Recombinant Xenopus laevis UPF0406 protein C16orf57 homolog

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 remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes 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 sediment 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 serve as a guideline.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer components, temperature, and the protein's inherent 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. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
usb1; U6 snRNA phosphodiesterase; EC 3.1.4.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-250
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
usb1
Target Protein Sequence
MALVSYSSSE EDEGETSEPP GRRLPPLPPP TTVLRMFQDM EGDEDLDENT KHEGRIRSFK HERGNWATYV YIPFQPQEEF LDLLDELVSV AAENGVLLTK MSEFHISQSQ TVVLRHHWIN PFVESLKDKL HCMYRFLCIA ERIKVYTNQE KTRTFLGLEV SVGMEHLLEV VSEVDRSLQE FNLQTFYQEP SFHVSLAWCV GDKYEKLKGS CLLELQKVID RFEDSDTLTR FNAEEIRCKA GNKTFCIPLL
Uniprot No.

Target Background

Function
This phosphodiesterase is responsible for U6 snRNA 3' end processing. It functions as an exoribonuclease (RNase), trimming the poly(U) tract from the pre-U6 snRNA molecule's terminal nucleotides, thereby generating mature U6 snRNA.
Database Links

KEGG: xla:379166

UniGene: Xl.9881

Protein Families
2H phosphoesterase superfamily, USB1 family
Subcellular Location
Nucleus.

Q&A

Basic Research Questions

  • What is the UPF0406 protein C16orf57 homolog in Xenopus laevis?

    The UPF0406 protein C16orf57 homolog in Xenopus laevis represents the amphibian ortholog of the human C16orf57 gene product (also known as USB1 or hUSB1). C16orf57 encodes a human protein that functions as a phosphodiesterase essential for the generation of the cyclic phosphate at the 3′ end of the U6 snRNA, a critical component of the RNA splicing machinery. The Xenopus homolog likely maintains similar enzymatic functions in amphibian RNA processing pathways, particularly in U6 snRNA biogenesis. The protein belongs to a family of uncharacterized proteins with conserved phosphodiesterase domains. Mutations in human C16orf57 are associated with several rare autosomal recessive diseases, including poikiloderma with neutropenia (PN), as well as some cases of Rothmund-Thomson syndrome (RTS) and dyskeratosis congenita (DC) .

  • What functional domains characterize the C16orf57 homolog?

    The C16orf57 homolog contains a conserved phosphodiesterase domain that is essential for its enzymatic activity in RNA processing. Based on studies of the human USB1 and its yeast ortholog, this domain catalyzes the removal of terminal nucleotides from U6 snRNA transcripts, leading to mature snRNA formation. The protein likely possesses RNA-binding motifs that facilitate substrate recognition. Structurally, the phosphodiesterase domain is particularly well-conserved across species, from yeast to humans, indicating its fundamental importance in cellular processes. In yeast, the ortholog USB1 (YLR132C) is essential for U6 snRNA biogenesis and cell viability, with its depletion leading to destabilization of U6 snRNA, splicing defects, and cell growth abnormalities .

  • How does C16orf57 homolog expression compare across developmental stages in Xenopus?

    While specific expression data for the C16orf57 homolog in Xenopus laevis is not comprehensively documented across all developmental stages, the protein is likely expressed in tissues with high rates of RNA processing and cell division. Based on deep proteomics studies of Xenopus laevis eggs that have identified thousands of proteins, RNA processing factors like C16orf57 homolog would be expected to be present during early development. The expression pattern may change throughout embryogenesis as different tissues form and mature. In particular, tissues requiring high levels of mRNA splicing activity would likely show enriched expression. Understanding these developmental expression patterns is crucial, given the importance of proper RNA processing during embryonic development and the potential implications for disease modeling studies .

  • What is the evolutionary significance of C16orf57 homologs?

    C16orf57 homologs show remarkable evolutionary conservation across diverse species, indicating their ancient and essential role in cellular function. This conservation extends from yeast (USB1) to amphibians (Xenopus) and mammals (human USB1/C16orf57), suggesting that the protein's role in U6 snRNA processing represents a fundamental cellular process that has been maintained throughout eukaryotic evolution. The high degree of conservation in the phosphodiesterase domain particularly underscores its critical enzymatic function. Comparative genomic analyses provide a unique opportunity to study both functional conservation and species-specific adaptations of this protein family. The presence of C16orf57 homologs across diverse species allows for effective cross-species functional studies and validates the use of model organisms like Xenopus for investigating the fundamental biology and disease relevance of this protein .

Expression and Purification Methods

  • What expression systems are most effective for recombinant Xenopus C16orf57 homolog production?

    Based on successful approaches with similar Xenopus proteins, several expression systems can be employed for recombinant C16orf57 homolog production:

    Expression SystemAdvantagesLimitationsOptimal Use Cases
    E. coliHigh yield, cost-effective, established protocolsLimited post-translational modificationsSmaller proteins without complex folding requirements
    Insect cellsBetter folding, some post-translational modificationsMore time-consuming, moderate costProteins requiring eukaryotic processing
    Mammalian cellsAuthentic post-translational modificationsLower yield, highest costProteins requiring specific mammalian modifications
    Cell-free systemsRapid production, handles toxic proteinsLower yield, more variableQuick expression screening

    E. coli is frequently the system of choice, as demonstrated with the successful production of other Xenopus recombinant proteins like UPF0694 transmembrane protein C14orf109 homolog A. For the C16orf57 homolog, an N-terminal His-tag fusion construct expressed in E. coli would likely provide sufficient yield and purity for most research applications .

  • What is the optimal purification protocol for recombinant Xenopus C16orf57 homolog?

    The recommended purification protocol for recombinant Xenopus C16orf57 homolog should include:

    a) Initial Capture:

    • Affinity chromatography using Ni-NTA resin for His-tagged protein

    • Washing with increasing imidazole concentrations to remove non-specific binding

    b) Intermediate Purification:

    • Ion exchange chromatography to separate based on charge differences

    • Optimal buffer conditions should be determined empirically (typical pH range 7.0-8.0)

    c) Polishing Step:

    • Size exclusion chromatography to remove aggregates and achieve >90% purity

    • Typical buffer: Tris/PBS-based with 6% Trehalose, pH 8.0

    d) Quality Control:

    • SDS-PAGE analysis to confirm >90% purity

    • Western blot to verify protein identity

    • Functional assay to confirm phosphodiesterase activity

    The purified protein should be lyophilized for storage or maintained in solution with appropriate stabilizers such as glycerol (final concentration 50%). This multi-step purification approach has proven effective for other recombinant Xenopus proteins and should yield high-purity C16orf57 homolog suitable for biochemical and structural studies .

  • What are the critical storage and handling requirements for maintaining C16orf57 homolog stability?

    For optimal stability and activity of the recombinant Xenopus C16orf57 homolog, follow these storage and handling guidelines:

    a) Long-term Storage:

    • Store lyophilized protein at -80°C

    • For solution storage, use Tris/PBS-based buffer with 6% Trehalose, pH 8.0

    • Add glycerol to 50% final concentration for frozen storage

    b) Working Solutions:

    • Reconstitute in deionized sterile water to 0.1-1.0 mg/mL

    • Store working aliquots at 4°C for up to one week

    • Avoid repeated freeze-thaw cycles as they significantly compromise protein integrity

    c) Handling Precautions:

    • Briefly centrifuge vials before opening to collect material at the bottom

    • Use low-binding tubes and pipette tips to minimize protein loss

    • When reconstituting, gently mix by inversion rather than vortexing

    d) Activity Preservation:

    • For enzymatic studies, add protease inhibitors to prevent degradation

    • Consider adding reducing agents if the protein contains critical cysteine residues

    • Periodically verify protein integrity by SDS-PAGE and activity assays

    These guidelines are based on successful approaches used with other recombinant Xenopus proteins, such as UPF0694 transmembrane protein and UBC4, and should maintain the structural and functional integrity of the C16orf57 homolog .

Functional Analysis Methods

  • What assays can effectively measure the phosphodiesterase activity of C16orf57 homolog?

    Several complementary assays can be employed to measure the phosphodiesterase activity of Xenopus C16orf57 homolog:

    a) Radiolabeled Substrate Assays:

    • Use 32P-labeled synthetic RNA substrates mimicking U6 snRNA

    • Monitor release of labeled phosphate by thin-layer chromatography

    • Quantify signal using phosphorimaging for precise activity measurements

    b) Fluorescence-Based Assays:

    • Utilize fluorescently labeled RNA substrates with quencher pairs

    • Measure fluorescence increase upon substrate cleavage

    • Enables high-throughput screening and real-time kinetic measurements

    c) HPLC/Mass Spectrometry Analysis:

    • Analyze reaction products to confirm specific cleavage patterns

    • Identify precise modification of the 3' end of U6 snRNA substrates

    • Determine whether cyclic phosphate structures are formed

    d) Gel-Based Assays:

    • Use denaturing polyacrylamide gels to separate substrate and products

    • Visualize with SYBR Gold or other RNA stains

    • Provide qualitative assessment of enzymatic activity

    These methods should be optimized with appropriate controls, including catalytically inactive mutants, to ensure specificity. Combining multiple assay approaches provides the most comprehensive characterization of the enzymatic properties of the C16orf57 homolog and its substrate specificity .

  • How can I design effective experiments to investigate C16orf57 homolog's role in U6 snRNA processing?

    To investigate the C16orf57 homolog's role in U6 snRNA processing, a multi-faceted experimental approach is recommended:

    a) In Vitro RNA Processing Assays:

    • Incubate purified recombinant C16orf57 homolog with in vitro transcribed U6 snRNA

    • Analyze resulting RNA products by sequencing and 3' end mapping

    • Compare wild-type protein activity with catalytically inactive mutants

    b) Cellular Depletion Studies:

    • Implement CRISPR/Cas9 or morpholino-based knockdown in Xenopus cells/embryos

    • Extract total RNA and specifically analyze U6 snRNA integrity and modification

    • Perform RNA-seq to assess global splicing changes

    c) Structure-Function Analysis:

    • Generate mutant versions of the protein based on disease-associated mutations

    • Compare their enzymatic activity with wild-type protein

    • Correlate functional deficits with structural predictions

    d) U6 snRNA Analysis:

    • Use 3' RACE or specialized sequencing approaches to characterize U6 snRNA 3' ends

    • Compare U6 snRNA stability between control and C16orf57-depleted samples

    • Assess formation of the characteristic cyclic phosphate at the 3' end

    These approaches would help determine whether the Xenopus C16orf57 homolog, like its human counterpart, functions as the long-sought U6 snRNA phosphodiesterase and elucidate its precise role in RNA processing pathways .

  • What proteomics approaches are most informative for studying C16orf57 homolog interactions?

    For comprehensive investigation of C16orf57 homolog protein interactions, employ these proteomics approaches:

    a) Immunoprecipitation-Mass Spectrometry (IP-MS):

    • Generate antibodies against the Xenopus C16orf57 homolog or use epitope-tagged versions

    • Perform immunoprecipitation from Xenopus egg or embryo extracts

    • Identify co-precipitated proteins by mass spectrometry

    • Compare results with control IPs to identify specific interactors

    b) Proximity Labeling:

    • Fuse the protein to BioID (biotin ligase) or APEX (peroxidase)

    • Express the fusion protein in Xenopus cells/embryos

    • Identify biotinylated proteins via streptavidin pulldown and mass spectrometry

    • Provides spatial information about protein interaction networks

    c) Cross-linking Mass Spectrometry (XL-MS):

    • Use chemical cross-linkers to stabilize transient interactions

    • Digest complexes and identify cross-linked peptides by mass spectrometry

    • Provides information about interaction interfaces

    d) Integrative Analysis:

    • Combine proteomic data with transcriptomics

    • Perform network analysis to identify functional protein complexes

    • Validate key interactions with targeted biochemical assays

    These approaches would leverage the extensive proteomics work already done in Xenopus (identifying >11,000 proteins in Xenopus eggs) and could reveal novel interaction partners of the C16orf57 homolog, potentially including other RNA processing factors or components of the spliceosome .

Advanced Research Applications

  • How can I develop a Xenopus model for diseases associated with C16orf57 mutations?

    Developing a Xenopus model for C16orf57-associated diseases requires a strategic approach:

    a) Genetic Engineering:

    • Generate specific mutations corresponding to human disease variants

    • Use CRISPR/Cas9 with homology-directed repair for precise genome editing

    • Create both complete knockout and specific disease mutations

    b) Phenotypic Characterization:

    • Analyze embryonic development with particular focus on:

      • Skin development (relevant to poikiloderma)

      • Hematopoiesis (relevant to neutropenia)

      • Growth and skeletal development

    • Document phenotypes using imaging, histology, and molecular markers

    c) Molecular Analysis:

    • Examine U6 snRNA processing via specialized RNA analysis

    • Perform transcriptome-wide analysis to identify misregulated genes

    • Assess splicing efficiency using RNA-seq approaches

    • Evaluate telomere maintenance despite no reported telomere length changes in RTS patients with C16orf57 mutations

    d) Rescue Experiments:

    • Test whether human wild-type C16orf57 can rescue phenotypes

    • Compare rescue efficiency with disease-associated mutants

    • Identify potential therapeutic targets through these rescue experiments

    Xenopus offers significant advantages for this disease modeling, including external development for easy observation, high fecundity for statistical power, and well-characterized developmental stages. This approach would provide valuable insights into the pathogenesis of rare diseases like poikiloderma with neutropenia, Rothmund-Thomson syndrome, and dyskeratosis congenita that have been associated with C16orf57 mutations .

  • What experimental approaches can resolve contradictory findings between C16orf57 mutations and telomere maintenance?

    To address the seemingly contradictory relationship between C16orf57 mutations and telomere maintenance, these experimental approaches are recommended:

    a) Comparative Telomere Analysis:

    • Measure telomere length in wild-type vs. C16orf57-mutant Xenopus using:

      • Telomere restriction fragment analysis

      • qPCR-based telomere length assays

      • Fluorescence in situ hybridization (FISH) for telomere visualization

    • Analyze telomere dynamics across multiple developmental stages

    • Compare results with published observations in human patients

    b) Mechanistic Investigations:

    • Examine expression of telomere maintenance genes in C16orf57-mutant backgrounds

    • Analyze alternative splicing of telomere-related transcripts

    • Investigate non-coding RNAs involved in telomere regulation

    c) DNA Damage Response Assessment:

    • Measure DNA damage markers (γH2AX, 53BP1) in C16orf57-deficient cells

    • Assess sensitivity to genotoxic agents

    • Evaluate repair kinetics following induced DNA damage

    d) Integrative Approach:

    • Perform multi-omics analysis (transcriptomics, proteomics, metabolomics)

    • Use network analysis to identify perturbed pathways

    • Develop computational models predicting indirect effects on telomere biology

    This comprehensive approach could resolve the paradox wherein C16orf57 mutations cause diseases typically associated with telomere dysfunction (dyskeratosis congenita) yet patients show no measurable changes in telomere length. The results may reveal novel connections between RNA processing pathways and genome stability mechanisms .

  • What structural analysis methods are most suitable for C16orf57 homolog characterization?

    For comprehensive structural characterization of the Xenopus C16orf57 homolog, employ these complementary approaches:

    a) X-ray Crystallography:

    • Optimize protein purification for high concentration and homogeneity

    • Screen multiple crystallization conditions

    • Collect high-resolution diffraction data

    • Solve structure using molecular replacement with human homolog (if available)

    b) Nuclear Magnetic Resonance (NMR):

    • Suitable for analyzing dynamic regions of the protein

    • Requires isotopically labeled protein (15N, 13C)

    • Provides information about protein dynamics in solution

    • Especially valuable for RNA-protein interaction studies

    c) Cryo-Electron Microscopy:

    • Particularly useful for larger complexes with RNA or protein partners

    • Can capture different functional states of the protein

    • Requires less protein than crystallography

    d) Computational Approaches:

    • Homology modeling based on human or yeast orthologs

    • Molecular dynamics simulations to study conformational changes

    • Normal Mode Analysis to capture dynamical properties essential for function

    • As demonstrated in recent protein design research, dynamics-informed modeling can capture functional aspects missed by static structures

    e) Small-Angle X-ray Scattering (SAXS):

    • Provides low-resolution envelope of protein in solution

    • Useful for confirming oligomeric state and general shape

    • Complements higher-resolution structural methods

    These approaches would provide insights into the catalytic mechanism of the C16orf57 homolog and help explain how disease-associated mutations disrupt protein function .

  • How can sequence-space exploration methods improve C16orf57 homolog functional studies?

    Sequence-space exploration methods offer powerful approaches to enhance C16orf57 homolog functional studies:

    a) Deep Mutational Scanning:

    • Generate comprehensive libraries of protein variants

    • Select for functional variants using appropriate assays

    • Sequence recovered variants to create detailed fitness landscapes

    • Identify residues critical for phosphodiesterase activity

    b) Epistasis Mapping:

    • Analyze how combinations of mutations affect protein function

    • Identify compensatory mutations that rescue deleterious effects

    • Map functional interaction networks within the protein structure

    c) Evolutionary Sequence Analysis:

    • Compare C16orf57 homologs across diverse species

    • Identify co-evolving residues suggesting functional interactions

    • Use this information to predict critical structural elements

    d) Computational Modeling:

    • Simulate sequence-space exploration in silico

    • Optimize experimental design for more efficient mapping

    • As demonstrated in recent research, this approach can guide experimental protocols by predicting optimal selection strength parameters

    e) Directed Evolution:

    • Apply selection pressure in cycles to evolve improved protein variants

    • Alternate between strong and weak selection as suggested by computational models

    • Develop proteins with enhanced stability or catalytic efficiency

    These methods would provide deeper insights into structure-function relationships of the C16orf57 homolog and potentially identify regions of therapeutic interest for disease-associated mutations .

Disease Implications and Therapeutic Applications

  • What connections exist between C16orf57 homolog function and disease phenotypes?

    The connections between C16orf57 homolog function and disease phenotypes span multiple cellular processes:

    a) RNA Processing Defects:

    • Primary function as U6 snRNA phosphodiesterase suggests splicing defects

    • Aberrant splicing may affect multiple downstream pathways

    • Tissue-specific effects could explain variable disease presentations

    b) Clinical Manifestations:

    • Poikiloderma (skin abnormalities) suggests epithelial defects

    • Neutropenia indicates hematopoietic dysfunction

    • Growth abnormalities point to developmental regulation roles

    c) Molecular Pathways:

    • Despite association with diseases typically linked to DNA repair/telomere maintenance:

      • No changes in telomere length were observed in patients with C16orf57 mutations

      • Suggests alternative pathological mechanisms

    • Possibility of indirect effects on genome stability through splicing defects

    d) Genotype-Phenotype Correlations:

    • Different mutations cause overlapping syndromes (PN, RTS, DC)

    • Location of mutations may determine severity and presentation

    • Understanding these correlations requires functional characterization of different variants

    This complex relationship between molecular function and disease phenotypes makes the C16orf57 homolog particularly interesting for studying how RNA processing defects can manifest as specific tissue abnormalities .

  • How might C16orf57 homolog research inform potential therapeutic approaches?

    Research on the C16orf57 homolog could inform therapeutic strategies through several avenues:

    a) Direct Enzyme Replacement:

    • Develop delivery methods for recombinant protein to affected tissues

    • Design stabilized enzyme variants with improved half-life

    • Target tissue-specific delivery to address prominent disease manifestations

    b) RNA-Based Therapies:

    • Antisense oligonucleotides to modulate splicing of key target genes

    • RNA editing approaches to correct processing defects

    • Small RNA delivery to compensate for misprocessed transcripts

    c) Small Molecule Screening:

    • Identify compounds that:

      • Stabilize mutant C16orf57 proteins

      • Enhance residual enzymatic activity

      • Activate compensatory pathways

    • Xenopus models provide excellent platforms for in vivo compound screening

    d) Gene Therapy Approaches:

    • Develop viral vectors for C16orf57 gene delivery

    • Explore CRISPR-based gene correction for specific mutations

    • Evaluate cell-based therapies using corrected patient cells

    e) Pathway-Based Interventions:

    • Target downstream pathways affected by C16orf57 dysfunction

    • Develop combinatorial approaches addressing multiple affected pathways

    • Personalize interventions based on specific mutation profiles

    The Xenopus model system is particularly well-suited for evaluating these therapeutic approaches due to its high-throughput capacity, transparent embryos allowing easy phenotypic assessment, and evolutionary conservation of key pathways affected in human disease .

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.