Coiled-coil domain-containing protein 25 (CCDC25) is a protein that, in humans, is encoded by the CCDC25 gene located on chromosome 8p21.1 . The protein is evolutionarily conserved among mammals and eukaryotes, such as Arabidopsis thaliana . Although the precise function of CCDC25 remains unclear, research suggests its involvement in cell signaling and regulation .
CCDC25 consists of a single polypeptide chain that is 208 amino acids in length, with a molecular weight of approximately 25 kDa . It is highly expressed in human tissues, with a nearly ubiquitous expression pattern, and is predicted to localize to the nucleus . The protein contains coiled-coil domains, which are known to mediate protein-protein interactions .
CCDC25 is widely expressed in mammalian cells, including hepatocytes and muscle cells . While it has not been detected in healthy bile duct epithelial cells, microarray experiments have shown that CCDC25 is upregulated in Metaphase II oocytes .
CCDC25 interacts with Smad2, a latent transcription factor involved in the TGF-β signaling pathway . It is also a predicted substrate for several kinases, including Casein Kinase 1, Casein Kinase 2, cGMP-dependent protein kinase, and the Insulin Receptor, all of which play roles in cell signaling .
CCDC25 has been found to be upregulated in cholangiocarcinoma (CCA) tissues, and serum CCDC25 levels may serve as a diagnostic marker for CCA . It regulates cell proliferation, migration, and invasion in clear cell renal cell carcinoma . Additionally, the cell division cycle 25 (Cdc25) protein family, to which CCDC25 belongs, is crucial in controlling cell proliferation, making it a potential target for cancer therapy .
TMCC1 (transmembrane and coiled-coil domain family 1) is an endoplasmic reticulum (ER) protein that contains coiled-coil domains . These domains facilitate homo- or hetero-dimerization or oligomerization with other TMCC proteins . TMCC1 interacts with ribosomal proteins through its coiled-coil domains .
Zebrafish ccdc25 (also known as zgc:64173) is a protein-coding gene that encodes coiled-coil domain containing protein 25 . While detailed structural analysis of zebrafish ccdc25 remains limited, the protein belongs to the coiled-coil domain family characterized by alpha-helical structures that form supercoiled regions facilitating protein-protein interactions.
Current genomic data indicates that zebrafish ccdc25 contains conserved coiled-coil motifs that likely mediate its biological functions through interaction with other proteins. The predicted protein structure shares homology with human CCDC25, which is a 208-amino acid protein with a molecular weight of approximately 25 kDa .
Comparative analysis methodology:
Sequence analysis using MUSCLE or CLUSTAL alignments to identify conserved regions
Secondary structure prediction using PSIPRED or similar tools
Domain identification using InterProScan or SMART databases
Homology modeling based on related coiled-coil domain proteins
While the search results don't provide specific temporal expression data for ccdc25 in zebrafish, insights can be gained from expression patterns of other genes in zebrafish. For developmental gene expression profiling, techniques similar to those used for analyzing slc39a14 (which is expressed during embryonic and early larval development between 3-120 hpf ) would be applicable.
For determining spatial expression patterns, methodologies include:
Whole-mount in situ hybridization (WISH) with RNA probes specific to ccdc25 at key developmental stages (similar to techniques used for slc39a14 )
Fluorescent in situ hybridization for co-localization studies
Transgenic reporter lines with the ccdc25 promoter driving fluorescent protein expression
RT-qPCR on dissected tissues to quantify expression levels
Previous studies of coiled-coil domain proteins in zebrafish have revealed diverse expression patterns, ranging from ubiquitous to highly tissue-specific. For example, studies of CCDC56 demonstrated its essential role in mitochondrial function, particularly in cytochrome c oxidase (COX) activity .
Based on successful strategies used for other zebrafish genes, CRISPR/Cas9 genome editing provides the most efficient approach for generating ccdc25 knockout models.
Recommended CRISPR/Cas9 protocol for ccdc25 knockout:
gRNA design:
Target early exons to ensure complete loss of function
Use algorithms like CHOPCHOP or CRISPRscan to design multiple gRNAs with high specificity
Validate gRNA efficiency using in vitro assays
Microinjection procedure:
Inject 2-3 nl of solution containing:
300 ng/μl Cas9 mRNA or 500 ng/μl Cas9 protein
25-50 ng/μl gRNA
0.05% phenol red as injection tracer
Target one-cell stage embryos
Mutation screening:
Establishing mutant lines:
Raise F0 injected fish to adulthood
Outcross to wild-type fish to identify germline transmission
Incross F1 heterozygotes to generate homozygous mutants for phenotypic analysis
For complete loss of function studies, additional strategies may be needed to address maternal contribution, such as generating maternal-zygotic mutants through germline replacement.
Characterizing protein-protein interactions is crucial for understanding ccdc25 function. Based on approaches used for other zebrafish proteins, the following methodological strategy is recommended:
Step-by-step interaction analysis protocol:
Bait protein preparation:
Generate epitope-tagged ccdc25 constructs (FLAG, HA, or GFP tags)
Express recombinant protein using bacterial or cell-free systems
Alternative: Create transgenic zebrafish expressing tagged ccdc25
Primary interaction screening:
Co-immunoprecipitation (Co-IP) followed by mass spectrometry
Yeast two-hybrid screening
BioID or proximity labeling approaches
Validation methods:
Reciprocal Co-IP experiments
GST pulldown assays with recombinant proteins
BiFC (Bimolecular Fluorescence Complementation) in zebrafish cells
In vivo confirmation:
Co-localization studies using confocal microscopy
FRET (Förster Resonance Energy Transfer) analysis
Genetic interaction studies through combined knockdown/knockout approaches
Given the coiled-coil structure of ccdc25, interactions are likely mediated through these domains, which should be considered when designing truncation constructs for domain-specific interaction mapping.
While specific information about ccdc25's role in zebrafish development is limited, insights can be drawn from research on other zebrafish proteins. Cell proliferation is compartmentalized at the posterior end of zebrafish embryos through regulated expression of mitotic factors like Cdc25a . Given that some coiled-coil domain proteins are involved in cell cycle regulation, ccdc25 might have similar functions.
Experimental approaches to investigate developmental roles:
Cell proliferation analysis:
BrdU or EdU incorporation assays in wild-type vs. ccdc25 mutants
Phospho-histone H3 immunostaining to detect mitotic cells
Cell cycle analysis using FACS in dissociated embryonic cells
Developmental phenotyping:
Detailed morphological analysis at key developmental stages
Time-lapse imaging of embryonic development
Lineage tracing using photoconvertible fluorescent proteins
Molecular pathway analysis:
Gene expression profiling using RNA-seq
Assessment of cell cycle regulators (cyclins, CDKs)
Analysis of developmental signaling pathways (Wnt, Notch, FGF)
Stat3/Cdc25a-dependent cell proliferation promotes embryonic axis extension during zebrafish gastrulation , suggesting that proteins involved in cell cycle regulation can have significant impacts on developmental processes. If ccdc25 functions in similar pathways, it might influence axis formation, gastrulation movements, or tissue-specific development.
Human CCDC25 has been implicated in several disease contexts, including hepatocellular carcinoma , cholangiocarcinoma , and metabolic syndrome . Zebrafish models can provide valuable insights into these conditions through comparative functional studies.
For disease modeling, key approaches include:
Generating ccdc25 mutations that mirror human disease variants
Creating liver-specific or other tissue-specific manipulations of ccdc25 expression
Combining ccdc25 manipulation with established disease models
Conducting drug screens to identify compounds that modulate ccdc25-dependent phenotypes
Understanding the subcellular localization of ccdc25 is crucial for elucidating its function. While CCDC25 has been reported on the cell surface of cancer cells in humans , its localization in zebrafish has not been specifically described.
Comprehensive localization analysis protocol:
Fluorescent fusion proteins:
Generate N- and C-terminal fluorescent protein fusions (GFP, mCherry)
Create stable transgenic lines with tissue-specific promoters
Validate functionality of fusion proteins through rescue experiments
Subcellular marker co-localization:
Co-expression with established organelle markers:
Nucleus: H2B-BFP
Mitochondria: MitoTracker or Tom20-RFP
ER: Sec61-mCherry
Golgi: GalT-RFP
Cell membrane: membrane-targeted fluorescent proteins
Super-resolution microscopy:
STED or PALM/STORM imaging for nanoscale localization
Live cell imaging to capture dynamic trafficking events
FRAP (Fluorescence Recovery After Photobleaching) to assess protein mobility
Biochemical fractionation:
Cell fractionation followed by Western blotting
Density gradient centrifugation for membrane subdomains
Protease protection assays for membrane topology
Similar approaches have been successfully used to determine subcellular localization of other zebrafish proteins, such as CCDC56 which was shown to localize to mitochondria .
Scientific research often produces seemingly contradictory results due to methodological differences or biological complexity. For ccdc25 research, systematic approaches can help reconcile discrepancies.
Strategies for resolving contradictory findings:
Methodological standardization:
Document complete experimental conditions including:
Zebrafish strain and genetic background
Developmental stage and temperature
Detailed protocol parameters
Use multiple detection methods (e.g., different antibodies, tagged constructs)
Implement blinded analysis where possible
Genetic and biological factors:
Test for genetic modifiers by using different zebrafish backgrounds
Consider maternal vs. zygotic contributions
Evaluate tissue-specific effects through conditional approaches
Assess dosage sensitivity through heterozygote analysis
Integration of multiple approaches:
Combine knockdown, knockout, and overexpression studies
Utilize both in vivo and in vitro systems
Perform time-course experiments to capture dynamic processes
Apply systems biology approaches to model complex interactions
Collaborative validation:
Independent replication in different laboratories
Data sharing through repositories
Multi-lab consortium approaches for complex phenotypes
Systematic documentation and reporting of negative results can be particularly valuable for resolving contradictions in emerging research areas.
Modern high-throughput technologies offer powerful approaches for rapidly advancing understanding of ccdc25 biology in zebrafish.
High-throughput methodologies applicable to ccdc25 research:
Transcriptomic approaches:
Single-cell RNA-seq to identify cell populations expressing ccdc25
Spatial transcriptomics to map expression in intact tissues
Time-series RNA-seq to capture dynamic expression changes
Differential expression analysis in ccdc25 mutants
Proteomic strategies:
Proximity labeling coupled with mass spectrometry
Phosphoproteomics to identify post-translational modifications
Thermal proteome profiling to identify protein interactions
CETSA (Cellular Thermal Shift Assay) for drug target engagement
Functional genomics:
CRISPR screens to identify genetic interactors
Chemical-genetic interaction screens
Enhancer/suppressor screens in sensitized backgrounds
Synthetic lethality approaches
High-content imaging:
Automated phenotyping of embryonic development
Machine learning-based image analysis
4D live imaging with computational tracking
High-throughput behavioral analysis
| Technology | Application to ccdc25 | Expected outcome | Key considerations |
|---|---|---|---|
| Single-cell RNA-seq | Cell type-specific expression profiling | Identification of ccdc25-expressing cell populations | Requires optimized dissociation protocols |
| BioID proximity labeling | Protein interaction network mapping | Comprehensive interactome of ccdc25 | Requires validation of key interactions |
| CRISPR screening | Genetic interaction identification | Genes that modify ccdc25 phenotypes | Screen design critical for success |
| Automated high-content imaging | Phenotypic profiling | Quantitative assessment of developmental impacts | Requires robust image analysis pipelines |
Based on human studies showing CCDC25's potential as a biomarker for multiple conditions , zebrafish models could be valuable for therapeutic development.
Therapeutic development framework:
Target validation in zebrafish:
Phenotypic rescue experiments with chemical compounds
Structure-function studies to identify critical domains
Conditional expression systems to define therapeutic windows
Drug discovery approaches:
Small molecule screens using embryo-based phenotypic assays
In silico docking to identified binding pockets
Peptide inhibitors targeting protein-protein interactions
Antisense approaches for targeted knockdown
Delivery system development:
Nanoparticle-based delivery testing in zebrafish
Tissue-specific targeting strategies
Evaluation of off-target effects
PK/PD studies in larval and adult zebrafish
Translational considerations:
Conservation of drug targets between zebrafish and humans
Biomarker development for treatment response
Combination therapies targeting multiple pathway components
Repurposing of approved drugs that modulate ccdc25 pathways
Given the association of CCDC25 with metabolic syndrome , liver cancer , and cholangiocarcinoma , therapeutic approaches targeting this protein could potentially address multiple pathological conditions.
Based on current knowledge and technological capabilities, several promising research directions emerge:
Developmental biology:
Characterize spatial and temporal expression patterns throughout development
Determine cell type-specific functions in key developmental processes
Investigate potential roles in tissue patterning and morphogenesis
Molecular mechanisms:
Define the ccdc25 interactome in different cellular contexts
Identify post-translational modifications and their functional significance
Determine structure-function relationships through domain analysis
Disease modeling:
Develop zebrafish models for CCDC25-associated human diseases
Investigate cancer-related functions, particularly in liver cancers
Explore potential roles in metabolic regulation
Therapeutic applications:
Screen for compounds that modulate ccdc25 function
Develop targeted approaches for disease intervention
Explore ccdc25 as a biomarker for disease progression or treatment response
The emerging role of human CCDC25 as a potential marker for conditions like metabolic syndrome suggests that zebrafish models could provide valuable insights into both basic biology and translational applications.