ZNF346 preferentially binds double-stranded RNA (dsRNA) or RNA-DNA hybrids, distinguishing it from typical DNA-binding zinc finger proteins . This activity is linked to:
Cell Growth Regulation: Modulates apoptosis and survival pathways .
Neuronal Protection: Inhibits cell cycle re-entry in neurons by upregulating p21 expression .
Hepatitis B virus (HBV) infection correlates with elevated ZNF346 expression in hepatocellular carcinoma (HCC), suggesting a role in viral oncogenesis .
ZNF346 serves as a prognostic biomarker in HCC, with overexpression linked to aggressive disease:
Multivariate Cox regression identifies ZNF346 as an independent risk factor for HCC (p < 0.05) .
ZNF346 is highly conserved across vertebrates, with orthologs in zebrafish (znf346), chickens, cats, dogs, and rodents . Functional studies in zebrafish confirm dsRNA-binding activity and nuclear localization .
Key reagents for studying ZNF346 include:
Antibodies:
Recombinant Protein: N-terminal His-tagged human ZNF346 (Purity: >90%; Bio-Techne Catalog #NBP2-22956) .
While ZNF346’s role in RNA metabolism and apoptosis is established, its mechanisms in HBV-associated HCC and neuronal protection warrant further exploration. Targeting ZNF346 could offer therapeutic avenues for cancers and neurodegenerative disorders.
Human ZNF346 is a 294 amino acid protein expressed from the ZNF346 gene. The full-length recombinant protein (ab171689) has a purity of >90% when expressed in Escherichia coli . ZNF346 contains multiple zinc finger domains that enable its nucleic acid binding capabilities. The amino acid sequence begins with MGSSHHHHHHSSGLVPRGSHM and continues through a series of zinc finger motifs essential for its molecular function . This protein is classically identified as a nuclear protein involved in RNA interactions.
While the search results don't provide direct information about evolutionary conservation, the functional importance of ZNF346 suggests conservation among vertebrates. Researchers investigating evolutionary aspects should perform comparative genomic analyses across species to determine conservation patterns of functional domains, particularly the zinc finger motifs responsible for RNA binding.
The Human Protein Atlas provides comprehensive data on ZNF346 expression across 44 normal human tissue types through immunohistochemistry-based profiling . Expression patterns vary across tissues, with both mRNA and protein level data available. Researchers can access tissue-specific expression data through the Tissue resource in the Human Protein Atlas, which offers visual representation of expression levels with reliability indicators based on available RNA-seq data and antibody-based profiling .
ZNF346 shows significant overexpression in hepatocellular carcinoma (HCC) compared to adjacent normal tissues. Analysis of 371 HCC samples and 160 normal tissues from TCGA and GTEx databases demonstrated significantly higher expression in tumor tissues . This overexpression was further validated in paired tumor and adjacent normal samples from 50 HCC patients, and confirmed at the protein level in HCC cell lines (HepG2) versus normal liver cells (LO2) .
For protein-level detection, Western blot analysis using validated antibodies is effective, as demonstrated in the comparison of ZNF346 levels between normal liver cells and HCC cell lines . For mRNA quantification, RT-qPCR remains the gold standard for targeted analysis, while RNA-seq provides a comprehensive view of expression in relation to other genes. When conducting knockdown experiments, researchers should validate efficiency at both mRNA level via RT-qPCR and protein level via Western blot, as demonstrated in HepG2 cells treated with si-ZNF346 .
Functional experiments demonstrate that ZNF346 promotes both proliferation and migration in hepatocellular carcinoma. Knockdown of ZNF346 using siRNA in HepG2 cells significantly inhibited cell proliferation as measured by CCK-8 assay . Migration capabilities were assessed through both wound healing and transwell assays, with results showing significantly reduced migration capacity in ZNF346-knockdown cells compared to control groups . These findings establish ZNF346 as a positive regulator of cancer cell proliferation and migration.
Gene Set Enrichment Analysis (GSEA) reveals that high ZNF346 expression is associated with specific biological pathways in HCC. While the search results don't detail all specific pathways, functional enrichment analyses demonstrate that ZNF346-related differentially expressed genes (DEGs) are involved in key cellular processes . The interaction between ZNF346 and these pathways provides potential therapeutic targets for intervention strategies in cancer treatment.
ZNF346 expression shows significant positive correlations with markers of immunosuppressive cells in the tumor microenvironment. Specifically, ZNF346 correlates with regulatory T cell (Treg) markers including FOXP3 (r=0.180, p=7.87×10⁻⁴), CCR8 (r=0.333, p=2.16×10⁻¹⁰), STAT5B (r=0.376, p=4.83×10⁻¹³), and TGFB (r=0.311, p=3.67×10⁻⁹) . It also correlates with cancer-associated fibroblast (CAF) markers like FAP (r=0.260, p=9.76×10⁻⁷), PDGFRα (r=0.123, p=2.25×10⁻²), and PDGFRβ (r=0.213, p=6.81×10⁻⁵) . These correlations suggest ZNF346 contributes to an immunosuppressive tumor microenvironment.
ZNF346 expression levels can predict resistance to immunotherapy. Patients with high ZNF346 expression demonstrate higher Tumor Immune Dysfunction and Exclusion (TIDE) scores and T-cell exclusion scores, both established predictors of immunotherapy resistance . Clinical validation in immunotherapy cohorts confirms that high ZNF346 expression correlates with poorer survival outcomes in patients receiving immune checkpoint inhibitors . This predictive value positions ZNF346 as a potential biomarker for immunotherapy response stratification.
Receiver Operating Characteristic (ROC) curve analysis demonstrates the diagnostic potential of ZNF346 in distinguishing HCC from normal liver tissue. The area under the curve (AUC) values indicate good discriminatory ability, making ZNF346 a promising diagnostic biomarker . For optimal diagnostic performance, researchers should consider combining ZNF346 with other established biomarkers in a diagnostic panel.
Escherichia coli serves as an effective expression system for recombinant human ZNF346 protein. The commercially available recombinant protein (ab171689) is expressed in E. coli with >90% purity, making it suitable for SDS-PAGE and mass spectrometry applications . For researchers seeking to produce their own recombinant ZNF346, the E. coli system offers an accessible and efficient method with established protocols for purification and quality control.
RNA interference via small interfering RNA (siRNA) provides an efficient method for ZNF346 knockdown in cellular models. The effectiveness of si-ZNF346 in reducing target expression should be validated at both mRNA (RT-qPCR) and protein (Western blot) levels, as demonstrated in HepG2 cells . For stable knockdown or knockout models, researchers might consider CRISPR-Cas9 genome editing approaches, though specific protocols for ZNF346 editing were not provided in the search results.
A comprehensive experimental design should include:
Knockdown validation: RT-qPCR and Western blot to confirm target reduction
Proliferation assays: CCK-8 assay with measurements at multiple time points (24, 48, 72 hours)
Migration assessment: Both wound healing assay (for collective migration) and transwell assay (for single-cell migration)
Control groups: Negative control siRNAs (NC) that do not target any genes
Statistical analysis: Multiple biological replicates with appropriate statistical tests
This design has successfully demonstrated ZNF346's promotion of proliferation and migration in HCC cells .
The high affinity of ZNF346 for dsRNA without sequence specificity suggests it may interact with various structured RNAs including non-coding RNAs. Researchers should investigate whether ZNF346 regulates specific oncogenic or tumor-suppressive RNAs through its binding activity. Techniques such as CLIP-seq (Crosslinking Immunoprecipitation followed by sequencing) could identify the RNA targets of ZNF346 in cancer cells, potentially revealing novel regulatory mechanisms.
While correlations between ZNF346 expression and immunosuppressive cell markers have been established , the causal mechanisms remain unclear. Advanced research should explore whether ZNF346 directly regulates genes involved in immune cell recruitment or function, or whether it affects signaling pathways that modulate the immune microenvironment. Single-cell RNA-seq of tumors with varying ZNF346 expression could provide insights into cell-type-specific effects.
Given that high ZNF346 expression predicts poor immunotherapy response , investigating the effects of ZNF346 inhibition on immunotherapy efficacy represents a promising research direction. Combination therapy experiments in animal models, where ZNF346 knockdown/inhibition is paired with immune checkpoint inhibitors, could reveal potential synergistic effects. This approach may overcome resistance mechanisms and improve outcomes in previously non-responsive tumors.
Cell Type | Gene Marker | Correlation | p-Value |
---|---|---|---|
Treg | FOXP3 | 0.180 | 7.87 × 10⁻⁴ |
Treg | CCR8 | 0.333 | 2.16 × 10⁻¹⁰ |
Treg | STAT5B | 0.376 | 4.83 × 10⁻¹³ |
Treg | TGFB | 0.311 | 3.67 × 10⁻⁹ |
CAF | FAP | 0.260 | 9.76 × 10⁻⁷ |
CAF | PDGFRα | 0.123 | 2.25 × 10⁻² |
CAF | PDGFRβ | 0.213 | 6.81 × 10⁻⁵ |
The table demonstrates significant positive correlations between ZNF346 expression and markers of immunosuppressive cells in the HCC tumor microenvironment .
ZNF346 is a zinc finger protein that preferentially binds to double-stranded RNA (dsRNA) or RNA/DNA hybrids, rather than DNA alone . The zinc finger domains are essential not only for dsRNA binding but also for the nucleolar localization of the protein . This binding specificity suggests that ZNF346 plays a significant role in RNA metabolism and cellular processes involving RNA.
ZNF346 is involved in various cellular processes, including cell growth and survival . It has been shown to bind with low affinity to double-stranded DNA (dsDNA) and single-stranded RNA (ssRNA), but with high affinity to dsRNA, indicating its potential role in RNA-related functions . The protein is also implicated in the intrinsic apoptotic signaling pathway mediated by p53, a crucial protein in regulating cell cycle and apoptosis .
Research on ZNF346 has provided insights into its role in RNA binding and cellular localization. Studies have shown that mutations in the zinc finger domains affect its binding properties and localization, highlighting the importance of these domains in its function . Recombinant ZNF346 is used in various research areas, including epigenetics and nuclear signaling .