) ensures antibodies do not bind paralogs like ZNF521 .
Neuroblastoma: SH-SY5Y cells treated with retinoic acid (RA) to study ZNF423’s role in differentiation .
Leukemia: SEM cells (KMT2A-AFF1 rearranged) for BMP2-induced SMAD1/5 signaling and ZNF423-EBF1 interactions .
Embryonic models: F9 teratocarcinoma cells for RA-responsive reporter assays (e.g., RARE-Luc) .
ZNF423’s dual functionality arises from its interaction partners:
Activation: In BMP signaling, ZNF423 binds SMAD1/5/9 to enhance transcription of BMP target genes (e.g., ID1). Co-IP assays with anti-SMAD1 antibodies and luciferase reporters under BMP-responsive promoters are used to quantify this activity .
Repression: ZNF423 inhibits EBF1-driven transcription in B-cell development by binding EBF1’s DNA-binding domain, blocking access to target sites like CD79A. FRET assays and ChIP-seq in ZNF423-overexpressing cells demonstrate reduced EBF1 occupancy .
In neuroblastoma, high ZNF423 correlates with RA sensitivity and favorable prognosis, whereas in B-cell acute lymphoblastic leukemia (ALL), overexpression predicts poor survival . To reconcile this:
Pathway-specific inhibition: Use small molecules (e.g., LDN-193189 for BMP inhibition) to isolate ZNF423’s BMP-dependent vs. RA-dependent effects .
Single-cell RNA-seq: Resolve heterogeneous ZNF423 expression within tumor subpopulations .
In vivo xenografts: Compare tumor growth in ZNF423-knockout vs. wild-type models under RA/BMP2 treatment .
Phosphoproteomics studies identify three critical PTMs:
S743 phosphorylation (by CDK2): Enhances ZNF423-SMAD1 binding in BMP signaling .
Ubiquitination at K892: Promotes proteasomal degradation; blocked by BMP2 via SMURF1 inhibition .
Acetylation at K45: Impairs EBF1 binding, assessed via co-IP with anti-acetyllysine antibodies .
Co-IP buffers: Use RIPA buffer with 300 mM NaCl to disrupt weak ZNF423-EBF1 interactions .
ChIP-seq peak calling: Combine MACS2 with manual curation to filter non-specific zinc finger binding sites .
Statistical thresholds: For RNA-seq, apply |log2FC| > 1.5 and FDR < 0.01 to identify ZNF423-regulated genes .