ZNF32 Human

Zinc Finger Protein 32 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Autophagy Regulation

ZNF32 inhibits autophagy initiation in breast cancer cells by activating the AKT/mTOR pathway, thereby reducing autophagosome formation (LC3-II expression) and protecting cells from excessive autophagy-induced death . Key findings:

  • Mechanism: ZNF32 overexpression suppresses Beclin-1 expression and enhances mTOR activity, blocking autophagosome formation .

  • Outcome: Protects cancer cells under oxidative stress (e.g., H₂O₂ or diamide exposure) .

Oxidative Stress Response

ZNF32 maintains mitochondrial membrane potential and enhances antioxidant capacity by upregulating C1QBP, a mitochondrial protein critical for ROS detoxification .

  • Regulation: Sp1 transcriptionally activates ZNF32 under moderate ROS levels, but repression occurs at cytotoxic ROS concentrations .

Cancer Progression

ZNF32 is upregulated in multiple cancers (e.g., breast, head and neck, hepatocellular carcinoma) and correlates with poor prognosis . Functional roles include:

  • Proliferation: Promotes cell cycle progression (S-phase enrichment) and colony formation .

  • Metastasis: Enhances migration and invasion in head and neck squamous cell carcinoma (HNSC) via CTLA-4 and PD-L1 upregulation .

Pan-Cancer Expression and Prognostic Value

Analysis of 33 cancer types from TCGA and GTEx reveals ZNF32’s differential expression and prognostic significance :

Immunotherapeutic Implications

ZNF32 influences tumor immune microenvironments by modulating immune checkpoint markers and immune cell infiltration :

  • Immune Checkpoints: Overexpression increases PD-L1 and CTLA-4 levels in HNSC cells .

  • Immune Infiltration: Positively correlates with naïve CD8+ T cells and effector memory T cells .

  • Therapeutic Response: High ZNF32 expression predicts better immunotherapy outcomes in NSCLC and renal cell carcinoma .

Diagnostic Biomarker

ZNF32’s overexpression in tumors (e.g., BRCA, HNSC) and association with advanced stages support its utility as a prognostic biomarker .

Therapeutic Target

  • Small-molecule inhibitors: Targeting ZNF32’s interaction with AKT/mTOR or immune checkpoints could reverse its pro-tumor effects .

  • Combination therapies: Pairing ZNF32 suppression with ROS-inducing agents may enhance chemotherapy efficacy .

Research Gaps and Future Directions

  • Mechanistic Insights: ZNF32’s downstream targets beyond C1QBP and mTOR remain poorly characterized .

  • Therapeutic Validation: Preclinical models are needed to test ZNF32-targeted therapies in vivo.

Product Specs

Introduction
Zinc Finger Protein 32 (ZNF32) is a member of the krueppel C2H2-type zinc-finger protein family. ZNF32 contains seven C2H2-type zinc fingers and is involved in transcriptional regulation.
Description
Recombinant human ZNF32 protein was expressed in E. coli. It is a single, non-glycosylated polypeptide chain containing 296 amino acids (amino acids 1-273) with a molecular weight of 33.4 kDa. ZNF32 is fused to a 23 amino acid His-tag at the N-terminus and purified by proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile filtered solution.
Formulation
ZNF32 protein is supplied in a solution at a concentration of 1 mg/ml with 20 mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store at -20°C. It is recommended to add a carrier protein (0.1% HSA or BSA) for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85% by SDS-PAGE analysis.
Synonyms
KOX30, Zinc finger protein 32, C2H2-546, Zinc finger protein KOX30, ZNF32.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMFGFPTA TLLDCHGRYA QNVAFFNVMT EAHHKYDHSE ATGSSSWDIQ NSFRREKLEQ KSPDSKTLQE DSPGVRQRVY ECQECGKSFR QKGSLTLHER IHTGQKPFEC THCGKSFRAK GNLVTHQRIH TGEKPYQCKE CGKSFSQRGS LAVHERLHTG QKPYECAICQ RSFRNQSNLA VHRRVHSGEK PYRCDQCGKA FSQKGSLIVH IRVHTGLKPY ACTQCRKSFH TRGNCILHGK IHTGETPYLC GQCGKSFTQR GSLAVHQRSC SQRLTL.

Q&A

What is ZNF32 and where is it located in the human genome?

ZNF32 (zinc finger protein 32) is a transcription factor belonging to the Krüppel family. It is also known by several synonyms including C2H2-546 and KOX30. The ZNF32 gene maps to chromosome region 10q23-q24 in humans. As a zinc finger protein, it contains DNA-binding domains that enable sequence-specific interactions with DNA, allowing it to regulate the expression of target genes. The protein was initially characterized alongside other zinc finger proteins in efforts to map human genes encoding this important class of transcriptional regulators .

What are the known structural characteristics of ZNF32?

ZNF32 contains C2H2-type zinc finger domains, which are the most common DNA-binding motifs found in human transcription factors. These domains consist of approximately 30 amino acid residues with conserved cysteine and histidine residues that coordinate a zinc ion, creating a finger-like projection that interacts with DNA. The specific arrangement of these domains determines the DNA sequence recognition properties of ZNF32, though detailed structural analyses through crystallography or NMR are not yet available in the literature. Understanding these structural features is essential for investigating how ZNF32 recognizes its target genes .

How does ZNF32 function as a transcription factor in normal cellular processes?

In normal cellular contexts, ZNF32 appears to play roles in regulating genes involved in fundamental processes like proliferation, cell cycle control, and stress response. The mouse homologue of ZNF32, Zfp637, has been shown to increase mTERT expression and telomerase activity, contributing to telomere length maintenance . Additionally, ZNF32 has been implicated in protecting cells against oxidative stress-induced apoptosis through regulation of C1QBP transcription . These functions suggest ZNF32 may have important roles in cellular homeostasis and stress response, though its normal physiological roles remain incompletely characterized compared to its functions in cancer contexts.

How does ZNF32 expression differ across various cancer types?

Comprehensive pan-cancer analyses have revealed that ZNF32 is highly expressed in multiple cancer types. Studies utilizing databases such as TCGA, CCLE, and others have shown significant ZNF32 upregulation in adrenocortical carcinoma (ACC), breast cancer (BRCA), and other cancer types . This consistently elevated expression across different malignancies suggests ZNF32 may play a common oncogenic role across cancer types, making it a potential biomarker for cancer detection and prognosis. Researchers investigating specific cancer types should consider examining ZNF32 expression in their experimental models to determine its relevance to their system.

What signaling pathways are affected by ZNF32 in cancer cells?

Transcriptome analysis of ZNF32 overexpression in breast cancer cells has identified several key signaling pathways affected by this transcription factor. These include:

  • Focal adhesion pathways

  • ECM-receptor interaction

  • PI3K-AKT signaling

  • HIPPO signaling

  • TNF signaling pathways

All these pathways are critically involved in cancer development and progression . The diversity of affected pathways suggests ZNF32 has broad regulatory impacts rather than influencing a single cancer-related mechanism. Researchers should consider investigating these pathways when studying the downstream effects of ZNF32 modulation in their experimental systems.

What experimental evidence demonstrates ZNF32's role in cancer cell proliferation and migration?

In vitro experiments have provided substantial evidence for ZNF32's role in promoting malignant cellular behaviors. In head and neck cancer cell lines (FaDu and CAL27), stable ZNF32 overexpression significantly enhanced:

  • Cell viability (demonstrated through CCK-8 assays)

  • Colony formation capacity

  • Cell migration (shown by Transwell migration assays)

  • Invasive abilities (revealed by Boyden invasion assays)

  • Cell cycle progression, specifically increasing the proportion of cells in S phase (determined by flow cytometry)

These functional assays collectively demonstrate that ZNF32 promotes proliferation, migration, and invasion - key hallmarks of cancer progression. Researchers studying ZNF32 should incorporate similar functional assessments to validate its effects in their cancer models of interest.

How does ZNF32 influence breast cancer progression specifically?

In breast cancer, ZNF32 has been shown to be involved in several critical processes:

  • It affects pathways crucial for cancer development including focal adhesion, ECM-receptor interaction, PI3K-AKT, HIPPO, and TNF signaling

  • It regulates multiple differentially expressed genes (DEGs) involved in cell proliferation, adhesion, and migration

  • It influences autophagy and stem cell-like properties of breast cancer cells

  • It alters the expression of 11 DEGs with fundamental changes in regulation modes, including CA9, CRLF1, and ENPP2P

Notably, 9 of these 11 DEGs contain potential transcriptional binding sequences for ZNF32 in their promoter regions, suggesting direct regulation . These findings provide a holistic perspective on ZNF32's molecular mechanisms in breast cancer progression and highlight potential downstream targets for therapeutic intervention.

What are the most effective methods for studying ZNF32 expression in experimental models?

For comprehensive analysis of ZNF32 expression and function, researchers should employ multiple complementary approaches:

  • RNA-seq for genome-wide transcriptional profiling and identification of differentially expressed genes following ZNF32 modulation

  • RT-qPCR for targeted validation of expression changes in ZNF32 and potential target genes

  • Western blotting for protein-level confirmation of ZNF32 expression changes

  • Immunohistochemistry for tissue-specific expression analysis

  • Bioinformatic analyses using databases such as TCGA, CCLE, TIMER2.0, KM-Plotter, cBioPortal, and ImmuCellAI for understanding expression patterns across cancer types and correlation with clinical parameters

When using bioinformatics approaches, researchers should exercise caution regarding data reduction techniques, as some common tools like the Jaccard estimator and MinHash estimator may introduce inaccuracies and inconsistencies in analysis results .

How can researchers establish reliable ZNF32 overexpression or knockdown models?

Based on published research, effective approaches for manipulating ZNF32 expression include:

  • Generation of stable ZNF32 overexpressing cell lines using lentiviral or plasmid vector systems, as demonstrated in head and neck cancer (FaDu and CAL27) and breast cancer cell lines

  • For validation, researchers should confirm expression changes at both mRNA (via RT-qPCR) and protein levels

  • Include appropriate vector-only controls for overexpression studies to account for non-specific effects

  • Consider developing inducible expression systems for temporal control of ZNF32 expression

  • For knockdown studies, siRNA, shRNA, or CRISPR-Cas9 approaches would be appropriate

These models provide the foundation for investigating ZNF32's functional roles and downstream effects in cancer cells.

What functional assays should be included in a comprehensive analysis of ZNF32 in cancer?

A thorough functional investigation of ZNF32 should include multiple assays addressing different aspects of cancer cell behavior:

  • Proliferation assays:

    • CCK-8 assay for cell viability assessment

    • Colony formation assay for clonogenic potential

    • Flow cytometry for cell cycle analysis and apoptosis assessment

  • Migration and invasion assays:

    • Transwell migration assay

    • Boyden invasion assay

    • Wound healing/scratch assay

  • Molecular analysis:

    • RNA-seq for global transcriptional changes

    • ChIP-seq to identify direct binding targets of ZNF32

    • Co-immunoprecipitation to identify protein interaction partners

These complementary approaches allow researchers to build a comprehensive understanding of ZNF32's functional impacts in cancer cells.

How does ZNF32 expression correlate with tumor immune characteristics?

Analysis of ZNF32 expression across cancer types has revealed significant correlations with various immune parameters:

  • Immune cell infiltration patterns

  • Microsatellite instability (MSI)

  • Tumor mutational burden (TMB)

  • Expression of immune checkpoint genes

These correlations suggest ZNF32 may play a role in shaping the tumor immune microenvironment, potentially affecting immunosurveillance and response to immunotherapy. The specific mechanisms underlying these correlations warrant further investigation to determine whether they represent causal relationships or simply associations.

What is the relationship between ZNF32 and immune checkpoint molecules?

Experimental evidence has demonstrated that ZNF32 overexpression significantly enhances the expression of key immune checkpoint molecules:

  • Increased CTLA-4 expression in ZNF32-overexpressing cells

  • Enhanced PD-L1 expression in ZNF32-overexpressing cells

  • No statistically significant effect on PD-L2 expression

These findings suggest ZNF32 may contribute to tumor immune evasion by upregulating inhibitory checkpoint molecules. This relationship has important implications for cancer immunotherapy, as it suggests that targeting ZNF32 might potentially enhance the efficacy of immune checkpoint inhibitors by reducing the expression of these inhibitory molecules.

How might researchers investigate ZNF32's role in immune modulation experimentally?

To explore ZNF32's immune regulatory functions, researchers should consider:

  • Co-culture experiments with cancer cells (with modulated ZNF32 expression) and immune cells to assess functional impacts on immune cell activation and function

  • Flow cytometric analysis of immune checkpoint molecule expression following ZNF32 modulation

  • In vivo studies using immunocompetent mouse models to evaluate how ZNF32 manipulation affects tumor immune infiltration

  • Analysis of cytokine production and signaling in response to ZNF32 modulation

  • Correlation studies between ZNF32 expression and response to immune checkpoint inhibitor therapy in patient cohorts

These approaches would provide mechanistic insights into how ZNF32 influences tumor-immune interactions and potentially identify strategies to target this pathway therapeutically.

What are the target genes directly regulated by ZNF32?

Transcriptome analysis in breast cancer cells has identified several potentially direct ZNF32 targets:

  • Among 11 differentially expressed genes (DEGs) showing fundamental changes in regulation following ZNF32 overexpression, 9 contained potential ZNF32 binding sequences in their promoter regions

  • These include genes such as CA9, CRLF1, and ENPP2P

  • These genes are involved in processes including cell proliferation, adhesion, and migration

To definitively identify direct ZNF32 targets, researchers should:

  • Perform ChIP-seq analysis to map genome-wide binding sites

  • Validate binding through reporter assays using wild-type and mutated promoter constructs

  • Employ techniques like CUT&RUN or CUT&Tag for more precise transcription factor binding site identification

How does ZNF32 contribute to drug resistance mechanisms in cancer?

ZNF32 has been implicated in multidrug resistance in lung adenocarcinoma, though the specific mechanisms require further investigation . Several potential mechanisms through which ZNF32 might contribute to drug resistance include:

  • Regulation of autophagy, which can protect cancer cells from therapy-induced cell death

  • Protection against oxidative stress-induced apoptosis via regulation of C1QBP transcription

  • Modulation of PI3K-AKT signaling, a pathway frequently involved in drug resistance mechanisms

  • Potential influence on cancer stem cell-like properties, which are associated with therapeutic resistance

Researchers investigating ZNF32's role in drug resistance should consider experimental approaches such as:

  • Comparing drug sensitivity profiles between ZNF32-high and ZNF32-low cancer cells

  • Analyzing changes in resistance-associated pathways following ZNF32 modulation

  • Evaluating combination approaches targeting ZNF32 alongside standard chemotherapeutics

What methodological challenges must researchers address when studying ZNF32?

Several important methodological considerations should guide ZNF32 research:

  • Data reduction challenges:

    • Caution is needed when using bioinformatics data sketching techniques like the Jaccard estimator and MinHash estimator, which can introduce inaccuracies

    • The minimizer Jaccard estimator has been shown to be biased and inconsistent, meaning estimates remain inaccurate regardless of sample size

  • Experimental design considerations:

    • Include appropriate controls for all experimental manipulations

    • Validate findings across multiple cell lines or models to ensure generalizability

    • Consider the temporal dynamics of ZNF32 regulation and function

  • Human subjects research compliance:

    • Ensure adherence to NIH policies and regulations for human subjects research

    • Obtain appropriate ethical approvals and informed consent

    • Consider inclusion policies for diverse populations in clinical studies

Addressing these methodological challenges is essential for producing robust, reproducible findings on ZNF32 function in cancer.

What are the most promising therapeutic applications of ZNF32 research?

Based on current knowledge of ZNF32 function, several therapeutic directions warrant investigation:

  • Development of strategies to inhibit ZNF32 expression or function in cancers where it promotes malignant behavior

  • Exploration of ZNF32 as a biomarker for patient stratification, particularly for immunotherapy response given its connections to immune checkpoint molecules

  • Investigation of combination approaches targeting ZNF32 alongside immune checkpoint inhibitors

  • Identification of synthetic lethal interactions with ZNF32 that could be therapeutically exploited

These approaches could potentially translate ZNF32 research findings into clinical applications for cancer treatment.

What key questions remain unanswered about ZNF32's role in human biology and disease?

Despite recent progress, several important questions about ZNF32 remain to be addressed:

  • What are the physiological roles of ZNF32 in normal tissues, and how does its dysregulation contribute to cancer initiation?

  • What is the complete set of direct transcriptional targets of ZNF32 across different cell types?

  • How does ZNF32 interact with other transcription factors and epigenetic regulators to influence gene expression?

  • What post-translational modifications regulate ZNF32 function?

  • How does ZNF32 contribute to tumor-stromal interactions beyond immune regulation?

Addressing these questions will provide a more complete understanding of ZNF32 biology and its potential as a therapeutic target.

Product Science Overview

Introduction

Zinc Finger Protein 32 (ZNF32) is a member of the zinc finger protein family, which is the largest family of transcription factors in the human genome. These proteins are characterized by their zinc finger domains, which are finger-like structures that bind to DNA and play a crucial role in gene regulation .

Structure

ZNF32 contains multiple zinc finger motifs, typically of the Cys2His2 type, which is the most common zinc finger motif. Each zinc finger domain consists of approximately 30 amino acids and forms a compact, spherical structure stabilized by a zinc ion. The zinc ion is coordinated by cysteine and histidine residues, which are essential for the structural integrity and function of the protein .

Function

ZNF32 functions primarily as a transcription factor, meaning it binds to specific DNA sequences and regulates the transcription of genes. This regulation is vital for various biological processes, including cell growth, differentiation, and apoptosis. ZNF32 is involved in the regulation of transcription by RNA polymerase II, which is responsible for transcribing most of the genes in eukaryotes .

Biological Significance

Zinc finger proteins, including ZNF32, play significant roles in numerous biological processes. They are involved in metabolism, autophagy, immune responses, and the maintenance and differentiation of stem cells. Due to their diverse functions, zinc finger proteins are implicated in various diseases, particularly cancers. For instance, alterations in zinc finger proteins have been associated with hepatocellular carcinoma, a common type of liver cancer .

Recombinant ZNF32

Recombinant ZNF32 refers to the protein produced through recombinant DNA technology. This involves inserting the gene encoding ZNF32 into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant ZNF32 is used in research to study its structure, function, and role in disease. It also has potential therapeutic applications, such as in gene therapy and as a target for drug development .

Research and Applications

Research on ZNF32 and other zinc finger proteins is ongoing, with a focus on understanding their roles in health and disease. Artificial zinc finger proteins are being designed for various applications, including gene editing and the development of new treatments for diseases. The unique structural and functional properties of zinc finger proteins make them valuable tools in biotechnology and medicine .

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 2024 Thebiotek. All Rights Reserved.