Recombinant Pongo abelii Zinc finger protein 22 (ZNF22)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we 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 purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Our standard shipping includes blue ice packs. Dry ice shipping requires advance notification 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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
ZNF22Zinc finger protein 22
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-224
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Pongo abelii (Sumatran orangutan) (Pongo pygmaeus abelii)
Target Names
ZNF22
Target Protein Sequence
MRLAKPKAGI SRSSSQGKAY ENKRKTGRQR QKWGMTIRFD SSFSRLRRSL DDKPYKCTEC EKSFSQSSTL FQHQKIHTGK KSHKCADCGK SFFQSSNLIQ HRRIHTGEKP YKCDECGESF KQSSNLIQHQ RIHTGEKPYQ CDECGRCFSQ SSHLIQHQRT HTGEKPYQCS ECGKCFSQSS HLRQHMKVHK EEKPRKTRGK NIRVKTHLPS WKAGTGRKSV AGLR
Uniprot No.

Target Background

Function
Binds DNA via the consensus sequence 5'-CAATG-3'. It may be involved in transcriptional regulation and potentially plays a role in tooth development.
Database Links
Protein Families
Krueppel C2H2-type zinc-finger protein family
Subcellular Location
Nucleus.

Q&A

What is ZNF22 and why is it important for comparative genomics research?

ZNF22 (zinc finger protein 22) is a protein-coding gene found in Pongo abelii (Sumatran orangutan) with Entrez Gene ID 100173890 . The importance of studying ZNF22 in comparative genomics stems from the evolutionary conservation of zinc finger proteins across species. Zinc finger proteins function as transcription factors that regulate gene expression by binding to specific DNA sequences. Studying ZNF22 in non-human primates like Pongo abelii provides valuable insights into the evolution of gene regulatory networks and can help illuminate human gene function through comparative analysis. The protein exists in multiple isoforms, including the standard zinc finger protein 22 and zinc finger protein 22 isoform X1, as documented in the NCBI Reference Sequence Database . The comparative analysis of ZNF22 structure and function across primate species can reveal evolutionary adaptations in transcriptional regulation mechanisms.

What are the known isoforms of Pongo abelii ZNF22 and how do they differ functionally?

The Pongo abelii ZNF22 gene produces multiple protein isoforms according to the NCBI Reference Sequence Database. The documented variants include:

mRNA ReferenceProtein ReferenceIsoform Name
XM_009245329.1XP_009243604.1zinc finger protein 22 isoform X1
XM_009245329.2XP_009243604.1zinc finger protein 22 isoform X1
NM_001133405.1NP_001126877.1zinc finger protein 22

These isoforms likely result from alternative splicing of the ZNF22 gene . While the exact functional differences between these isoforms have not been fully characterized in the literature, they may exhibit varying DNA-binding specificities, protein-protein interactions, or subcellular localizations. Research methods to determine functional differences typically include:

  • DNA binding assays (EMSA, ChIP-seq) to identify isoform-specific target sequences

  • Reporter gene assays to assess transcriptional regulatory activity

  • Co-immunoprecipitation studies to identify differential protein interaction partners

  • Localization studies using fluorescent-tagged constructs to determine subcellular distribution

The conservation of specific isoforms across species may indicate functionally important variants that have been maintained throughout evolutionary history.

What are the optimal expression systems for producing recombinant Pongo abelii ZNF22?

The optimal expression system for recombinant Pongo abelii ZNF22 depends on research objectives, required protein yields, and downstream applications. Several expression systems can be considered:

Expression vector selection typically includes tags for purification and detection. Based on approaches used for similar zinc finger proteins, an N-terminal His-tag is commonly employed . Expression should be optimized by testing various conditions:

ParameterVariables to Test
Induction temperature16°C, 25°C, 30°C, 37°C
Induction duration3h, 6h, overnight, 24h
Inducer concentration0.1-1.0 mM IPTG (bacterial) or 1-10% FBS (mammalian)
Media supplementsZnCl₂ (50-100 μM) to ensure zinc finger domain folding

For mammalian expression, transfection protocols similar to those used for other zinc finger proteins can be applied, followed by cell harvesting, PBS washing, and lysis using freeze-thaw cycles in the presence of protease inhibitors .

What purification strategies yield the highest purity and structural integrity for recombinant ZNF22?

Purification of recombinant ZNF22 should maintain the structural integrity of the zinc finger domains, which is crucial for DNA-binding functionality. A multi-step purification strategy is recommended:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin for His-tagged ZNF22, with washing buffers containing 20-40 mM imidazole to reduce non-specific binding .

  • Intermediate purification: Ion exchange chromatography (typically cation exchange as zinc finger proteins are generally basic) to separate differently charged species.

  • Polishing step: Size exclusion chromatography to remove aggregates and achieve high purity.

Throughout the purification process, buffers should contain:

ComponentConcentrationPurpose
ZnCl₂50-100 μMMaintain zinc finger domain integrity
DTT or βME1-5 mMPrevent cysteine oxidation
NaCl150-300 mMMaintain solubility
Glycerol10-20%Enhance stability
Protease inhibitors1XPrevent degradation

Quality control should include SDS-PAGE analysis, Western blotting using anti-His tag antibodies, and DNA-binding activity assays to confirm functional integrity . For structural studies, circular dichroism (CD) spectroscopy can be used to verify proper folding of the zinc finger domains.

How does the DNA-binding mechanism of Pongo abelii ZNF22 compare to other zinc finger proteins?

While the specific DNA-binding mechanism of Pongo abelii ZNF22 has not been fully characterized, insights can be drawn from studies of other zinc finger proteins. Classical C2H2 zinc finger proteins typically bind DNA through specific interactions between amino acid residues in the α-helix of each finger and nucleotides in the major groove of DNA .

The DNA-binding specificity is determined by key residues at positions -1, 2, 3, and 6 relative to the α-helix of each zinc finger domain. Based on studies of multi-zinc finger proteins like ZNF217, which uses only two of its eight zinc fingers (fingers 6 and 7) for DNA contact , ZNF22 might similarly employ a subset of its zinc finger domains for specific DNA recognition.

The binding mechanism likely involves:

  • Initial scanning of DNA through electrostatic interactions

  • Specific recognition of target sequences by key residues in the zinc finger α-helices

  • Stabilization of the complex through additional contacts involving residues in linker regions

Unlike the typical three-finger arrays separated by canonical TGE(K/R)P linkers seen in proteins like Zif268, ZNF22 may utilize a non-canonical arrangement of zinc fingers for DNA recognition . Experimental approaches to characterize its binding mechanism include:

  • Electrophoretic mobility shift assays (EMSA) to determine DNA-binding specificity

  • DNase I footprinting to identify protected DNA regions

  • Systematic evolution of ligands by exponential enrichment (SELEX) to determine consensus binding sequences

  • X-ray crystallography or NMR spectroscopy to resolve the structural details of ZNF22-DNA complexes

What are the predicted consensus binding sequences for ZNF22 based on its zinc finger domain composition?

Predicting the consensus binding sequence for ZNF22 requires analysis of its zinc finger domains and comparison with known zinc finger-DNA interactions. While the exact binding preferences of Pongo abelii ZNF22 have not been experimentally determined, computational approaches can provide preliminary predictions.

Based on studies of other zinc finger proteins, each zinc finger typically recognizes 3-4 base pairs of DNA. The amino acid residues at positions -1, 2, 3, and 6 relative to the α-helix of each finger are particularly important for base-specific contacts . By analyzing these residues in ZNF22 and comparing them to zinc fingers with known specificities, approximate binding preferences can be predicted.

For example, studies of ZNF217 identified a preferred binding site of (T/A)(G/A)CAGAA(T/G/C), which differs from previously proposed sequences . Similarly, ZNF22 likely has its own unique binding preferences.

To experimentally determine the consensus binding sequence, several approaches can be employed:

  • SELEX (Systematic Evolution of Ligands by Exponential Enrichment): This method involves iterative selection of DNA sequences that bind with high affinity to ZNF22, followed by sequencing and motif analysis.

  • Protein-binding microarrays: These provide comprehensive analysis of binding to all possible DNA sequences of a given length.

  • ChIP-seq (Chromatin Immunoprecipitation followed by sequencing): This approach identifies genomic binding sites in cellular contexts.

  • In vitro binding assays with mutational analysis: Systematic mutation of potential binding sites can reveal critical nucleotides for recognition.

The results from these methods would generate a position weight matrix representing the binding preferences of ZNF22, which could then be used to predict potential target genes regulated by this transcription factor.

What are the critical parameters for successful cloning and expression of ZNF22 cDNA in standard expression vectors?

Successful cloning and expression of ZNF22 cDNA requires attention to several critical parameters:

  • Codon optimization: When expressing Pongo abelii ZNF22 in heterologous systems, codon optimization for the host organism can significantly improve expression levels. This is particularly important when expressing in E. coli, which has different codon usage preferences than primates.

  • Vector selection: The choice of expression vector should be based on:

    • Promoter strength (e.g., T7, CMV, CAG)

    • Fusion tags (His, GST, MBP, SUMO) that can enhance solubility and facilitate purification

    • Presence of appropriate cloning sites

    • Copy number and stability

  • Insert design:

    • Include Kozak sequence for efficient translation in eukaryotic systems

    • Consider removing the native stop codon if C-terminal fusion tags are desired

    • Include appropriate restriction sites flanking the ORF

    • Consider optimization of GC content in the 5' region

  • Expression conditions:

    • Supplementation with zinc ions (50-100 μM ZnCl₂) is crucial for proper folding of zinc finger domains

    • Lower temperatures (16-25°C) often improve soluble expression

    • Induction parameters should be optimized (time, inducer concentration)

The ZNF22 cDNA can be obtained from resources like the NCBI Reference Sequence Database, where multiple variants are documented (NM_001133405.1, XM_009245329.1, XM_009245329.2) . Commercial ORF clones are also available and can be subcloned into appropriate expression vectors using standard molecular cloning techniques.

How can researchers efficiently engineer zinc finger variants of ZNF22 for altered DNA-binding specificity?

Engineering zinc finger variants with altered DNA-binding specificity can be achieved through several approaches, drawing from established zinc finger engineering methodologies:

  • Bipartite selection strategy: This method involves parallel selection of two DNA-binding domains using phage display libraries. As described for other zinc finger proteins, the procedure involves:

    • PCR amplification of selected zinc fingers

    • Restriction digestion with DdeI

    • Random recombination by re-ligation

    • Recovery of desired recombinants by selective PCR

  • Rationally designed mutations: Based on the recognition code of zinc fingers, specific residues at positions -1, 2, 3, and 6 relative to the α-helix can be mutated to alter binding specificity. This approach requires:

    • Structural analysis or homology modeling of ZNF22

    • Identification of key residues that contact DNA

    • Site-directed mutagenesis of these residues

    • Functional validation through binding assays

  • Directed evolution: This strategy employs random mutagenesis followed by selection:

    • Error-prone PCR to generate libraries of ZNF22 variants

    • Phage or yeast display to select variants with desired binding properties

    • Iterative rounds of selection to refine specificity

  • Modular assembly: Given that zinc fingers often function as independent modules, novel zinc finger arrays can be constructed by combining fingers with known specificities:

    • Selection of individual fingers with desired triplet recognition

    • Design of appropriate linkers between fingers

    • Assembly of the composite array

    • Validation of DNA-binding specificity

The high-throughput nature of these approaches allows for parallel engineering of multiple variants, potentially yielding thousands of zinc finger transcription factors per year using automated systems . Validation of engineered variants should include binding affinity measurements and specificity profiling to ensure the desired DNA recognition properties have been achieved.

What are the potential applications of recombinant ZNF22 in transcriptional regulation studies?

Recombinant ZNF22 can serve as a valuable tool in transcriptional regulation studies, offering several research applications:

  • Identification of genomic targets: Recombinant ZNF22 can be used in chromatin immunoprecipitation followed by sequencing (ChIP-seq) experiments to identify its genomic binding sites. This approach reveals the genes potentially regulated by ZNF22 and enables construction of gene regulatory networks.

  • Transcriptional activation/repression studies: By fusing ZNF22 DNA-binding domains with various effector domains (activators like VP64 or repressors like KRAB), researchers can study the impact of ZNF22-mediated regulation on target gene expression. Similar approaches have been used with other zinc finger proteins to modulate gene expression in experimental settings .

  • Protein-protein interaction analysis: Recombinant ZNF22 can be used in pull-down assays, co-immunoprecipitation, or yeast two-hybrid screens to identify cofactors and other interaction partners involved in transcriptional complexes.

  • Comparative studies across species: Given its presence in Pongo abelii, comparative studies with human and other primate ZNF22 orthologs can reveal evolutionary conservation and divergence in transcriptional regulation mechanisms.

  • Structural studies: Purified recombinant ZNF22 can be used for crystallography or NMR studies to determine its three-dimensional structure, especially in complex with DNA, providing insights into its binding mechanism.

  • Development of transcription factor-based tools: Similar to other engineered zinc finger proteins, ZNF22 domains could potentially be used to develop synthetic transcription factors for targeted gene regulation in research or therapeutic applications .

How can ZNF22 be utilized in comparative genomics studies between human and non-human primates?

ZNF22 presents an excellent candidate for comparative genomics studies between humans and non-human primates, offering insights into transcriptional regulation evolution:

  • Sequence comparison analysis: Alignment of ZNF22 sequences from humans, Pongo abelii, and other primates can reveal:

    • Conserved domains indicating functional importance

    • Species-specific variations that might confer adaptive advantages

    • Selection pressures acting on different regions of the protein

  • Binding site conservation and divergence: Comparing the genomic binding sites of ZNF22 across primate species can reveal:

    • Core conserved target genes that likely serve fundamental functions

    • Species-specific targets that might contribute to phenotypic differences

    • Evolution of regulatory networks

  • Functional conservation testing: Experimental approaches include:

    • Cross-species complementation assays to determine if ZNF22 from one species can functionally replace that of another

    • Reporter gene assays with promoters from different species to assess regulatory potential

    • Binding affinity measurements to compare DNA interaction strengths

  • Gene expression profiling: RNA-seq studies following ZNF22 overexpression or knockdown in cells from different primate species can reveal:

    • Conserved and divergent downstream effects

    • Species-specific regulatory networks

    • Potential roles in primate evolution

  • Structural comparisons: Recombinant proteins from different species can be used for structural studies to identify subtle differences that might affect function.

The methodological approach would typically involve:

  • Cloning and expressing ZNF22 from multiple primate species

  • Performing parallel functional assays under identical conditions

  • Computational analysis of binding sites and target genes

  • Integration of functional and genomic data to build evolutionary models of transcriptional regulation

What are common challenges in expressing and purifying functional ZNF22, and how can they be addressed?

Researchers frequently encounter several challenges when working with zinc finger proteins like ZNF22:

  • Poor solubility and aggregation:

    • Solution: Use solubility-enhancing fusion partners like MBP, SUMO, or Thioredoxin

    • Lower expression temperature to 16-18°C

    • Add stabilizing agents (glycerol, arginine, zinc ions) to buffers

    • Consider refolding from inclusion bodies if necessary

  • Loss of zinc during purification:

    • Solution: Include 50-100 μM ZnCl₂ in all buffers

    • Avoid strong chelating agents (EDTA) in buffers

    • Monitor zinc content using colorimetric assays (PAR assay)

  • Proteolytic degradation:

    • Solution: Add protease inhibitor cocktails to lysis buffers

    • Perform purification at 4°C

    • Minimize processing time

    • Consider using protease-deficient expression hosts

  • Poor DNA-binding activity:

    • Solution: Verify correct folding using circular dichroism spectroscopy

    • Ensure zinc incorporation through atomic absorption spectroscopy

    • Optimize buffer conditions (salt concentration, pH) for binding assays

    • Use freshly prepared protein for activity assays

  • Low expression yields:

    • Solution: Optimize codon usage for expression host

    • Test different promoters and expression strains

    • Consider using cell-free expression systems for toxic proteins

    • Scale up culture volume rather than extending expression time

A systematic troubleshooting approach should involve:

IssueDiagnostic MethodPotential Solutions
Insoluble expressionSDS-PAGE of soluble vs. insoluble fractionsChange fusion tags, lower temperature, add solubilizing agents
Improper foldingCircular dichroism spectroscopyAdd zinc, optimize reducing conditions, try refolding
AggregationSize exclusion chromatography, DLSAdjust buffer composition, add stabilizing agents
Low puritySDS-PAGE, Western blotAdd intermediate purification steps, optimize wash conditions
Poor activityEMSA, fluorescence polarizationEnsure proper folding, optimize binding conditions

What advanced analytical techniques are most informative for studying ZNF22-DNA interactions?

Several advanced analytical techniques provide valuable insights into ZNF22-DNA interactions:

  • Surface Plasmon Resonance (SPR):

    • Provides real-time kinetic data (kon, koff) and equilibrium binding constants (KD)

    • Allows comparison of binding affinities to different DNA sequences

    • Can detect conformational changes upon binding

    • Methodology: Immobilize biotinylated DNA on streptavidin sensor chips, flow ZNF22 protein at various concentrations, analyze binding/dissociation curves

  • Isothermal Titration Calorimetry (ITC):

    • Measures thermodynamic parameters (ΔH, ΔS, ΔG)

    • Determines binding stoichiometry

    • No labeling or immobilization required

    • Methodology: Titrate ZNF22 into DNA solution while measuring heat changes

  • Bio-Layer Interferometry (BLI):

    • Similar to SPR but uses optical interference patterns

    • Good for high-throughput screening of binding interactions

    • Requires less sample than SPR

    • Methodology: Immobilize DNA on biosensors, dip into ZNF22 solutions, measure binding in real-time

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):

    • Maps protein regions involved in DNA binding

    • Identifies conformational changes upon binding

    • Provides structural information without crystallization

    • Methodology: Compare deuterium uptake of free vs. DNA-bound ZNF22

  • Single-molecule FRET (smFRET):

    • Observes binding events at single-molecule resolution

    • Detects conformational dynamics and heterogeneity

    • Provides insights into binding mechanisms

    • Methodology: Label ZNF22 and DNA with FRET pairs, observe energy transfer changes upon binding

  • X-ray Crystallography and Cryo-EM:

    • Provides atomic-resolution structures of ZNF22-DNA complexes

    • Reveals specific protein-DNA contacts

    • Informs structure-based engineering

    • Methodology: Crystallize or vitrify purified ZNF22-DNA complexes, collect diffraction or image data

  • ChIP-sequencing (ChIP-seq):

    • Identifies genomic binding sites in cellular contexts

    • Reveals physiologically relevant targets

    • Can be combined with gene expression data to assess regulatory impact

    • Methodology: Crosslink protein-DNA in cells, immunoprecipitate ZNF22-bound DNA, sequence and map to genome

Each of these techniques provides complementary information, and combining multiple approaches yields the most comprehensive understanding of ZNF22-DNA interactions. For example, SPR and ITC provide quantitative binding parameters, while structural techniques reveal the molecular basis for recognition specificity.

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.