Recombinant Danio rerio Ankyrin Repeat Domain-Containing Protein 13C (ANKRD13C) is a protein derived from zebrafish (Danio rerio), expressed through recombinant DNA technology. This protein is part of the ankyrin repeat domain family, which plays a crucial role in protein-protein interactions and is involved in various cellular processes.
Origin: Zebrafish (Danio rerio)
Expression System: Yeast
Tag: His tag for purification and detection
Sequence: Comprises amino acids 1-488
Ankyrin repeat proteins are characterized by their modular structure, consisting of repeating units that form a specific binding interface. Each repeat typically folds into two antiparallel α-helices followed by a β-hairpin or a long loop, creating an L-shaped domain that facilitates protein-protein interactions .
Protein-Protein Interactions: Ankyrin repeats mediate interactions with other proteins, which are crucial for various cellular functions such as signaling, transcription regulation, and cytoskeleton integrity .
Disease Association: Mutations in ankyrin repeat proteins have been linked to several diseases, including cancers and genetic disorders .
Biological Studies: ANKRD13C can be used in studies focusing on protein-protein interactions and cellular signaling pathways.
Therapeutic Targets: Understanding the function of ANKRD13C could provide insights into potential therapeutic targets for diseases related to ankyrin repeat proteins.
ANKRD13C is expressed in yeast, which offers an efficient eukaryotic system for protein production. The His tag facilitates purification using affinity chromatography, making it suitable for various biochemical assays .
| Expression System | Advantages | Disadvantages |
|---|---|---|
| Yeast | Economical, efficient for eukaryotic proteins | Limited post-translational modifications compared to mammalian systems |
| Mammalian Cells | High-quality proteins with natural modifications | High cost, low expression levels |
| Baculovirus | High yield, suitable for complex proteins | Requires insect cells |
Functional Characterization: Investigating the specific functions and interactions of ANKRD13C.
Disease Association: Exploring potential links between ANKRD13C and diseases related to ankyrin repeat proteins.
Functions as a molecular chaperone for G protein-coupled receptors, regulating their biogenesis and trafficking from the endoplasmic reticulum.
ANKRD13C is a protein characterized by ankyrin repeat domains that functions as a molecular chaperone for G protein-coupled receptors (GPCRs). In zebrafish and other organisms, it regulates the folding and maturation of newly synthesized GPCRs . This protein is primarily associated with the cytosolic side of endoplasmic reticulum (ER) membranes, where it interacts with the cytoplasmic C-terminus of GPCRs .
The functional significance of ANKRD13C includes:
Promotion of GPCR biogenesis by inhibiting degradation of newly synthesized receptors
Regulation of protein exit from the ER
Control of protein trafficking through the biosynthetic pathway
Retention of misfolded/unassembled forms of receptors in the ER, directing them to proteasome-mediated degradation
Research methodology for functional characterization typically involves protein interaction studies, subcellular localization experiments, and functional assays measuring receptor expression levels under conditions of ANKRD13C overexpression or knockdown.
Zebrafish ANKRD13C is a 488 amino acid protein that contains multiple functional domains . While the search results don't provide the complete structural characterization specifically for zebrafish ANKRD13C, we can infer from related ankyrin repeat domain-containing proteins that the structure likely includes:
Ankyrin repeat domains - critical for protein-protein interactions
Potential PEST sequences - involved in rapid intracellular proteolysis
Possible coiled-coil motifs - important for protein oligomerization
Nuclear localization signals (NLS) may be present or absent depending on specific function
The full amino acid sequence begins with: MTGEKIRSVR KERKSGLDLL EPDEEPAATG PAKHRGSKIF SGGNHRISRS SSSPGDPDGA YP...
Methodological approaches to study ANKRD13C structure include recombinant protein expression (typically in yeast systems for eukaryotic proteins), followed by purification and structural analysis through techniques such as X-ray crystallography or NMR spectroscopy.
While the search results don't provide specific developmental expression data for ANKRD13C in zebrafish, we can gain insights from the expression patterns of related ankyrin repeat domain-containing proteins. For example, ankrd1a and ankrd1b (counterparts of mammalian ANKRD1) show distinct spatiotemporal expression during development:
ankrd1a shows mild increase at 72 hpf (1.74±0.24 fold increase relative to 24 hpf)
ankrd1b is markedly upregulated from 24 hpf onward, peaking at 72 hpf (92.18±36.95 fold increase relative to 24 hpf)
These paralogs exhibit non-overlapping expression patterns during skeletal muscle development, with ankrd1a predominantly in trunk somites and ankrd1b in tail somites
To study ANKRD13C expression patterns, researchers should employ:
Quantitative PCR (qPCR) with appropriate reference genes (e.g., rpl13a as used for other ankyrin repeat proteins)
In situ hybridization to visualize spatial expression
Developmental time-course experiments from early embryogenesis through adult stages
For optimal expression of recombinant zebrafish ANKRD13C, the following methodological approach is recommended:
Expression System Selection:
The yeast expression system is particularly suitable for zebrafish ANKRD13C as it combines economic efficiency with the eukaryotic post-translational modifications necessary for proper protein folding . This system enables modifications such as glycosylation, acylation, and phosphorylation that ensure native protein conformation.
Protein Characteristics for Optimal Expression:
Addition of purification tag: His-tag for affinity purification
Expression vector: Choose vectors optimized for yeast expression with inducible promoters
Purification Protocol:
Cell lysis under non-denaturing conditions to preserve protein structure
Affinity chromatography using Ni-NTA columns for His-tagged proteins
Size exclusion chromatography for further purification
Quality control through SDS-PAGE and Western blotting
Researchers should aim for >90% purity for experimental applications . Alternative expression systems (E. coli, mammalian cells, or baculovirus infection) may be considered depending on specific experimental requirements, though these have trade-offs in terms of cost, yield, and post-translational modifications.
ANKRD13C functions as a molecular chaperone for GPCRs, with several key interaction mechanisms identified through cellular studies:
Interaction Mechanism:
ANKRD13C directly binds to the cytoplasmic C-terminus of GPCRs
This interaction occurs at the endoplasmic reticulum membrane on the cytosolic side
The binding shows specificity toward GPCRs, as ANKRD13C does not affect the expression of unrelated proteins like GFP, GRK2, or VSVG
Functional Consequences of Interaction:
Initial increase in receptor protein levels upon co-expression
Inhibition of degradation of newly synthesized receptors
Prolonged interaction results in ER retention of misfolded/unassembled receptors
Direction of misfolded receptors to proteasome-mediated degradation
Experimental Approaches to Study Interactions:
Co-immunoprecipitation to detect physical interaction
Yeast two-hybrid screening (which originally identified ANKRD13C as a GPCR-interacting protein)
Fluorescence microscopy showing co-localization with ER markers
Pulse-chase experiments to track receptor biogenesis and degradation
siRNA knockdown studies to assess effects of reduced ANKRD13C expression
In Vivo Zebrafish Model Preparation:
Select adult zebrafish (both sexes, aged 60-90 days, sizes 3.5 ± 0.5 cm and weight 0.4 ± 0.1 g)
Maintain in appropriate conditions: glass aquariums (30 x 15 x 20 cm), dechlorinated water with air pumps and submerged filters, at 25°C and pH 7.0, with a 14-day circadian cycle (10 h light/dark)
Acclimate for at least 24 hours before experiments
Gene Expression Analysis:
Quantitative PCR (qPCR) using appropriate reference genes (e.g., rpl13a)
Reaction conditions: initial denaturation at 95°C for 10 min, 40 cycles of denaturation at 95°C for 15 s, annealing and elongation at 60°C
Data analysis using the 2^(-ΔΔCt) method for relative quantification
Genetic Manipulation Approaches:
Morpholino knockdown for transient loss-of-function
CRISPR/Cas9 for generating stable genetic models
Transgenic overexpression using tissue-specific promoters
Protein Analysis Techniques:
Western blotting with specific antibodies
Immunohistochemistry for tissue localization
Co-immunoprecipitation for protein interaction studies
Functional Studies:
Tissue-specific expression analysis in response to physiological stimuli
Exercise protocols to study stress responses (e.g., forced swimming for 3 hours, twice daily, for five consecutive days)
Behavioral assays to assess phenotypic outcomes
While direct data on ANKRD13C responsiveness to stress is limited in the search results, insights can be drawn from related ankyrin repeat proteins. The effect of physical stress (endurance exercise) on ankyrin repeat proteins in zebrafish has been documented:
Exercise Protocol Impact on Ankyrin Repeat Proteins:
Protocol: Two 3-hour bouts of forced swimming daily for five consecutive days
Tissue-specific responses measured 3 hours after the final exercise bout:
Methodological Approach for Studying Stress Responses:
Design appropriate stress models (e.g., exercise, oxidative stress, temperature variation)
Tissue sampling at different time points post-stress
Gene expression analysis via qPCR with reference genes like rpl13a
Protein analysis through Western blotting
Subcellular localization studies using immunofluorescence
Based on the function of ankyrin repeat proteins as stress-responsive elements, researchers should consider examining ANKRD13C in the context of:
Mechanical stress responses in muscle tissue
Oxidative stress conditions
Metabolic stress scenarios
Temperature stress paradigms
When designing experiments with recombinant zebrafish ANKRD13C, researchers should implement the following controls:
For Expression Studies:
Empty vector controls to account for expression system effects
Unrelated protein controls (e.g., GFP) to demonstrate specificity
Wild-type vs. mutated ANKRD13C to identify functional domains
Dose-response experiments to determine optimal protein concentrations
For Interaction Studies:
Non-GPCR membrane proteins as negative controls
Multiple GPCR subtypes to assess interaction specificity
Truncated ANKRD13C constructs lacking specific domains
Competition assays with known interacting proteins
For Functional Assays:
ER stress inducers to assess role in stress response
Pulse-chase experiments with and without ANKRD13C expression
siRNA knockdown with rescue experiments using recombinant protein
For In Vivo Studies:
Age-matched and sex-balanced zebrafish groups
Vehicle-only treatments in parallel with experimental conditions
Behavioral monitoring to assess systemic effects
To systematically compare zebrafish ANKRD13C with mammalian orthologs, researchers should implement the following methodological approaches:
Sequence and Structural Analysis:
Multiple sequence alignment to identify conserved domains
Phylogenetic analysis to understand evolutionary relationships
Synteny analysis to examine genomic context conservation
Protein structure prediction and comparison
Based on related ankyrin repeat proteins, expect moderate sequence conservation between species, with higher conservation in functional domains like ankyrin repeats . For example, zebrafish Ankrd1a, Ankrd1b, and Ankrd2 show 56%, 46%, and 51% identity with their human counterparts, respectively .
Functional Comparison:
Parallel expression studies in zebrafish and mammalian cells
Cross-species rescue experiments
GPCR interaction profiles across species
Comparative response to cellular stressors
Experimental Design Table for Cross-Species Comparison:
| Analysis Type | Zebrafish System | Mammalian System | Assessment Methods |
|---|---|---|---|
| Expression Pattern | In situ hybridization, qPCR | Immunohistochemistry, RNA-seq | Tissue distribution, developmental timing |
| Subcellular Localization | Fluorescent tagging in zebrafish cells | Fluorescent tagging in mammalian cells | Confocal microscopy, subcellular fractionation |
| Protein Interactions | Co-IP from zebrafish tissues | Co-IP from mammalian tissues | Mass spectrometry, Western blot |
| Functional Impact | Morpholino/CRISPR in zebrafish | siRNA/CRISPR in mammalian cells | Receptor trafficking, protein degradation assays |
| Stress Response | Exercise protocol for zebrafish | Mechanical/oxidative stress in cell culture | qPCR, Western blot, proteomics |
When facing conflicting results from different expression systems for zebrafish ANKRD13C, researchers should systematically evaluate:
Source of Variation Analysis:
Expression system differences: Yeast expression provides eukaryotic post-translational modifications that may be critical for proper ANKRD13C folding and function, while bacterial systems may yield higher amounts but lack these modifications
Protein tag influence: His-tag location (N- vs C-terminal) may differentially affect protein function
Protein purity variations: Aim for >90% purity for consistent results
Buffer composition effects on protein stability and activity
Methodological Approach to Resolve Conflicts:
Side-by-side comparison of proteins from different expression systems using multiple functional assays
Structural analysis to identify potential differences in protein folding
Mass spectrometry to confirm post-translational modifications
Limited proteolysis to assess protein conformation
Decision Table for Expression System Selection:
| Research Goal | Recommended System | Advantages | Limitations |
|---|---|---|---|
| Structural studies | Yeast | Native-like folding, post-translational modifications | Moderate yield |
| High-throughput screening | E. coli | High yield, cost-effective | May lack proper folding/modifications |
| Functional studies | Yeast or mammalian | Most physiologically relevant | Lower yield, higher cost |
| In vivo applications | Mammalian | Minimal immunogenicity, complete modifications | Highest cost, lowest yield |
Common Experimental Challenges and Solutions:
Low Protein Expression Yields
Problem: Recombinant ANKRD13C may express poorly in certain systems
Solution: Optimize codon usage for expression system, adjust induction conditions, consider fusion partners to enhance solubility
Protein Aggregation
Problem: ANKRD13C may form aggregates during expression or purification
Solution: Express at lower temperatures, include stabilizing agents in buffers, consider detergents for membrane-associated portions
Non-specific Interactions
Problem: His-tagged proteins may show non-specific binding in interaction studies
Solution: Include imidazole controls, perform reciprocal co-IP, validate with multiple detection methods
Inconsistent qPCR Results
Developmental Variability
Problem: Expression timing may vary between zebrafish clutches
Solution: Precise staging of embryos, larger sample sizes, consistent husbandry conditions
Methodological Controls to Implement:
Include wild-type controls alongside recombinant protein experiments
Perform dose-response studies to identify optimal protein concentrations
Validate antibody specificity through knockdown/knockout controls
Include positive controls with known behavior in each experimental set
Several cutting-edge technologies hold significant potential for advancing our understanding of zebrafish ANKRD13C:
CRISPR/Cas9 Genome Editing:
Generate precise knockouts or domain-specific mutations in zebrafish ANKRD13C
Create fluorescent protein fusions at endogenous loci for live imaging
Implement tissue-specific or inducible CRISPR systems for temporal control
Advanced Imaging Approaches:
Super-resolution microscopy to visualize ANKRD13C-GPCR interactions at the nanoscale
Live cell imaging with optogenetic tools to manipulate ANKRD13C activity
Light-sheet microscopy for whole-organism visualization of ANKRD13C dynamics
Proteomics Applications:
Proximity labeling (BioID, APEX) to identify the complete ANKRD13C interactome
Quantitative proteomics to measure changes in the proteome upon ANKRD13C manipulation
Cross-linking mass spectrometry to map interaction interfaces
Computational Methods:
Molecular docking studies to predict ANKRD13C-GPCR binding, similar to approaches used for other proteins in zebrafish
AlphaFold or RoseTTAFold for accurate structure prediction
Network analysis to position ANKRD13C within cellular signaling pathways
Single-Cell Technologies:
scRNA-seq to identify cell populations expressing ANKRD13C during development
Spatial transcriptomics to map expression patterns with tissue context
Cell-specific proteomics to measure ANKRD13C levels across different cell types
Research on zebrafish ANKRD13C offers valuable insights into fundamental aspects of protein quality control:
Evolutionary Conservation of Chaperone Functions:
Comparing ANKRD13C function across species can reveal evolutionarily conserved mechanisms of protein quality control
The moderate conservation observed in related ankyrin repeat proteins (46-56% identity between zebrafish and human) suggests both conserved and divergent aspects of function
GPCR-Specific Quality Control:
ANKRD13C selectively regulates GPCR biogenesis and trafficking, distinguishing it from general chaperones
Research can reveal how specialized chaperones recognize specific protein families
Understanding this selectivity may lead to targeted therapeutic approaches for GPCR-related diseases
ER-Associated Degradation (ERAD) Mechanisms:
This provides a model system to study how quality control decisions (fold vs. degrade) are made
The balance between retention, folding assistance, and degradation reveals fundamental principles of proteostasis
Stress Response Integration: