KEGG: xla:380474
UniGene: Xl.3177
Xenopus laevis nudcd1.S is a protein-coding gene also known as cml66. As an S homeolog in the allotetraploid X. laevis genome, it represents one of the two copies resulting from genome duplication. The gene has the Entrez Gene ID 380474 . Due to the allotetraploid nature of X. laevis (in contrast to the diploid X. tropicalis), researchers should consider both homeologs when designing experiments. The genomic structure reflects evolutionary conservation of NudC domain-containing proteins, which are characterized by specific coiled-coil domains that mediate protein-protein interactions essential for function.
Expression and purification of recombinant Xenopus laevis nudcd1 can be accomplished through several validated approaches:
Bacterial expression systems: Use E. coli BL21(DE3) with pET vectors containing the Xenopus nudcd1 coding sequence. Optimize induction conditions (IPTG concentration, temperature, duration) to maximize soluble protein yield.
Mammalian expression systems: HEK293T cells transfected with vectors containing Xenopus nudcd1 often produce properly folded protein with post-translational modifications.
Purification strategy:
His-tagged recombinant nudcd1 can be purified using Ni-NTA affinity chromatography
GST-tagged constructs can be purified via glutathione-sepharose
Ion exchange chromatography followed by size exclusion chromatography yields high purity
For functional studies, researchers should verify protein integrity through SDS-PAGE, Western blotting, and activity assays before experimental use.
Several experimental approaches are available for investigating nudcd1 function in Xenopus:
Xenopus egg extracts: Meiotic Xenopus egg extracts provide an excellent system for studying protein function in microtubule organization and cell cycle regulation. This system has been successfully used for related NudC domain proteins and could be applied to nudcd1 .
Morpholino knockdown: Antisense morpholino oligonucleotides can be injected into Xenopus embryos to transiently knock down nudcd1 expression during early development.
CRISPR/Cas9 genome editing: Modern genome editing tools have been successfully applied in Xenopus models to generate gene knockouts or mutations .
mRNA overexpression: Injection of in vitro transcribed nudcd1 mRNA allows for gain-of-function studies.
Xenopus transgenic models: Stable transgenic lines expressing tagged versions of nudcd1 or under tissue-specific promoters provide long-term models.
| Experimental Approach | Advantages | Limitations | Typical Applications |
|---|---|---|---|
| Egg extracts | Cell-free system, biochemical accessibility | Limited to early developmental processes | Protein interactions, microtubule studies |
| Morpholino knockdown | Rapid, targeted gene knockdown | Transient effects, potential off-targets | Early developmental studies |
| CRISPR/Cas9 | Permanent genetic modification | More technically challenging | Long-term functional studies |
| mRNA overexpression | Quick gain-of-function | Transient effects, potential overexpression artifacts | Rescue experiments, structure-function analysis |
| Transgenic models | Stable expression, tissue-specific studies | Time-consuming to generate | Long-term, tissue-specific analysis |
While direct evidence for nudcd1's role in Xenopus microtubule organization is limited, insights can be gained from studies of related NudC domain proteins. In Xenopus egg extracts, the N-terminal coiled-coil domain of Ndel1 (another NudC-related protein) serves as a regulated scaffold for dynein-dependent microtubule self-organization . This interaction requires binding to both dynein and LIS1, suggesting a conserved mechanism that may apply to nudcd1.
Methodology for investigating nudcd1-microtubule interactions:
Co-immunoprecipitation: Using antibodies against nudcd1 to pull down potential interacting partners like dynein components and LIS1 from Xenopus egg extracts.
Microtubule co-sedimentation assays: Determine if recombinant nudcd1 directly binds microtubules or affects their polymerization kinetics.
Aster formation assays: Ran-mediated aster formation in Xenopus egg extracts with and without nudcd1 depletion/addition can reveal functional roles in microtubule organization .
Live imaging: Fluorescently tagged nudcd1 in Xenopus cells allows visualization of dynamic interactions with the cytoskeleton during cell division or migration.
Based on research in cancer models, nudcd1 likely participates in proliferation and metastasis-related signaling pathways such as IGF1R-ERK1/2 . To investigate these pathways in Xenopus:
Phosphorylation analysis: Western blot analysis using phospho-specific antibodies can detect activation of potential downstream effectors like ERK1/2 following nudcd1 overexpression or knockdown in Xenopus cells or embryos.
Pathway inhibition studies: Using small molecule inhibitors of specific pathway components (e.g., ERK inhibitors) in conjunction with nudcd1 manipulation can reveal pathway dependencies.
Gene expression profiling: RNA-seq analysis of nudcd1-depleted versus control Xenopus embryos or cells can identify affected gene networks and signaling pathways.
Epistasis experiments: Sequential knockdown of nudcd1 and pathway components can determine hierarchical relationships.
Studies in non-Xenopus systems have shown that NudCD1 promotes phosphorylation of IGF1R and ERK1/2 proteins , suggesting its role in activating these proliferation-associated pathways. This provides a starting point for Xenopus-specific investigations.
When designing knockdown experiments for nudcd1 in Xenopus laevis, researchers should consider:
Target specificity: Due to the allotetraploid nature of X. laevis, both homeologs (L and S) of nudcd1 may need to be targeted. Sequence alignment of both homeologs is essential for designing effective knockdown reagents.
Knockdown methods:
Morpholinos: Design against the translation start site or splice junctions. Typically inject 10-20 ng of morpholino at the 1-2 cell stage.
CRISPR/Cas9: Design gRNAs targeting conserved exons in both homeologs. Co-inject with Cas9 protein (500 pg) or mRNA (300 pg).
shRNA: Design using algorithms that predict efficient knockdown. For cultured cells, viral transduction achieves high efficiency.
Controls:
Include mismatch or scrambled controls
Perform rescue experiments with morpholino-resistant mRNA
Validate knockdown efficiency by qRT-PCR and Western blot
Phenotypic analysis:
Based on cancer studies, examine proliferation using EdU or BrdU incorporation
Assess migration/invasion through transplantation assays
Examine cell cycle progression and apoptosis markers
Similar approaches for NudCD1 knockdown in cancer cells using shRNA have revealed its roles in proliferation, migration, and invasion , which can guide experimental design in Xenopus.
Research on nudcd1 in Xenopus provides valuable insights into human disease mechanisms, particularly cancer:
Cancer biology: NudCD1 functions as an oncoprotein in multiple cancer types, including non-small cell lung cancer and pancreatic cancer . Xenopus models offer a system to study the developmental context of these oncogenic functions.
Comparative analysis:
Pathway conservation: The IGF1R-ERK1/2 pathway modulated by NudCD1 in cancer cells is conserved in Xenopus, allowing for comparative studies.
Therapeutic targeting: Understanding nudcd1 function in Xenopus can help identify conserved domains or interactions as potential therapeutic targets.
The unique advantages of Xenopus for disease modeling include rapid embryonic development, external fertilization, and amenability to genetic manipulation . These features facilitate high-throughput screening and in vivo validation of mechanisms identified in human disease studies.
Understanding structure-function relationships of nudcd1 is crucial for designing informative experiments:
Domain analysis: NudC domain proteins typically contain:
N-terminal regions with regulatory functions
Central NudC domain mediating protein-protein interactions
C-terminal regions with variable functions
Truncation constructs: Generate and express different fragments of Xenopus nudcd1 to determine:
Which domains are essential for specific functions
Regions required for protein-protein interactions
Domains necessary for subcellular localization
Point mutations: Introduce site-specific mutations in conserved residues to assess:
Effects on protein stability and folding
Impact on binding to interaction partners
Changes in signaling pathway activation
Chimeric proteins: Exchange domains between nudcd1 and other NudC domain proteins to determine functional conservation.
Structural analysis: Use recombinant protein for:
X-ray crystallography or cryo-EM structural determination
Circular dichroism to assess secondary structure
Limited proteolysis to identify stable domains
These approaches can be combined with functional assays (proliferation, migration, protein interaction) to correlate structure with specific cellular functions, similar to studies in cancer models where NudCD1 promotes cell proliferation and metastasis via specific signaling pathways .
For accurate detection and differentiation of endogenous versus recombinant nudcd1:
Western blotting:
For endogenous detection: Use antibodies against conserved epitopes of Xenopus nudcd1
For recombinant detection: Epitope tags (His, FLAG, HA) allow specific recognition
Quantification: Densitometry analysis with loading controls (β-actin, GAPDH)
Immunohistochemistry/Immunofluorescence:
Tissue fixation: 4% paraformaldehyde for embryos, methanol for better epitope preservation
Antibody optimization: Titration experiments to determine optimal concentration
Controls: Include no-primary controls and pre-immune serum controls
qRT-PCR:
Design primers spanning exon-exon junctions to avoid genomic DNA amplification
For recombinant constructs: Target vector-specific sequences or tags
Normalization: Use validated reference genes (ef1α, odc1) for Xenopus
Protein mass spectrometry:
Identify post-translational modifications
Distinguish between L and S homeologs
Quantify relative abundance of endogenous versus recombinant protein
| Detection Method | Sensitivity | Specificity | Quantitative Capability | Sample Requirements |
|---|---|---|---|---|
| Western blot | Medium-High | High with validated antibodies | Semi-quantitative | Protein lysates (50-100 μg) |
| Immunostaining | Medium | Variable, depends on antibody | Qualitative | Fixed tissue/cells |
| qRT-PCR | Very High | High with optimized primers | Highly quantitative | RNA (50-100 ng) |
| Mass spectrometry | High | Very High | Quantitative | Purified protein (μg range) |
Similar detection methods have been successfully applied in studies of NudCD1 in cancer tissues and cell lines .
Common challenges in recombinant nudcd1 expression and solutions include:
Low solubility:
Modify buffer conditions (pH, salt concentration, detergents)
Express as fusion protein with solubility enhancers (MBP, SUMO, thioredoxin)
Lower induction temperature (16-20°C) to slow folding
Use lysis buffers containing mild detergents (0.1% Triton X-100)
Poor yield:
Optimize codon usage for expression system
Test different promoters or expression systems
Use Terrific Broth or auto-induction media for bacterial expression
Scale up culture volume or cell density
Degradation:
Add protease inhibitors during purification
Express truncated stable domains
Identify and mutate protease-sensitive sites
Reduce purification time and temperature
Lack of activity:
Ensure proper folding through circular dichroism analysis
Add co-factors or binding partners during purification
Use gentle elution conditions
Test different tags and their positions (N- vs C-terminal)
Aggregation:
Include stabilizing agents (glycerol, arginine)
Perform size exclusion chromatography to remove aggregates
Adjust protein concentration and storage conditions
Consider refolding protocols if necessary
Validation techniques include thermal shift assays to assess stability, dynamic light scattering to detect aggregation, and functional assays based on known activities of nudcd1 in cellular proliferation and migration .
When analyzing proliferation and migration data in nudcd1 studies:
Proliferation assays:
EdU/BrdU incorporation: Quantify percentage of positive cells across multiple fields
MTT/CCK-8: Generate growth curves over 24-72 hours
Colony formation: Count and measure colony size and number
Cell cycle analysis: Determine changes in cell cycle distribution by flow cytometry
Studies in cancer cells demonstrate that NudCD1 knockdown impedes proliferation , providing a benchmark for expected effects.
Migration/invasion assays:
Wound healing: Measure closure rate over time
Transwell assays: Count cells that traverse the membrane
3D matrix invasion: Analyze distance and directionality of migration
NudCD1 knockdown has been shown to inhibit migration and invasion of cancer cells , suggesting similar functions may exist in Xenopus models.
Data normalization and statistics:
Use appropriate normalization to control samples
Apply statistical tests suitable for data distribution (t-test, ANOVA)
Present both biological and technical replicates (n≥3)
Calculate effect sizes and confidence intervals, not just p-values
Pathway analysis:
Interpretation challenges:
Distinguish between direct and indirect effects
Consider developmental context in Xenopus studies
Account for compensatory mechanisms in knockdown models
Integrate findings with existing knowledge of nudcd1 function
When comparing nudcd1 function between Xenopus and other species:
Evolutionary conservation:
Sequence homology analysis: Align nudcd1 sequences across species to identify conserved domains
Phylogenetic analysis: Determine evolutionary relationships and potential functional divergence
Domain structure: Compare organization of functional domains across species
Functional equivalence testing:
Cross-species rescue experiments: Can human NudCD1 rescue Xenopus nudcd1 knockdown phenotypes?
Heterologous expression: Express Xenopus nudcd1 in mammalian cells to assess function
Chimeric proteins: Create fusion proteins with domains from different species to test domain-specific functions
Contextual differences:
Signaling pathway conservation:
Experimental system differences:
Cell-free systems (Xenopus egg extracts) versus cellular systems
Temperature considerations (Xenopus optimal temperature ~18-22°C vs. mammalian 37°C)
Developmental rate differences between species
By systematically accounting for these considerations, researchers can make valid cross-species comparisons and identify truly conserved functions of nudcd1.
Several cutting-edge technologies show promise for advancing nudcd1 research:
Single-cell analysis:
scRNA-seq to identify cell populations affected by nudcd1 manipulation
Single-cell proteomics to detect protein level changes
Spatial transcriptomics to map nudcd1 expression patterns in developing embryos
Advanced imaging:
Light sheet microscopy for whole-embryo imaging with minimal phototoxicity
Super-resolution microscopy to visualize subcellular localization
Live imaging with optogenetic tools to manipulate nudcd1 function with spatiotemporal precision
CRISPR technologies:
Protein interaction mapping:
BioID or APEX proximity labeling to identify nudcd1 interactome
FRET/BRET sensors to detect dynamic protein interactions
Hydrogen-deuterium exchange mass spectrometry for structural dynamics
Organoid models:
Development of Xenopus organoids to study nudcd1 in organ-specific contexts
Co-culture systems to examine cell-cell interactions mediated by nudcd1
These technologies will enable more precise dissection of nudcd1 function in development and disease models, building upon current understanding of its roles in proliferation, migration, and signaling pathway activation .
Xenopus nudcd1 research has several potential applications for therapeutic development:
Target validation:
Xenopus models provide in vivo validation of nudcd1 as a therapeutic target
Developmental phenotypes can reveal potential side effects of targeting NudCD1
Genetic interaction studies can identify synthetic lethal relationships
Drug discovery:
High-throughput screening using Xenopus embryos or egg extracts
Structure-based drug design using recombinant Xenopus nudcd1
Phenotypic rescue assays to validate compound efficacy
Pathway targeting:
Biomarker development:
Precision medicine approaches:
Patient-derived xenografts in Xenopus to test personalized therapies
CRISPR-engineered Xenopus models carrying patient-specific mutations
Rapid testing of drug resistance mechanisms