Recombinant Human Uncharacterized Protein C2orf66, commonly referred to as C2orf66, is a protein encoded by the C2orf66 gene located on chromosome 2 in humans. Despite its designation as "uncharacterized," this protein is part of ongoing research efforts aimed at understanding its potential functions and implications in human biology and disease.
Gene Location: The C2orf66 gene is situated on chromosome 2, which is one of the largest human chromosomes and contains numerous genes involved in various biological processes .
Protein Function: Currently, the specific biological functions of the C2orf66 protein remain largely uncharacterized. This means that while it is known to be a protein-coding gene, its precise role in cellular processes or disease mechanisms is not well understood .
Recombinant Protein: Recombinant proteins are produced through genetic engineering techniques where the gene encoding the protein is inserted into an expression vector and then expressed in a host organism (e.g., bacteria, yeast, or mammalian cells). This allows for large-scale production of the protein for research or therapeutic purposes .
| Research Area | Methodology | Potential Outcomes |
|---|---|---|
| Bioinformatics | Sequence alignment, structural prediction | Insights into potential protein functions or interactions |
| Cellular Studies | Immunofluorescence, Western blotting | Understanding protein localization and expression patterns |
| Functional Assays | Cell culture experiments, knockout models | Determining the protein's role in cellular processes or disease |
NCBI Gene Database: For information on the C2orf66 gene and its genomic context.
UniProt: Provides details on the protein sequence and any known functional data.
GeneCards: Offers comprehensive information on the gene, including potential pathways and disorders.
MyBioSource: Supplies recombinant C2orf66 protein for research purposes.
C2orf66 remains classified as an uncharacterized protein encoded by a gene located on chromosome 2 in humans. Like many proteins studied in immunological contexts, its functional characterization requires systematic experimental approaches combining genetic, biochemical, and cell biological methods. When approaching uncharacterized proteins, researchers typically begin with sequence analysis tools to identify conserved domains and potential functional motifs, similar to approaches used for characterizing novel immune signaling proteins. Protein prediction algorithms can suggest potential structural elements, but these must be validated experimentally through techniques like those used to characterize TLR-interacting proteins.
When producing recombinant uncharacterized proteins like C2orf66, selection of an appropriate expression system is critical. For initial characterization studies, bacterial expression systems (E. coli) offer high yield but may not provide proper folding or post-translational modifications. For functional studies, mammalian expression systems are preferred despite lower yields.
The methodology for mammalian cell expression typically involves:
Cloning the C2orf66 gene into a mammalian expression vector with appropriate promoter
Transfecting HEK293T or CHO cells using optimized transfection protocols
Selecting stable transfectants using antibiotic selection
Verifying expression using Western blotting
This approach parallels methods used for expressing recombinant human cytokines in research settings, where proper folding and post-translational modifications are critical for functional studies.
Purification of recombinant C2orf66 typically employs affinity chromatography approaches, with methodology varying based on the expression system and fusion tags employed:
| Purification Step | Methodology | Expected Outcome | Quality Control |
|---|---|---|---|
| Affinity Chromatography | Ni-NTA for His-tagged C2orf66 | >80% purity | SDS-PAGE |
| Size Exclusion | Superdex 200 column | >90% purity, removal of aggregates | Dynamic light scattering |
| Ion Exchange | Resource Q column | >95% purity | Protein staining |
| Endotoxin Removal | Polymyxin B columns | <0.1 EU/μg protein | LAL assay |
For functional studies, ensuring removal of endotoxin contamination is particularly important as it can activate TLR4 signaling pathways and confound experimental results, similar to concerns when studying pathogen recognition receptor pathways.
Validation of recombinant C2orf66 requires multiple analytical approaches:
SDS-PAGE and Western blotting with anti-tag antibodies for initial confirmation
Mass spectrometry for definitive protein identification and assessment of modifications
Circular dichroism to confirm proper secondary structure formation
Size exclusion chromatography to assess oligomeric state
Thermal shift assays to determine protein stability
These approaches provide complementary evidence of protein identity and structural integrity before proceeding to functional studies.
Identifying interaction partners represents a critical step in characterizing uncharacterized proteins like C2orf66. Several complementary approaches should be employed:
| Approach | Methodology | Advantages | Limitations |
|---|---|---|---|
| Yeast Two-Hybrid | Screen against human cDNA libraries | Unbiased discovery | High false-positive rate |
| Co-Immunoprecipitation | Pull-down with anti-tag antibodies | Detects native complexes | Requires high-quality antibodies |
| Proximity Labeling | BioID or APEX2 fusion constructs | Maps protein neighborhoods | Requires genetic manipulation |
| Crosslinking MS | Chemical crosslinking with MS analysis | Captures transient interactions | Complex data analysis |
| Surface Plasmon Resonance | Immobilized C2orf66 with candidate ligands | Quantitative binding kinetics | Requires candidate selection |
These methodologies parallel approaches used to identify novel binding partners in immune signaling pathways. For example, similar techniques revealed that bacterial tRNA can interact with TLR8 to trigger innate immune responses, demonstrating how binding partner identification can reveal unexpected functional roles.
Investigating potential signaling roles requires systematic perturbation studies:
Generate cell lines overexpressing C2orf66 and measure activation of common signaling pathways (NF-κB, MAPK, JAK-STAT) using reporter assays
Perform C2orf66 knockdown or knockout studies using siRNA or CRISPR/Cas9
Conduct phosphoproteomic analysis comparing wild-type and C2orf66-deficient cells
Examine C2orf66 expression changes during cellular activation with various stimuli
This methodological approach parallels studies of novel immune signaling components, where researchers examine how protein perturbation affects downstream signaling cascades. For instance, studies of TLR8 signaling revealed its unique ability to induce secretion of IL-12p70 in the absence of signals from other receptors, demonstrating how targeted signaling studies can reveal specialized functions.
Computational analysis provides crucial guidance for experimental studies of uncharacterized proteins:
| Computational Approach | Tool Examples | Information Provided |
|---|---|---|
| Sequence Homology | BLAST, HHpred | Related proteins with known functions |
| Domain Prediction | InterProScan, SMART | Conserved functional domains |
| Structural Modeling | AlphaFold2, I-TASSER | Predicted 3D structure |
| Intrinsic Disorder | PONDR, IUPred | Regions lacking stable structure |
| Post-translational Modification | NetPhos, GPS | Potential modification sites |
| Subcellular Localization | DeepLoc, PSORT | Predicted cellular compartment |
These computational predictions should guide experimental design but require validation. This parallels approaches used in characterizing novel immune signaling components, where computational analysis often provides initial hypotheses about protein function that are subsequently tested experimentally.
Selection of relevant biological systems is critical for functional characterization:
Begin with expression analysis across tissue and cell types using publicly available RNA-seq databases
Prioritize cell types with highest expression
Consider disease contexts where C2orf66 expression is altered
Include immune cells (monocytes, macrophages, dendritic cells) given the importance of protein discovery in immune contexts
Develop reporter cell lines from relevant cell types
This systematic approach to biological system selection parallels successful strategies used in characterizing novel components of immune signaling pathways, where understanding cell type-specific expression patterns provides crucial context for functional studies.
CRISPR/Cas9 approaches offer powerful tools for functional characterization:
Design multiple sgRNAs targeting different exons of C2orf66 to ensure complete knockout
Create tagged versions of C2orf66 at endogenous loci using HDR-mediated knock-in
Generate conditional knockout models to study tissue-specific functions
Perform CRISPR screens to identify genetic interactions
Use CRISPRi/CRISPRa to modulate expression without complete deletion
The guide RNA design process should prioritize specificity while minimizing off-target effects:
| Consideration | Methodology | Validation Approach |
|---|---|---|
| Target Selection | Target conserved exons, avoid alternative splice sites | Sequence verification |
| Off-target Prediction | Use algorithms (CRISPOR, CHOPCHOP) | Whole genome sequencing |
| Delivery Method | Lentiviral vs. nucleofection based on cell type | Delivery efficiency measurement |
| Clonal Selection | Single-cell sorting and expansion | Genotyping PCR and sequencing |
| Phenotype Validation | Rescue experiments with wildtype C2orf66 | Functional assays |
When facing conflicting experimental results, systematic troubleshooting is essential:
Carefully examine differences in experimental systems (cell types, stimulation conditions)
Verify reagent quality and specificity, particularly antibodies
Implement orthogonal methodologies to validate findings
Consider genetic background effects in different cell lines
Evaluate protein interaction networks in different contexts
These approaches parallel strategies used in resolving contradictory findings in immune signaling research. For instance, apparently conflicting reports about TLR8 polymorphisms and tuberculosis susceptibility were resolved through meta-analysis and population-specific studies, demonstrating how methodological rigor can clarify seemingly contradictory results.
Rigorous control implementation is critical for reliable characterization:
Use multiple cell lines to avoid cell type-specific artifacts
Include empty vector/untransfected controls in overexpression studies
Employ scrambled/non-targeting controls in knockdown experiments
Validate knockout phenotypes with rescue experiments
Include positive controls for pathway activation in signaling studies
Implement biological replicates across independent experiments
These control strategies parallel approaches used in immune signaling research, where careful control implementation is essential for distinguishing specific effects from experimental artifacts.
Post-translational modification (PTM) analysis requires integrated approaches:
| PTM Type | Detection Methodology | Functional Validation |
|---|---|---|
| Phosphorylation | Phospho-specific antibodies, LC-MS/MS with phosphopeptide enrichment | Phosphomimetic and phosphodeficient mutants |
| Ubiquitination | Ubiquitin pull-down, K-ε-GG antibody enrichment | Lysine-to-arginine mutants |
| Glycosylation | Glycosidase treatment, lectin binding, glycoproteomic MS | N/O-glycosylation site mutants |
| SUMOylation | SUMO-IP, MS analysis | Consensus site mutations |
| Acetylation | Anti-acetyl-lysine antibodies, MS | Lysine-to-arginine mutants |
Understanding PTMs can provide critical insights into protein regulation and function, as demonstrated in studies of immune signaling proteins where phosphorylation events often serve as molecular switches that control pathway activation.
Development of specific antibodies against uncharacterized proteins requires careful design:
Perform epitope prediction to identify antigenic regions unique to C2orf66
Generate multiple antibodies against different regions (N-terminal, C-terminal, internal domains)
Use both peptide and recombinant protein immunization strategies
Rigorously validate antibody specificity using knockout cells and overexpression controls
Characterize each antibody for specific applications (Western blot, IP, flow cytometry)
These approaches parallel successful strategies used in developing antibodies against novel immune signaling components, where antibody specificity is critical for accurately interpreting experimental results.
Connecting uncharacterized proteins to disease contexts requires integrative approaches:
Analyze expression patterns in disease-relevant tissues using public databases
Examine genetic variants in C2orf66 associated with disease phenotypes
Investigate protein interactions with known disease-associated proteins
Perform pathway analysis to position C2orf66 within known disease networks
Consider evolutionary conservation as an indicator of functional importance
This systematic approach parallels strategies used to connect novel immune signaling components to disease processes. For example, polymorphisms in TLR8 were associated with altered progression of HIV and tuberculosis through genome-wide association studies, demonstrating how genetic approaches can reveal unexpected disease connections.
Selection of appropriate model systems is critical for translational studies:
| Model System | Advantages | Limitations | Best Applications |
|---|---|---|---|
| Cell Lines | Genetic manipulation, high throughput | Limited physiological relevance | Mechanistic studies, initial screening |
| Primary Human Cells | Physiological relevance | Donor variability, limited lifespan | Validation of mechanisms in human context |
| Mouse Models | In vivo systems, genetic manipulation | Species differences | Physiological studies, disease models |
| Patient Samples | Direct disease relevance | Limited availability, experimental constraints | Correlation studies, biomarker validation |
| Organoids | 3D architecture, tissue-specific functions | Technical challenges, limited throughput | Complex cellular interactions |
The selection of appropriate model systems should be guided by the specific research question and the translational goals of the study.
Troubleshooting strategies for difficult-to-express proteins include:
Optimize codon usage for the expression system
Test multiple fusion tags and their placement (N-terminal vs. C-terminal)
Evaluate different cell lysis and extraction buffers
Consider expressing individual domains if full-length protein is unstable
Implement co-expression with potential binding partners
Test expression at lower temperatures to improve folding
These approaches parallel strategies used to overcome expression challenges with immune signaling proteins, where protein structure and stability often present technical hurdles.
When facing experimental inconsistencies:
Standardize experimental protocols across all studies
Validate key reagents, particularly antibodies and recombinant proteins
Implement positive and negative controls in each experiment
Consider cell density, passage number, and culture conditions as variables
Evaluate timing of measurements for dynamic processes
Employ multiple methodologies to measure the same outcome
These troubleshooting approaches reflect best practices in immune signaling research, where experimental standardization is essential for generating reproducible results.