Zinc finger proteins, characterized by the C2H2-type zinc finger motif, constitute a superfamily of DNA and RNA-binding proteins that play key regulatory roles during embryogenesis . Xenopus laevis, or the African clawed frog, is a widely used model organism in developmental biology . Within this context, Recombinant Xenopus laevis Zinc finger protein 238.2 (ZNF238.2) refers to a specific zinc finger protein from Xenopus laevis produced using recombinant DNA technology.
ZNF238, also known as ZBTB18, RP58, or ZFP238, is a sequence-specific DNA-binding protein with transcriptional repression activity . It can bind to the consensus DNA sequence 5'-[AC]ACATCTG[GT][AC]-3', which contains the E box core, and may be involved in chromosome organization within the nucleus . Zinc finger proteins like ZNF238 act as transcriptional regulators and participate in chromatin assembly .
ZNF238
Zinc finger protein 238
C2H2 171
RP58
TAZ 1
ZBTB18
58 kDa repressor protein
Zinc finger protein C2H2-171
Transcriptional repressor RP58
Translin-associated zinc finger protein 1
Zinc finger and BTB domain-containing protein 18
TAZ-1
C2H2-171
| Category | Information |
|---|---|
| Gene Name | ZNF238 zinc finger protein 238 $$Homo sapiens ] |
| Official Symbol | ZNF238 |
| Gene ID | 10472 |
| mRNA Refseq | NM_006352 |
| Protein Refseq | NP_006343 |
| MIM | 608433 |
| UniProt ID | Q99592 |
| Chromosome Location | 1q44-ter |
| Function | DNA binding; metal ion binding; sequence-specific DNA binding; sequence-specific DNA binding transcription factor activity; zinc ion binding |
Zinc finger proteins are known to have important regulatory functions in embryogenesis . A study has identified a large collection of C2H2-type zinc finger proteins from Xenopus laevis, some of which are highly conserved and differentially expressed during the embryonic development of the central nervous system .
The ZBTB18 gene, which encodes ZNF238, is located on chromosome 1q44 . Variants in this gene are associated with autosomal dominant intellectual disability, referred to as mental retardation 22 (MRD22) . Almost all patients with ZBTB18-related conditions exhibit some degree of intellectual disability (100%) and language retardation (96.8%) . Many also have dysmorphic facial features (60%), corpus callosum abnormalities (58.8%), and ataxia (57.9%) .
Intellectual disability
Language retardation
Dysmorphic facial features (e.g., prominent forehead, hypertelorism, flat nasal bridge, epicanthal folds, round face, and malformed low-set ears)
Corpus callosum abnormalities
Ataxia
Epilepsy
Microcephaly
Growth retardation
Eye diseases (myopia, strabismus, and visual impairment)
Gastrointestinal problems
Bruxism
Clinodactyly of the 5th finger
Abnormality of hair or hair pattern
Cerebellar tonsillar hypoplasia
Febrile seizure
Recombinant Xenopus laevis Zinc finger protein 238.2 (znf238.2)
This transcriptional repressor plays a crucial role in various developmental processes. It exhibits specific binding to the consensus DNA sequence 5'-[AC]ACATCTG[GT][AC]-3', encompassing the E box core. Its mechanism of action involves recruiting chromatin remodeling multiprotein complexes.
KEGG: xla:379322
UniGene: Xl.23523
Recombinant Xenopus laevis Zinc finger protein 238.2 (znf238.2) is a specific zinc finger protein from the African clawed frog produced using recombinant DNA technology. It belongs to the C2H2-type zinc finger motif superfamily of DNA and RNA-binding proteins that play key regulatory roles during embryogenesis. This protein, also known as ZBTB18, RP58, or ZFP238, functions as a sequence-specific DNA-binding protein with transcriptional repression activity. It recognizes and binds to the consensus DNA sequence 5'-[AC]ACATCTG[GT][AC]-3', which contains the E box core, and is likely involved in chromosome organization within the nucleus. The protein participates in chromatin assembly and acts as a transcriptional regulator, contributing to the complex genetic control mechanisms during development.
Characterizing znf238.2 binding specificity requires multiple complementary approaches:
Electrophoretic Mobility Shift Assays (EMSA): Use purified recombinant znf238.2 with synthesized oligonucleotides containing the consensus sequence 5'-[AC]ACATCTG[GT][AC]-3' to confirm direct binding . Compare binding affinity to mutated sequences to map critical nucleotides.
Chromatin Immunoprecipitation (ChIP): Implement ChIP assays using specific antibodies against znf238.2 followed by sequencing to identify genome-wide binding sites in Xenopus embryonic tissues.
DNase I Footprinting: This technique can precisely map the protected regions where znf238.2 binds to DNA, providing nucleotide-level resolution of binding sites.
Systematic Evolution of Ligands by Exponential Enrichment (SELEX): This iterative method can identify high-affinity binding sequences from random oligonucleotide pools, helping refine our understanding of znf238.2 binding motifs.
Surface Plasmon Resonance (SPR): Quantify binding kinetics and affinity constants for znf238.2 interaction with various DNA sequences.
The combined data from these approaches will provide comprehensive binding specificity profiles and reveal potential cooperative binding effects with other transcription factors.
Distinguishing znf238.2 from other zinc finger proteins in Xenopus laevis can be challenging given that the C2H2-type zinc finger proteins are encoded by a multigene family comprising several hundred members . Consider these methodological approaches:
Structural characteristics: znf238.2 contains specific C2H2-type zinc finger motifs that bind DNA in a sequence-specific manner. Unlike dsRBP-ZFa, which primarily binds dsRNA, znf238.2 preferentially binds DNA sequences containing the E box core .
Specific domains: Focus on regions outside the zinc finger motifs, as ZFPs can be subdivided into distinct subfamilies based on conserved sequence features in these regions . Use antibodies targeting the unique N-terminal domains for immunoprecipitation experiments.
DNA binding specificity: Perform competitive binding assays with labeled consensus sequences. znf238.2 binds specifically to 5'-[AC]ACATCTG[GT][AC]-3', which can distinguish it from other ZFPs with different sequence preferences.
Phosphorylation patterns: Take advantage of the differential phosphorylation patterns. Unlike the FAR-ZFP subfamily proteins, which are phosphorylated by CK II in their N-terminal domains, znf238.2 may exhibit distinct phosphorylation characteristics .
Mass spectrometry analysis: Implement targeted proteomics approaches to identify unique peptide signatures that distinguish znf238.2 from other zinc finger family members.
The choice of expression system significantly impacts the yield and functionality of recombinant znf238.2:
Bacterial expression systems (E. coli): While cost-effective and high-yielding, bacterial systems often produce insoluble zinc finger proteins due to improper folding in the absence of appropriate post-translational modifications. If attempting bacterial expression, consider:
Using specialized strains like Rosetta(DE3) that supply rare codons
Expressing as a fusion with solubility tags (MBP, SUMO, or TRX)
Including zinc in the growth medium (0.1-0.5 mM ZnCl₂) to facilitate proper folding
Lowering induction temperature to 16-18°C to reduce inclusion body formation
Insect cell expression (Baculovirus): This system provides superior folding and post-translational modifications compared to bacteria:
The Sf9 or High Five™ insect cell lines are recommended
Co-express with chaperones to improve proper folding
Include 0.1 mM ZnSO₄ in the media during protein expression
Optimize infection MOI and harvest time (typically 48-72 hours post-infection)
Xenopus oocyte expression: For the most native-like protein:
Microinject capped znf238.2 mRNA into Xenopus oocytes
Include the required conserved N-terminal domains for proper folding
Extract protein under native conditions with mild detergents
Verify zinc incorporation using atomic absorption spectroscopy
Each system offers distinct advantages, but insect cell expression typically provides the best balance between yield and functional protein quality for zinc finger proteins like znf238.2.
Successful purification of functional znf238.2 requires careful attention to maintaining protein stability throughout the process:
Always include zinc ions (0.05-0.1 mM ZnCl₂) in all purification buffers to maintain finger structure
Use reducing agents (1-5 mM DTT or 2-10 mM β-mercaptoethanol) to prevent oxidation of cysteine residues
Include glycerol (10-20%) to enhance stability
Maintain pH between 7.0-8.0 to preserve zinc coordination
Add protease inhibitors to prevent degradation
For affinity purification, use mild elution conditions to prevent protein unfolding
During ion exchange chromatography, use shallow gradients to separate properly folded protein from misfolded species
Consider size exclusion chromatography as a final polishing step
Avoid freeze-thaw cycles; store purified protein at 4°C for short-term use
Verify zinc content using atomic absorption spectroscopy
Confirm proper folding using circular dichroism spectroscopy
Test DNA binding activity with EMSA before and after each purification step
Monitor protein homogeneity by dynamic light scattering
Evidence suggests that zinc finger domains require physiological zinc concentrations for structural stability, as RNA binding by zinc finger proteins like Zar2 requires the presence of Zn²⁺ and conserved cysteines in the C-terminal domain .
Verification of functional recombinant znf238.2 requires multiple assays targeting different aspects of its activity:
Electrophoretic Mobility Shift Assay (EMSA): The gold standard for confirming DNA binding. Use oligonucleotides containing the consensus sequence 5'-[AC]ACATCTG[GT][AC]-3'. A positive result shows retarded migration of the protein-DNA complex.
Fluorescence Anisotropy: Measure real-time binding kinetics using fluorescently labeled target DNA sequences.
Filter Binding Assay: A quantitative approach to determine binding affinity constants.
Reporter Gene Assay: Transfect cells with a reporter construct containing znf238.2 binding sites upstream of a luciferase gene. Co-transfect with znf238.2 expression vector and measure repression of luciferase activity.
In vitro Transcription Assay: Use a cell-free transcription system with templates containing znf238.2 binding sites to assess direct repression of transcription.
Circular Dichroism (CD): Confirm proper secondary structure formation.
Thermal Shift Assay: Assess protein stability through melting temperature determination.
Limited Proteolysis: Properly folded proteins show resistance to proteolytic degradation at specific sites.
Inductively Coupled Plasma Mass Spectrometry (ICP-MS): Quantify zinc content.
PAR Assay (4-(2-pyridylazo)resorcinol): A colorimetric assay for zinc binding.
A fully functional znf238.2 should demonstrate specific DNA binding with the expected affinity (nanomolar range) and exhibit transcriptional repression activity when introduced into appropriate cellular contexts.
Investigating znf238.2's role in embryonic development requires a multi-faceted approach:
Perform whole-mount in situ hybridization to map znf238.2 expression patterns throughout Xenopus embryonic development.
Use immunohistochemistry with specific antibodies to localize the protein in developing tissues.
Implement RT-qPCR to quantify expression levels during different developmental stages.
Employ morpholino oligonucleotides for targeted knockdown of znf238.2 in Xenopus embryos.
Use CRISPR/Cas9 technology for precise gene editing to create znf238.2 knockout models.
Analyze resulting phenotypes with particular attention to neuronal development, as zinc finger proteins play crucial roles in central nervous system development.
Microinject synthetic znf238.2 mRNA into specific blastomeres at early cleavage stages.
Create tissue-specific overexpression using appropriate promoters.
Examine phenotypic consequences on tissue patterning and cell differentiation.
Perform ChIP-seq at different developmental stages to identify stage-specific binding sites.
Combine with RNA-seq after znf238.2 manipulation to correlate binding with gene expression changes.
Validate individual targets using reporter gene assays.
Identify protein binding partners through co-immunoprecipitation followed by mass spectrometry.
Investigate chromatin remodeling complex interactions since znf238.2 is involved in chromosome organization.
Zinc finger proteins are known to have important regulatory functions during embryogenesis, and some are highly conserved and differentially expressed during development of the central nervous system.
Phosphorylation can significantly impact zinc finger protein function, as evidenced by studies on related proteins. While specific data on znf238.2 phosphorylation is limited, research on other Xenopus zinc finger proteins provides methodological guidance:
Phosphorylation of zinc finger proteins can alter DNA binding affinity and specificity
It may regulate nuclear-cytoplasmic shuttling
It can modify interactions with co-factors and chromatin remodeling complexes
Temporal regulation of activity during development may be phosphorylation-dependent
Identification of phosphorylation sites:
Kinase identification:
Functional impact assessment:
Site-directed mutagenesis of identified/predicted phosphorylation sites (Ser/Thr→Ala to prevent phosphorylation; Ser/Thr→Asp to mimic phosphorylation)
DNA binding assays with phosphorylated vs. non-phosphorylated protein
Reporter gene assays to assess transcriptional activity changes
Developmental regulation:
Analysis of phosphorylation changes during developmental stages
Correlation with expression patterns of relevant kinases
Phospho-specific antibody staining in developing embryos
Research has shown that zinc finger proteins of the FAR-ZFP subfamily are targets for CK II-mediated phosphorylation, and expression of these proteins during oogenesis coincides with CK II activity in unfertilized eggs . The target sites for phosphorylation are typically localized within conserved N-terminal domains rather than within the zinc finger regions themselves .
Investigating znf238.2 interactions with chromatin and nuclear factors requires specialized techniques that preserve native interactions:
ChIP-seq: The gold standard for genome-wide binding site identification. Use high-quality antibodies against znf238.2 or epitope tags if working with recombinant protein.
CUT&RUN or CUT&Tag: These techniques offer higher signal-to-noise ratios than traditional ChIP and require fewer cells.
ATAC-seq: Combined with znf238.2 manipulation to identify changes in chromatin accessibility.
HiChIP: To investigate how znf238.2 affects three-dimensional chromatin architecture.
Proximity labeling (BioID, APEX): Identify proteins in close proximity to znf238.2 in living cells.
Co-immunoprecipitation coupled with mass spectrometry: Identify stable interaction partners.
FRET/BRET assays: For studying dynamic interactions in living cells.
Yeast two-hybrid screening: To discover novel interaction partners.
Sequential ChIP (Re-ChIP): To identify co-occupancy of znf238.2 with other factors at specific genomic loci.
Chromatin fractionation: Determine association with different chromatin states (heterochromatin vs. euchromatin).
CRISPR screens: Identify genes that genetically interact with znf238.2.
FRAP (Fluorescence Recovery After Photobleaching): Analyze dynamic binding to chromatin.
Single-molecule tracking: Study real-time behavior of individual znf238.2 molecules.
These methodologies are particularly relevant as znf238.2 may be involved in chromosome organization within the nucleus and participates in chromatin assembly. Understanding these interactions will provide insight into how znf238.2 contributes to the complex regulatory networks controlling gene expression during development.
Zinc finger proteins exhibit diverse nucleic acid binding preferences, with some binding DNA (like znf238.2) and others binding RNA. The structural and functional differences between these proteins provide insights into their specialized roles:
znf238.2 binds to specific DNA sequences (5'-[AC]ACATCTG[GT][AC]-3') containing the E box core and functions as a transcriptional repressor. In contrast, RNA-binding zinc finger proteins like dsRBP-ZFa bind double-stranded RNA with nanomolar dissociation constants in a sequence-independent manner . dsRBP-ZFa prefers A-form helices and can also bind RNA-DNA hybrids .
The solution structure of RNA-binding zinc finger proteins reveals distinct features: dsRBP-ZFa has two zinc finger domains connected by an unstructured linker (24 residues), with each finger having a highly electropositive surface that maps to a helix-kink-helix motif . This represents a novel motif for double-stranded RNA binding that does not contain the previously described dsRNA binding motif .
These differences highlight how zinc finger domains have evolved specialized functions while maintaining the core C2H2 structure, allowing them to participate in diverse regulatory processes from transcriptional control to RNA metabolism.
The regulatory mechanisms of znf238.2 and Zar family zinc finger proteins represent distinct but potentially complementary systems functioning during oocyte maturation and early development:
znf238.2 functions primarily as a DNA-binding transcriptional repressor
Zar2 binds specifically to RNA, targeting the Translational Control Sequence (TCS) in maternal mRNAs like Wee1
Zar2 is present throughout oogenesis but levels decrease during oocyte maturation
While specific znf238.2 expression data during oogenesis is limited, zinc finger proteins generally show stage-specific expression
Zar2 represses translation of maternal mRNAs in immature oocytes, with this repression being relieved during oocyte maturation as Zar2 is degraded
This creates a temporal control system for maternal mRNA translation
znf238.2, as a transcriptional regulator, likely controls gene expression at the DNA level rather than post-transcriptionally
Zar2 RNA binding requires the presence of Zn²⁺ and conserved cysteines in the C-terminal domain, suggesting a zinc finger structure
znf238.2 contains C2H2-type zinc finger motifs that mediate DNA binding
RNA immunoprecipitation to determine if znf238.2 might also bind specific RNA targets
ChIP-seq to identify if Zar2 has any DNA binding activity
Co-immunoprecipitation to test for potential interactions between these proteins
Dual reporter assays to compare their respective regulatory impacts
The complementary activities - with Zar2 controlling translation of maternal mRNAs while znf238.2 potentially regulates transcription - suggest a multi-layered regulatory system during early development. This combination of transcriptional and translational control likely provides precise temporal and spatial regulation of gene expression during the critical transition from maternal to zygotic control of development.
Several cutting-edge methodologies show promise for deepening our understanding of znf238.2 function:
Single-cell RNA-seq combined with CRISPR screening: This approach can reveal cell-type-specific functions of znf238.2 during development.
Single-cell ATAC-seq: Mapping chromatin accessibility changes in individual cells after znf238.2 manipulation.
Single-cell ChIP-seq: Although technically challenging, this emerging method could map znf238.2 binding in rare cell populations.
Cryo-EM studies: To visualize znf238.2 in complex with DNA and co-factors at near-atomic resolution.
AlphaFold2 and related AI structure prediction: To model full-length znf238.2 structure, including domains not previously crystallized.
Hydrogen-deuterium exchange mass spectrometry (HDX-MS): To map conformational changes upon DNA binding or protein-protein interactions.
Base editing and prime editing: For precise introduction of specific mutations without double-strand breaks.
Epigenome editing: Using dCas9 fused to epigenetic modifiers to alter the chromatin state at znf238.2 binding sites.
Optogenetic control of znf238.2 activity: To achieve temporal precision in functional studies.
Spatial mapping of znf238.2-dependent gene expression: To correlate znf238.2 activity with tissue-specific transcriptional programs.
Multiplexed FISH techniques: To visualize multiple znf238.2 target genes simultaneously in tissue sections.
Engineered zinc finger domains: Creating synthetic znf238.2 variants with altered binding specificities.
Cell-free expression systems: For high-throughput functional screening of znf238.2 variants.
These emerging technologies will help overcome current limitations in understanding the precise roles of znf238.2 in development and disease, particularly in mapping its position within complex gene regulatory networks that control embryonic patterning and cellular differentiation.
Despite advances in understanding zinc finger protein biology, several critical questions about znf238.2 remain unresolved:
What is the complete repertoire of znf238.2 target genes during different developmental stages?
How does znf238.2 cooperate or compete with other transcription factors at shared regulatory elements?
What determines the context-specific activities of znf238.2 in different tissues?
How is znf238.2 itself regulated during development to ensure proper temporal control of target genes?
Does znf238.2 participate in maternal-to-zygotic transition regulation?
Are there feedback loops involving znf238.2 that create oscillatory gene expression patterns?
To what extent do other zinc finger proteins compensate for loss of znf238.2 function?
Are there paralogs that evolved specialized functions from a common ancestral gene?
What unique functions does znf238.2 perform that cannot be compensated by other factors?
Does znf238.2 recruit specific chromatin modifiers to establish repressive chromatin states?
How stable are znf238.2-chromatin interactions during mitosis?
Can znf238.2 function as an activator in specific contexts, despite its general classification as a repressor?
How has znf238.2 function diverged between Xenopus laevis and other vertebrates?
What selection pressures drove the evolution of the znf238.2 DNA binding specificity?
While variants in human ZBTB18 are associated with intellectual disability, what is the molecular basis for these effects?
Could znf238.2 be involved in amphibian-specific developmental processes with no mammalian parallel?
Addressing these questions will require integrative approaches combining genomics, proteomics, developmental biology, and computational modeling to place znf238.2 in its proper context within the complex regulatory networks governing vertebrate development.
When designing experiments involving znf238.2, researchers should consider several practical aspects to ensure robust and reproducible results:
Always include zinc in buffers (0.05-0.1 mM ZnCl₂) when working with recombinant znf238.2
Verify protein integrity before experiments using activity assays and structural analyses
Avoid repeated freeze-thaw cycles that may compromise zinc finger domain structure
Consider expressing fusion proteins with stability tags for improved handling
Include appropriate negative controls such as zinc finger mutants that cannot bind DNA
When performing knockdown or knockout studies, validate specificity with rescue experiments
For binding studies, include competition controls with non-specific DNA/RNA
Control for potential off-target effects when using morpholinos or CRISPR/Cas9
Remember that Xenopus laevis is pseudotetraploid with potentially redundant gene copies
Consider the developmental stage carefully when interpreting results, as zinc finger protein functions may be stage-specific
For cross-species comparisons, account for divergence in binding specificity and regulatory networks
When studying maternal proteins, differentiate between protein stored in oocytes and newly synthesized protein
Generating highly specific antibodies may be difficult due to sequence similarity with other zinc finger proteins
ChIP experiments may require optimization due to the transient nature of some znf238.2-DNA interactions
Consider using epitope tags when antibody quality is a concern, validating that tags don't disrupt function
When expressing recombinant protein, codon optimization for the expression system may improve yields
By addressing these practical considerations, researchers can design more robust experiments that advance our understanding of znf238.2 function in development and potentially reveal insights applicable to human developmental disorders associated with zinc finger protein dysfunction.