Recombinant Xenopus laevis CXXC-type zinc finger protein 4 (cxxc4)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates. Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. The tag type will be determined during the production process. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
cxxc4; idaxCXXC-type zinc finger protein 4; Inhibition of the Dvl and axin complex protein; xIdax
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-217
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
Target Protein Sequence
MHRNDSQRLG KPGGAPESLQ MANNNFLSTL SPEHCRPLAG ECMNKLKCGA AEAEIMNLPE RVGTFSAIPA LGGISLPPGV IVMTALHSPA AASAAVTDSA FQIANLADCP QNNSSGAGGN PAKKKRKRCG VCVPCKRLIN CGVCSSCRNR KTGHQICKFR KCEELKKKPG TSLEVRGDDS FFPCLASSLI PPFSPPFQCF LSPFKMHHSF SESPSRL
Uniprot No.

Target Background

Function
This protein acts as a negative regulator of the Wnt signaling pathway, crucial for anterior neural structure formation. Ectopic expression induces ventralization (PubMed:15108311, PubMed:11113207). It preferentially binds to DNA containing cytidine-phosphate-guanosine (CpG) dinucleotides compared to CpH (H=A, T, and C), hemimethylated-CpG, and hemimethylated-hydroxymethyl-CpG.
Subcellular Location
Cytoplasm.

Q&A

How do researchers distinguish CXXC-type zinc finger proteins from other zinc-binding domains in X. laevis?

Distinguishing CXXC-type zinc finger proteins requires detailed structural and sequence analysis:

  • Sequence analysis techniques: Examination of conserved CXXC motifs and their spacing patterns. Unlike canonical C4 zinc fingers, CXXC-type proteins feature distinctive cysteine arrangements.

  • Spectroscopic methods: NMR spectroscopy can reveal chemical shift deviations consistent with rubredoxin turns, which are characteristic of certain zinc finger proteins .

  • Metal content determination: ESI-MS (Electrospray Ionization Mass Spectrometry) analysis to determine the precise number of zinc atoms present, as seen in studies of other zinc finger proteins like Churchill .

  • Mutation studies: Targeted mutations of potential zinc-coordinating residues (cysteines and histidines) can help identify essential ligands. For example, in studies of Churchill, mutations such as C17S, C17A, and C17V produced varying effects on protein folding, helping to elucidate zinc coordination patterns .

  • Secondary structure analysis: CD spectroscopy and NMR can distinguish CXXC proteins from classical zinc fingers by identifying characteristic structural elements. Traditional C2H2 zinc fingers typically contain ββα motifs, while CXXC proteins may display alternative secondary structure arrangements.

What experimental approaches are most effective for purifying recombinant Xenopus laevis cxxc4 protein?

Purification of recombinant Xenopus laevis cxxc4 requires careful consideration of protein stability and zinc coordination:

Recommended purification protocol:

  • Expression system selection: E. coli BL21(DE3) or specialized strains designed for proper disulfide bond formation are preferred for zinc finger proteins.

  • Expression construct design:

    • Include an N-terminal affinity tag (His6 or GST) for initial purification

    • Consider fusion protein approaches like those used for other zinc finger proteins

    • Include a protease cleavage site for tag removal

  • Buffer optimization:

    • Maintain zinc throughout purification (typically 10-50 μM ZnCl₂ or ZnSO₄)

    • Include reducing agents (1-5 mM DTT or β-mercaptoethanol) to prevent cysteine oxidation

    • pH 7.5-8.0 is optimal for zinc finger stability

  • Purification steps:

    • Initial IMAC or GST-affinity chromatography

    • Ion exchange chromatography (typically cation exchange as zinc finger proteins tend to be basic)

    • Size exclusion chromatography as a final polishing step

  • Quality control:

    • UV-visible spectroscopy to confirm proper zinc coordination

    • Circular dichroism to verify secondary structure integrity, similar to methods used for dsRBP-ZFa

    • ESI-MS to confirm the presence of the expected number of zinc ions

How does protein phosphorylation affect the function of Xenopus laevis zinc finger proteins and how can this be studied for cxxc4?

Evidence suggests that phosphorylation plays a crucial role in regulating X. laevis zinc finger protein function. Studies on FAR and FAX domain-containing zinc finger proteins have shown they are targets for CK II-mediated phosphorylation .

Methodological approach to study phosphorylation:

  • Identification of potential phosphorylation sites:

    • In silico prediction using tools like NetPhos or GPS

    • Comparison with known phosphorylation sites in related proteins

    • Focus on conserved N-terminal domains where phosphorylation often occurs in X. laevis zinc finger proteins

  • In vitro phosphorylation assays:

    • Express recombinant cxxc4 protein

    • Incubate with purified kinases (starting with CK II based on precedent)

    • Detect phosphorylation by:

      • ³²P-ATP incorporation

      • Phospho-specific antibodies

      • Mass spectrometry

  • Site-directed mutagenesis to create phosphomimetic variants:

    • Mutate target serine/threonine residues to alanine (phospho-null)

    • Mutate target residues to glutamic acid (phosphomimetic)

    • Compare functional properties of wild-type and mutant proteins

  • Temporal analysis of phosphorylation during development:

    • Extract protein from X. laevis embryos at different developmental stages

    • Immunoprecipitate cxxc4

    • Analyze phosphorylation status

  • Functional consequences assessment:

    • DNA/RNA binding assays with phosphorylated vs. unphosphorylated protein

    • Protein-protein interaction studies before and after phosphorylation

    • Cellular localization analysis using phospho-specific antibodies

What methods are most reliable for studying the developmental expression patterns of cxxc4 in Xenopus laevis?

Understanding the developmental expression of cxxc4 requires multiple complementary approaches:

  • RNA expression analysis:

    • Semi-quantitative RT-PCR: As demonstrated for other X. laevis genes like xWnt11 and Siamois

    • Quantitative real-time RT-PCR: Provides more precise quantification of expression levels across developmental stages

    • In situ hybridization: To visualize spatial expression patterns in embryos

  • Protein expression analysis:

    • Western blotting: Using specific antibodies against cxxc4

    • Immunohistochemistry: To visualize protein localization in tissues

    • Mass spectrometry: For quantitative proteomics across developmental stages

  • Reporter gene assays:

    • Clone the cxxc4 promoter region upstream of a reporter gene

    • Inject into Xenopus embryos to monitor temporal and spatial expression patterns

    • Use deletion constructs to identify key regulatory elements

  • Transgenic approaches:

    • Generate transgenic Xenopus expressing fluorescent protein fusions

    • Monitor expression in living embryos through development

  • Expression data integration table:

    Developmental StageRNA Expression MethodProtein DetectionReporter Gene Activity
    OocyteRT-qPCRWestern blotN/A
    BlastulaRT-qPCR, ISHIHC, WesternPromoter activity
    GastrulaRT-qPCR, ISHIHC, WesternPromoter activity
    NeurulaRT-qPCR, ISHIHC, WesternPromoter activity
    TadpoleRT-qPCR, ISHIHC, WesternPromoter activity

What NMR spectroscopy approaches are most informative for characterizing the zinc coordination in cxxc4?

Based on successful structural studies of other zinc finger proteins, several NMR approaches are particularly valuable:

  • Heteronuclear NMR experiments:

    • ¹⁵N-HSQC to monitor backbone amide resonances

    • ¹³C-HSQC focused on cysteine and histidine side chains

    • Triple-resonance experiments for sequential assignment

  • Specific experiments for zinc coordination:

    • ¹H-¹⁵N HMQC spectra to identify histidine tautomeric states, which change upon zinc coordination

    • Chemical shift analysis of cysteine Cβ resonances (typically shifted downfield upon zinc coordination)

    • NOE patterns between zinc-coordinating residues

  • Dynamic measurements:

    • ¹⁵N relaxation experiments (T₁, T₂, heteronuclear NOE) to identify regions stabilized by zinc binding

    • Hydrogen-deuterium exchange to probe structural stability around zinc sites

  • Metal substitution approaches:

    • Replace Zn²⁺ with paramagnetic Co²⁺ to obtain distance constraints

    • Use EDTA titration to monitor unfolding as zinc is removed, as demonstrated for dsRBP-ZFa

  • Data processing and structure calculation:

    • Use CYANA or XPLOR-NIH with specific zinc coordination restraints

    • Validate with PROCHECK-NMR and other structure validation tools

How can researchers distinguish between DNA and RNA binding specificity in cxxc4?

Understanding nucleic acid binding preferences requires careful experimental design:

  • Comparative binding assays:

    • Gel mobility shift assays with equal concentrations of DNA and RNA substrates

    • Competition experiments with labeled and unlabeled nucleic acids

    • Measure binding constants for different substrates under identical conditions

  • Structural characterization of complexes:

    • Chemical shift perturbation mapping by NMR to identify interaction surfaces

    • Cross-linking followed by mass spectrometry to identify contact points

    • X-ray crystallography or cryo-EM of protein-nucleic acid complexes

  • Mutational analysis:

    • Target residues in potential binding surfaces (e.g., electropositive surfaces similar to those identified in dsRBP-ZFa )

    • Analyze the effect on DNA vs. RNA binding

    • Create chimeric proteins by swapping domains with proteins of known specificity

  • In vivo validation:

    • ChIP assays for DNA binding as performed for Kaiso

    • CLIP (Cross-Linking Immunoprecipitation) for RNA interactions

    • Functional studies using reporter constructs

  • Analytical framework for binding specificity:

    PropertyDNA BindingRNA BindingAssay Method
    Affinity (KD)Measure nM-μM rangeMeasure nM-μM rangeEMSA, SPR, ITC
    SpecificitySequence motifSequence/structureSELEX, binding site selection
    StructureB-form recognitionA-form/loops recognitionNMR, X-ray crystallography
    KineticsAssociation/dissociation ratesAssociation/dissociation ratesSPR, stopped-flow
    CompetitionDNA vs. RNA preferenceRNA vs. DNA preferenceCompetition EMSA

What are the recommended methods for studying cxxc4 involvement in cellular signaling pathways?

Based on studies of related zinc finger proteins, several approaches can be used to investigate cxxc4's role in signaling:

  • Gene knockdown/knockout approaches:

    • Morpholino antisense oligonucleotides for targeted knockdown

    • CRISPR/Cas9 gene editing to generate mutant Xenopus

    • Analyze phenotypic consequences on development and specific signaling pathways

  • Overexpression studies:

    • Inject cxxc4 mRNA into X. laevis embryos

    • Use tissue-specific or inducible promoters

    • Monitor effects on known signaling targets (e.g., Wnt pathway components)

  • Interaction studies:

    • Yeast two-hybrid screening to identify protein partners

    • Co-immunoprecipitation followed by mass spectrometry

    • Proximity labeling (BioID or APEX) in Xenopus cells or tissues

  • Pathway-specific reporter assays:

    • Design reporter constructs for relevant pathways (e.g., Wnt, BMP)

    • Co-express with wild-type or mutant cxxc4

    • Measure reporter activity in response to pathway stimulation

  • Integration with RNA-seq data:

    • Perform RNA-seq after cxxc4 manipulation

    • Conduct pathway enrichment analysis

    • Validate key targets by RT-qPCR, similar to approaches used for studying Kaiso targets like Siamois

How can researchers determine if cxxc4 functions primarily through chromatin modification or direct transcriptional regulation?

Distinguishing between these mechanisms requires multiple experimental approaches:

  • Chromatin association analysis:

    • ChIP-seq to map genome-wide binding sites

    • ATAC-seq to correlate binding with chromatin accessibility

    • Co-localization with histone modifications by sequential ChIP

  • Protein complex identification:

    • Immunoprecipitation coupled with mass spectrometry

    • Size exclusion chromatography to identify native complexes

    • Density gradient ultracentrifugation to separate distinct complexes

  • Biochemical activity assays:

    • In vitro transcription assays with purified components

    • Histone modification assays to test direct enzymatic activity

    • DNA methylation analysis at target sites

  • Genetic interaction studies:

    • Combined knockdown of cxxc4 with chromatin modifiers

    • Epistasis analysis with transcription factors

    • Rescue experiments with mutant forms lacking specific interaction domains

  • Microscopy approaches:

    • Super-resolution imaging of nuclear localization

    • FRAP (Fluorescence Recovery After Photobleaching) to measure dynamics

    • Live cell imaging with fluorescently tagged proteins

What are the key considerations when analyzing potential crosstalk between cxxc4 and other zinc finger proteins in X. laevis?

Investigating functional relationships between zinc finger proteins requires:

  • Expression pattern correlation:

    • Compare developmental timing of expression

    • Analyze tissue distribution using in situ hybridization

    • Quantify relative expression levels by RT-qPCR

  • Protein-protein interaction studies:

    • Direct binding assays with purified proteins

    • Co-immunoprecipitation from Xenopus extracts

    • FRET/BRET approaches for detecting interactions in live cells

  • Functional redundancy assessment:

    • Single and combined knockdown experiments

    • Rescue experiments with related proteins

    • Domain swapping to identify functional equivalence

  • Target gene overlap analysis:

    • Compare ChIP-seq or RNA-seq datasets

    • Analyze binding motifs for similarity

    • Perform competition binding assays for shared targets

  • Evolutionary relationship analysis:

    • Phylogenetic analysis of zinc finger domains

    • Compare with syntenic regions in other vertebrates

    • Analyze conservation of key functional residues

What strategies can overcome the challenges in expressing soluble, correctly folded recombinant cxxc4?

Expression of properly folded zinc finger proteins presents several challenges:

  • Optimizing expression conditions:

    • Test multiple expression strains (BL21(DE3), Rosetta, SHuffle)

    • Reduce induction temperature (16-20°C)

    • Use auto-induction media or low IPTG concentrations (0.1-0.5 mM)

    • Include zinc in growth media (10-50 μM ZnCl₂)

  • Protein fusion strategies:

    • Test multiple solubility tags (GST, MBP, SUMO, Trx)

    • Include a flexible linker between tag and cxxc4

    • Use dual affinity tags for improved purification

  • Refolding approaches if necessary:

    • Isolate inclusion bodies and solubilize in denaturants

    • Perform gradual dialysis to remove denaturants

    • Include redox buffer systems (reduced/oxidized glutathione)

    • Add zinc during refolding process

  • Quality control methods:

    • Circular dichroism to verify secondary structure, similar to methods used for dsRBP-ZFa

    • Thermal stability assays (DSF/DSC)

    • Limited proteolysis to identify folded domains

    • Dynamic light scattering to assess aggregation

  • Truncation strategies:

    • Express individual zinc finger domains

    • Design constructs based on secondary structure prediction

    • Test multiple N and C-terminal boundaries

How can researchers address the challenge of specificity when studying cxxc4 function in X. laevis embryos?

Ensuring specificity in functional studies requires several control measures:

  • Knockdown validation:

    • Use multiple non-overlapping morpholinos

    • Include control morpholinos with mismatches

    • Validate knockdown efficiency by Western blot or RT-qPCR

    • Perform rescue experiments with morpholino-resistant mRNA

  • CRISPR/Cas9 strategies:

    • Design multiple guide RNAs targeting different exons

    • Validate editing efficiency by sequencing

    • Screen for off-target effects using whole genome sequencing

    • Generate F0 mosaic embryos and F1 stable lines for comparison

  • Antibody validation:

    • Test antibody specificity against recombinant protein

    • Perform peptide competition assays

    • Validate using knockout/knockdown samples as negative controls

    • Test cross-reactivity with related zinc finger proteins

  • Target validation:

    • Confirm direct binding to targets by ChIP-qPCR

    • Use reporter constructs with wild-type and mutated binding sites

    • Perform rescue experiments with targeted gene overexpression

  • Controls for developmental timing:

    • Include stage-matched controls for all experiments

    • Monitor developmental markers to ensure proper staging

    • Design time-course experiments to capture temporal dynamics

What are the best approaches for differentiating the functions of cxxc4 from other CXXC-domain proteins in Xenopus development?

Distinguishing between related zinc finger proteins requires:

  • Domain-specific functional analysis:

    • Generate chimeric proteins by domain swapping

    • Express isolated domains and test their function

    • Perform structure-guided mutagenesis of specific residues

  • Temporal control of expression/function:

    • Use hormone-inducible protein systems (e.g., GR fusion)

    • Apply photoactivatable morpholinos for stage-specific knockdown

    • Employ optogenetic approaches for spatial-temporal control

  • Comparative genomics and transcriptomics:

    • Perform RNA-seq after manipulation of individual family members

    • Identify unique and overlapping target genes

    • Conduct motif analysis of binding sites

  • Double and triple knockdown/knockout experiments:

    • Generate combinatorial loss-of-function models

    • Test for synergistic or additive effects

    • Perform rescue experiments with individual proteins

  • Evolutionary approach:

    • Compare functions across multiple model organisms

    • Analyze conservation of binding sites and interaction partners

    • Reconstruct ancestral proteins to test functional evolution

What emerging technologies are likely to advance our understanding of cxxc4 function in the next five years?

Several cutting-edge approaches show promise for zinc finger protein research:

  • Single-cell multi-omics:

    • Single-cell RNA-seq to map expression in rare cell populations

    • Single-cell ATAC-seq to correlate with chromatin accessibility

    • Spatial transcriptomics to preserve tissue context

    • Integration of multiple data types at single-cell resolution

  • Advanced imaging techniques:

    • Live imaging of fluorescently tagged cxxc4 during development

    • Super-resolution microscopy of nuclear organization

    • Multiplexed protein imaging using DNA-PAINT or similar approaches

    • Correlative light and electron microscopy for ultrastructural context

  • Protein engineering approaches:

    • Designed zinc finger arrays with altered specificity

    • Split protein complementation for detecting interactions in vivo

    • Degron-based approaches for rapid protein degradation

    • Proximity-dependent labeling for identifying transient interactions

  • High-throughput functional genomics:

    • CRISPR screens in Xenopus tropicalis

    • Massively parallel reporter assays for enhancer analysis

    • Base editing for precise genetic modifications

    • Perturb-seq for linking genotype to transcriptional phenotype

  • Computational integration:

    • Machine learning approaches for predicting zinc finger binding

    • Systems biology modeling of developmental gene networks

    • Molecular dynamics simulations of zinc finger-nucleic acid interactions

    • Multi-scale modeling from molecular to tissue levels

How can contradictory findings about cxxc4 function be reconciled through improved experimental design?

Resolving contradictions requires systematic approaches:

  • Standardization of experimental conditions:

    • Define consistent developmental stages for experiments

    • Standardize protein expression and purification protocols

    • Use consistent nucleic acid binding assay conditions

    • Adopt uniform morpholino or CRISPR design guidelines

  • Direct replication studies:

    • Perform side-by-side comparisons of conflicting protocols

    • Exchange reagents between laboratories

    • Pre-register experimental designs and analysis plans

    • Conduct multi-lab collaborative studies

  • Resolution of technical limitations:

    • Identify sources of antibody cross-reactivity

    • Test for morpholino off-target effects

    • Control for genetic background differences

    • Account for maternal contribution in early development

  • Integration of multiple approaches:

    • Combine in vitro biochemical data with in vivo functional studies

    • Correlate structural information with functional outcomes

    • Link genomic binding data with transcriptional effects

    • Connect developmental phenotypes with molecular mechanisms

  • Context-dependent function analysis:

    • Test function across multiple developmental stages

    • Analyze tissue-specific effects

    • Investigate environmental influences on function

    • Examine effects of post-translational modifications

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.