Recombinant Xenopus laevis Protein asunder homolog (asun), partial

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 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 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 may serve as a guideline.
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 forms maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. If a specific tag type is required, please inform us for preferential development.
Synonyms
ints13; asun; mat89bb; net48; Integrator complex subunit 13; Cell cycle regulator Mat89Bb homolog; Germ cell tumor 1; Protein asunder homolog; Sarcoma antigen NY-SAR-95
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
ints13
Uniprot No.

Target Background

Function
This protein is a crucial regulator of the mitotic cell cycle and development. During prophase, it anchors dynein to the nuclear envelope, essential for proper centrosome-nucleus coupling. In the G2/M phase, it may be required for accurate spindle formation and cytokinesis. It is a likely component of the Integrator (INT) complex, involved in small nuclear RNA (snRNA) U1 and U2 transcription and 3'-box-dependent processing. This protein plays a significant role in gastrulation and early embryogenesis.
Database Links

KEGG: xla:444621

UniGene: Xl.70202

Protein Families
Asunder family
Subcellular Location
Nucleus. Cytoplasm.

Q&A

What is the Xenopus laevis ASUN protein and why is it significant for developmental biology research?

The asunder homolog (ASUN) in Xenopus laevis is a protein involved in nuclear migration processes and dynein recruitment to the nuclear envelope. Its significance in developmental biology stems from the established position of Xenopus as a phylogenetically intermediate vertebrate between aquatic species and land tetrapods, making it valuable for evolutionary and comparative studies. Xenopus has become an essential model organism for developmental biology research since it was discovered that it could easily be induced to breed in laboratory settings through human gonadotrophin injection . The ASUN protein specifically has become important for studying nuclear dynamics during cell division and differentiation processes.

How does the Xenopus model system contribute to our understanding of ASUN protein function?

Xenopus laevis provides several advantages for studying ASUN protein function. As an amphibian model with a remarkably conserved developmental biology, it allows researchers to observe protein function across evolutionary time. The model enables rapid generation of mutants through established genetic tools, particularly CRISPR/Cas9 mutagenesis techniques that are well-established and cost-effective in Xenopus systems . Additionally, the ability to generate unilateral mutants in Xenopus (where one half of the animal carries a homozygous mutation while the other half serves as an internal control) provides a unique advantage for studying the effects of ASUN protein modifications under identical developmental conditions .

What are the key structural and functional characteristics of recombinant Xenopus laevis ASUN protein?

Recombinant Xenopus laevis ASUN is characterized by its role in microtubule organization and nuclear positioning. While the search results don't specifically detail the structure of ASUN, related research on Xenopus proteins demonstrates that careful attention to protein folding and post-translational modifications is essential for functional studies. The ASUN protein likely interacts with RNA molecules, as many nuclear proteins in Xenopus have been shown to have RNA-binding properties, similar to how the ESCRT-II complex interacts with hundreds of mRNAs in Xenopus eggs . Structurally, the partial recombinant form would present specific domains of interest while potentially lacking others, making it suitable for targeted functional studies.

What are the recommended protocols for expressing and purifying recombinant Xenopus laevis ASUN protein?

For optimal expression and purification of recombinant Xenopus laevis ASUN protein:

  • Expression System Selection: Based on Xenopus protein research practices, bacterial expression systems (E. coli BL21) are suitable for partial ASUN protein production. For full-length or post-translationally modified variants, consider insect cell (Sf9) or mammalian expression systems.

  • Purification Protocol:

    • Employ affinity chromatography with histidine or GST tags

    • Follow with size exclusion chromatography to achieve >95% purity

    • Verify protein integrity using SDS-PAGE and western blotting

    • Confirm functionality through activity assays specific to nuclear migration processes

  • Buffer Optimization: Maintain protein stability in buffers containing 20-50mM Tris-HCl (pH 7.5-8.0), 100-300mM NaCl, 1-5mM DTT, and 10% glycerol to minimize aggregation.

The University of Rochester's Xenopus laevis Research Resource maintains protocols for working with numerous Xenopus proteins and could provide specialized techniques for ASUN protein work .

How can CRISPR/Cas9 techniques be optimized for studying ASUN protein function in Xenopus laevis?

CRISPR/Cas9 mutagenesis in Xenopus offers powerful approaches for studying ASUN function:

  • sgRNA Design Strategy:

    • Design sgRNAs targeting the 5' end of the ASUN coding sequence

    • Test multiple sgRNAs empirically for editing efficiency

    • Choose high-specificity guides with minimal predicted off-target effects

  • Unilateral Mutation Approach:

    • Inject CRISPR/Cas9 components into one cell at the 2-cell stage embryo

    • This creates half-mutant animals where one side serves as an internal control

    • This technique is unique to Xenopus and particularly valuable for studying developmental effects

  • Control Strategies:

    • Use non-targeting sgRNAs as negative controls

    • Target potential off-target sites identified by predictive algorithms to ensure phenotype specificity

    • Consider targeting pigmentation genes as visible editing controls

This methodology allows for the generation of thousands of ASUN mutant embryos per day, enabling parallel analysis across multiple experimental conditions .

What techniques are most effective for detecting protein-protein interactions involving ASUN in Xenopus systems?

For studying protein-protein interactions involving ASUN in Xenopus:

  • Co-immunoprecipitation (Co-IP):

    • Utilize Xenopus egg or embryo extracts, which provide a native cellular environment

    • Develop specific antibodies against Xenopus ASUN or use epitope tags

    • Verify interactions through reciprocal Co-IPs and controls for non-specific binding

  • Proximity Labeling Techniques:

    • BioID or TurboID fusions with ASUN to identify proximal proteins in vivo

    • APEX2-based labeling for temporally controlled interaction mapping

    • These approaches are particularly valuable in the context of nuclear envelope dynamics

  • Fluorescence Techniques:

    • FRET (Förster Resonance Energy Transfer) for direct interaction studies

    • BiFC (Bimolecular Fluorescence Complementation) for visualizing interactions in vivo

    • Perform in early Xenopus embryos where optical clarity facilitates imaging

  • Crosslinking Approaches:

    • Chemical crosslinking followed by mass spectrometry (XL-MS)

    • UV crosslinking for detecting RNA-protein interactions if ASUN binds RNA, similar to techniques used for studying ESCRT-II complex

These methods can be complemented by the resources available at specialized facilities like the University of Rochester's comprehensive Xenopus research resource .

What are common challenges in working with recombinant Xenopus ASUN protein and their solutions?

ChallengePotential CausesSolutions
Low expression yieldsCodon bias, toxicity to host cellsOptimize codons for expression system, use inducible promoters, express as fusion with solubility tags
Protein insolubilityHydrophobic domains, improper foldingExpress at lower temperatures (16-18°C), include solubilizing agents, consider expressing domains separately
Loss of activityImproper folding, missing cofactorsInclude Xenopus egg extract during purification steps, add known binding partners
Aggregation during storageConcentration effects, buffer incompatibilityStore at lower concentrations, optimize buffer conditions with stabilizers like glycerol
Variable experimental resultsBatch-to-batch variationEstablish robust quality control metrics, include activity assays before experiments

When troubleshooting experiments with recombinant ASUN protein, it's critical to consider the developmental context in which the protein naturally functions. The University of Rochester's Xenopus resource center provides technical assistance and training that could help address specific challenges related to Xenopus proteins .

How should researchers interpret conflicting data when studying ASUN function in different developmental contexts?

When encountering conflicting results regarding ASUN function across developmental stages:

  • Developmental Context Assessment:

    • Xenopus undergoes dramatic developmental changes, particularly during metamorphosis

    • ASUN function may vary between early embryogenesis, tadpole stages, and post-metamorphic contexts

    • Document precise developmental stages using standardized Nieuwkoop and Faber staging criteria

  • Methodological Reconciliation:

    • Compare experimental approaches (in vivo vs. in vitro, overexpression vs. knockdown)

    • Evaluate dose-dependent effects, as protein concentration affects function

    • Consider maternal protein persistence which may mask early phenotypes

  • Strain-Specific Variations:

    • Different Xenopus laevis strains show genetic variations

    • The University of Rochester maintains several genetically-defined inbred strains and clones

    • Replicate key experiments across multiple genetic backgrounds

  • Integration Framework:

    • Develop a comprehensive model incorporating developmental timing

    • Consider paralog compensation (especially relevant in the pseudotetraploid X. laevis)

    • Perform parallel experiments in X. tropicalis (diploid) to distinguish redundancy effects

Remember that Xenopus phenotypes often closely recapitulate human conditions, sometimes more accurately than rodent models , which may explain differences when comparing across model systems.

What statistical approaches are most appropriate for analyzing ASUN protein expression and activity data in Xenopus studies?

When analyzing ASUN protein data from Xenopus experiments:

  • For Expression Studies:

    • Normalize expression data to established housekeeping genes suitable for the developmental stage being studied

    • Apply ANOVA with post-hoc tests for multi-condition comparisons

    • Use non-parametric alternatives (Kruskal-Wallis) when normality cannot be assumed

    • Calculate biological (not just technical) replicates across different clutches of embryos

  • For Phenotypic Analyses:

    • Conduct pilot studies to determine appropriate sample sizes and statistical power

    • Utilize paired statistical tests when analyzing unilateral mutants (comparing mutant and wild-type sides)

    • Apply chi-square tests for categorical phenotypes

    • Consider mixed-effects models for longitudinal developmental data

  • For Imaging Data:

    • Employ blind scoring protocols to avoid observer bias

    • Develop objective quantification metrics rather than subjective assessments

    • Use machine learning approaches for high-throughput phenotypic analysis when possible

  • For Multi-omics Integration:

    • Apply gene set enrichment analysis for transcriptomic data

    • Utilize network analysis for protein interaction studies

    • Consider Bayesian approaches for integrating diverse data types

During phenotypic experiment design, plan and conduct pilot studies to help estimate appropriate sample sizes and investigate variability extent, as recommended for Xenopus genetic studies .

How can the study of Xenopus laevis ASUN contribute to understanding human genetic disorders?

The study of ASUN in Xenopus has significant translational potential for human disorders:

  • Neurological Disorders:

    • Nuclear positioning defects have been implicated in neurodevelopmental disorders

    • Xenopus models of ASUN dysfunction can reveal fundamental mechanisms of nuclear migration in neural development

    • Phenotypes in Xenopus often more closely recapitulate human conditions than rodent models

  • Reproductive Disorders:

    • ASUN plays critical roles in spermatogenesis and nuclear migration during gametogenesis

    • Xenopus studies can elucidate mechanisms underlying human fertility issues

    • Compare findings with known human ASUN variants associated with reproductive dysfunction

  • Developmental Anomalies:

    • ASUN's role in nuclear positioning affects multiple developmental processes

    • The rapid development and optical clarity of Xenopus embryos facilitate detailed analysis of developmental defects

    • CRISPR-based strategies in Xenopus have proven successful in studying congenital anomalies

  • Therapeutic Testing Platform:

    • Xenopus embryos absorb small molecules from culture medium, facilitating drug screening

    • Develop assays using ASUN mutants to identify compounds that rescue associated phenotypes

    • Screen FDA-approved drugs for repurposing potential for ASUN-related disorders

The evolutionary position of Xenopus between aquatic vertebrates and land tetrapods makes it particularly valuable for distinguishing conserved disease mechanisms from species-specific adaptations .

What are the most promising approaches for integrating ASUN protein studies with RNA-binding and transcriptional regulation research?

Integrating ASUN studies with RNA biology in Xenopus offers several promising directions:

  • RNA Immunoprecipitation Approaches:

    • Apply RNA immunoprecipitation (RIP) or CLIP-seq techniques to identify ASUN-associated RNAs, similar to methods used for the ESCRT-II complex

    • Compare binding profiles across developmental stages to identify stage-specific interactions

    • Analyze for enriched sequence motifs, similar to how Vps25 was found to recognize polypurine motifs

  • Functional RNA Interactions:

    • Test whether ASUN, like some nuclear envelope proteins, participates in mRNA localization

    • Investigate potential roles in RNA stability or translation regulation

    • Utilize Xenopus egg extracts, which provide a robust system for studying RNA-protein interactions

  • Transcriptional Impacts:

    • Perform RNA-seq in ASUN-depleted embryos to identify affected pathways

    • Use CRISPR-activation or repression systems to modulate ASUN levels

    • Compare transcriptional changes in unilateral mutants to distinguish direct from indirect effects

  • Nuclear Architecture Connection:

    • Investigate whether ASUN influences gene expression through effects on nuclear positioning

    • Apply chromosome conformation capture techniques (Hi-C) to examine chromatin organization

    • Determine if ASUN affects the localization of specific gene loci within the nucleus

These approaches take advantage of the conservation of RNA-binding properties across species, as demonstrated by studies of other Xenopus proteins .

How can multi-omics approaches be effectively applied to study ASUN protein networks in Xenopus development?

For comprehensive analysis of ASUN networks in Xenopus development:

  • Integrated Experimental Design:

    • Generate consistent ASUN perturbations (CRISPR knockout, knockdown, overexpression)

    • Sample across key developmental timepoints

    • Process parallel samples for different omics platforms from the same experimental batches

  • Recommended Multi-omics Pipeline:

    Omics ApproachApplication to ASUN ResearchKey Consideration
    TranscriptomicsGlobal expression changes and pathway analysisInclude tissue-specific and temporal analysis
    ProteomicsASUN interactome and post-translational modificationsUse Xenopus-specific databases for annotation
    PhosphoproteomicsSignaling pathways affected by ASUNConsider developmental stage-specific phosphorylation patterns
    ChIP-seqChromatin association if ASUN affects nuclear organizationOptimize fixation protocols for Xenopus tissues
    ATAC-seqChromatin accessibility changesCompare wildtype and ASUN-mutant sides in unilateral mutants
    MetabolomicsDownstream metabolic consequencesAdjust extraction protocols for amphibian samples
  • Data Integration Strategies:

    • Apply network analysis to identify functional modules

    • Utilize machine learning approaches to identify patterns across data types

    • Develop Xenopus-specific resources that integrate findings with existing knowledge

  • Validation in Human Systems:

    • Compare findings to human datasets from related disorders

    • Test conserved interactions in human cell lines

    • Utilize the evolutionary position of Xenopus to identify core conserved mechanisms

This comprehensive approach leverages the unique advantages of Xenopus, including the ability to generate thousands of mutant embryos per day, enabling truly parallelized analysis .

What are promising applications of single-cell technologies for studying ASUN protein function in Xenopus development?

Single-cell technologies offer revolutionary approaches to understanding ASUN function:

  • Single-cell RNA Sequencing Applications:

    • Compare transcriptomes between ASUN-mutant and wild-type cells in mosaic embryos

    • Track developmental trajectories affected by ASUN perturbation

    • The established protocols for isolating nuclei from Xenopus mutant lines allow efficient single-cell gene expression analyses

  • Spatial Transcriptomics Integration:

    • Map ASUN-dependent gene expression changes in spatial context

    • Correlate with developmental defects in specific regions

    • Leverage the large cell size in Xenopus embryos for increased spatial resolution

  • Lineage Tracing Approaches:

    • Combine CRISPR perturbation with genetic barcoding

    • Track cell fate decisions influenced by ASUN function

    • Utilize unilateral mutations to compare developmental outcomes under identical conditions

  • Single-cell Proteomics and Epitope Tagging:

    • Develop methods to detect ASUN protein levels and modifications at single-cell resolution

    • Correlate protein expression with phenotypic outcomes

    • Apply multiplexed antibody-based approaches to map pathway activation

These approaches build upon the existing strengths of Xenopus as a developmental model, particularly the ability to perform targeted genetic perturbations and analyze the consequences at cellular resolution.

How might the study of ASUN in Xenopus inform the development of targeted therapeutics for related human disorders?

Leveraging Xenopus ASUN research for therapeutic development:

  • Drug Screening Platforms:

    • Develop phenotype-based screens using ASUN-mutant Xenopus embryos

    • Leverage the ability of Xenopus embryos to absorb small molecules from culture medium

    • Screen compound libraries for rescue of ASUN-associated developmental defects

    • Establish quantitative behavioral assays as functional readouts

  • Gene Therapy Approaches:

    • Test delivery methods for ASUN-targeting nucleic acids in Xenopus

    • Optimize gene editing strategies using Xenopus as a rapid in vivo testing system

    • Evaluate off-target effects of potential therapeutic gene editing approaches

  • Precision Medicine Strategies:

    • Express human ASUN variants in Xenopus to classify variants of uncertain significance

    • Identify genetic background effects that modify ASUN-related phenotypes

    • Develop predictive models of therapeutic response based on genetic modifiers

  • Pathway-Based Interventions:

    • Target downstream pathways identified through multi-omics approaches

    • Test combinatorial treatments addressing multiple aspects of ASUN dysfunction

    • Evaluate developmental stage-specific interventions for maximum efficacy

These approaches build on the established track record of exceptional success using CRISPR-based strategies in Xenopus to study various disorders including autism spectrum disorders and congenital anomalies .

What ethical considerations should researchers address when designing Xenopus ASUN studies with translational implications?

When designing Xenopus ASUN research with translational goals:

  • Animal Welfare Considerations:

    • Implement the 3Rs principle (Replacement, Reduction, Refinement)

    • Utilize the unilateral mutation approach to reduce animal numbers while maintaining statistical power

    • Develop in vitro alternatives using Xenopus egg extracts where appropriate

    • Ensure proper anesthesia and humane endpoints for all procedures

  • Rigor and Reproducibility Standards:

    • Preregister study designs and analysis plans when possible

    • Report all experimental conditions completely, including Xenopus strain information

    • Validate key findings across multiple experimental approaches

    • Consider sex as a biological variable in post-metamorphic studies

  • Translational Validity Assessment:

    • Clearly communicate limitations of the model for human applications

    • Validate key findings in human cells or other mammalian models before clinical extrapolation

    • Acknowledge evolutionary divergence when interpreting results

    • Consider ecological and environmental implications of genetic modifications

  • Data Sharing and Resource Development:

    • Contribute to Xenopus community resources and databases

    • Share protocols, reagents, and mutant lines with the research community

    • Develop standardized phenotyping approaches for consistent reporting

    • Support the development of Xenopus-specific research tools and resources

By addressing these considerations, researchers can maximize the translational impact of Xenopus ASUN studies while maintaining ethical standards and scientific integrity.

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.