C1orf144 (also known as SZRD1, UPF0485 protein C1orf144, and DKFZp566C0424) is a human gene product encoded by C1orf144 on chromosome 1p36.13 . Its recombinant form is produced via heterologous expression systems for research purposes.
| Attribute | Detail |
|---|---|
| Gene Aliases | SZRD1, DKFZp566C0424 |
| UniProt ID | Q7Z422 |
| Predicted Function | SUZ domain-containing protein; putative MAPK-activating protein |
| Molecular Weight | ~17 kDa (fragment aa 75-151) |
Recombinant C1orf144 is produced using multiple hosts, each offering distinct advantages:
| Host System | Yield | Turnaround Time | Post-Translational Modifications |
|---|---|---|---|
| E. coli/Yeast | High | Short (weeks) | Minimal (no eukaryotic modifications) |
| Insect Cells | Moderate | Medium | Glycosylation, disulfide bonds |
| Mammalian Cells | Low | Long (months) | Full PTMs (e.g., phosphorylation) |
Source: Production methods optimized in
E. coli/Yeast: Preferred for high-throughput studies due to cost efficiency and speed .
Insect/Mammalian Cells: Critical for functional studies requiring proper folding and activity .
Despite its unknown function, C1orf144 has been investigated in molecular and cellular contexts:
MAPK Activation: Hypothesized to act as a MAPK-activating protein based on sequence homology .
RNA Binding: Contains a SUZ domain, suggesting possible RNA interaction .
Recombinant C1orf144 is used as:
Control Fragment: For blocking experiments in Western blot (WB) and immunohistochemistry (IHC) .
Antigen Source: Used to generate polyclonal antibodies (e.g., HPA024221, PA5-55018) .
| Application | Antibody Example | Technique | Concentration |
|---|---|---|---|
| Immunoblotting | PA5-55018 | WB | 1:1000 dilution |
| Immunohistochemistry | HPA024221 | IHC (tissue arrays) | 1:1000–1:2500 |
| Immunofluorescence | HPA024221 | IF | 0.25–2 μg/mL |
Sources: Antibody validation data from
C1orf144 (Chromosome 1 Open Reading Frame 144) is a protein-coding gene located on chromosome 1, which is the largest human chromosome spanning approximately 260 million base pairs and constituting about 8% of the human genome. The protein product has been provisionally designated as UPF0485 protein C1orf144, pending further characterization of its structure and function. The "UPF" prefix indicates an uncharacterized protein family, suggesting that its biological roles remain to be fully elucidated through systematic research .
Multiple expression systems can be employed for recombinant C1orf144 production, each with distinct advantages. E. coli and yeast expression systems typically offer superior yields and shorter production timelines, making them cost-effective choices for initial characterization studies. For applications requiring post-translational modifications that may be essential for proper protein folding or activity, insect cells with baculovirus expression systems or mammalian cell expression systems are recommended. These advanced systems can provide the necessary cellular machinery to ensure proper glycosylation, phosphorylation, and other modifications that might be critical for maintaining the native conformation and functionality of C1orf144 .
The structural characteristics of C1orf144 remain largely undefined, as indicated by its UPF (Uncharacterized Protein Family) designation. The protein belongs to the UPF0485 family, suggesting shared structural motifs with other members of this family. Preliminary structural analyses may involve computational prediction methods, circular dichroism spectroscopy to determine secondary structure content, and ultimately X-ray crystallography or NMR spectroscopy for high-resolution structural determination. Researchers should consider both the native protein structure and potential structural changes that might occur upon interaction with binding partners or activators .
C1orf144 activators constitute a specialized class of chemical compounds designed to modulate C1orf144 activity. These molecules typically function by interacting with specific regulatory elements within the C1orf144 gene, particularly in promoter or enhancer regions, to enhance transcription and subsequent translation into functional protein products. While the precise mechanisms vary among different activators, they fundamentally act as molecular switches that amplify C1orf144 activity. Some compounds, such as Troglitazone, may indirectly influence C1orf144 expression through activation of nuclear receptors like PPARγ, which regulate gene expression networks. Zinc may also play a role as a cofactor for various transcription factors that could potentially influence C1orf144 expression .
Designing experiments to determine C1orf144 function requires a multi-faceted approach:
Loss-of-function studies: Implement CRISPR-Cas9 gene editing, RNA interference (RNAi), or antisense oligonucleotides to downregulate or eliminate C1orf144 expression. Following knockdown or knockout, comprehensive transcriptomic and proteomic analyses can identify altered pathways and interacting partners.
Gain-of-function studies: Overexpress C1orf144 using appropriate vectors and promoters, then evaluate phenotypic changes through cell proliferation assays, metabolic assessments, or functional readouts specific to the cellular context being investigated.
Subcellular localization: Employ immunofluorescence, cell fractionation, or expression of fluorescently-tagged C1orf144 to determine its intracellular distribution, which can provide crucial insights into potential functions.
Interactome mapping: Conduct co-immunoprecipitation, yeast two-hybrid assays, or proximity-dependent biotin identification (BioID) to identify proteins that physically interact with C1orf144, establishing a network of potential functional associations .
Pathway analysis: Similar to investigations with related chromosome 1 proteins (like C1orf74), researchers should examine connections to established signaling pathways, such as MAPK signaling, which has been implicated in the function of other chromosome 1 proteins .
Purification of recombinant C1orf144 requires careful consideration of protein stability and activity:
Affinity chromatography: Design constructs with appropriate affinity tags (His-tag, GST, MBP) positioned to minimize interference with protein folding and function. Consider implementing protease cleavage sites to remove tags post-purification.
Buffer optimization: Systematically screen buffer compositions to identify conditions that maximize protein stability. Critical parameters include:
pH range (typically 6.5-8.0 for most proteins)
Salt concentration (150-500 mM NaCl)
Reducing agents (DTT, β-mercaptoethanol, TCEP)
Stabilizing additives (glycerol, arginine, trehalose)
Chromatographic sequence: Implement a multi-step purification strategy, potentially including ion exchange, size exclusion, and hydrophobic interaction chromatography after initial affinity purification to achieve high purity.
Aggregation prevention: Monitor protein aggregation using dynamic light scattering or size exclusion chromatography with multi-angle light scattering (SEC-MALS).
Activity assays: Develop specific activity assays based on predicted or identified functions to confirm that purified C1orf144 retains its biological activity throughout the purification process .
Posttranslational modifications (PTMs) potentially critical for C1orf144 function require special consideration:
PTM prediction and identification: Employ bioinformatic tools to predict potential modification sites, followed by mass spectrometry analysis of natively expressed protein to identify actual PTMs.
Expression system selection: Based on identified PTMs, select appropriate expression systems:
Phosphorylation: Mammalian cells or insect cells
Glycosylation: Mammalian cells for complex glycans; yeast for high-mannose structures
Acetylation/methylation: Mammalian cells
Co-expression strategies: Consider co-expressing relevant kinases, phosphatases, or other modifying enzymes to enhance specific PTM acquisition in recombinant systems.
Site-directed mutagenesis: Create PTM site mutants (e.g., phosphomimetic mutations) to assess the functional significance of specific modifications.
Preservation protocols: Incorporate phosphatase inhibitors, deacetylase inhibitors, or other PTM-preserving reagents during purification to maintain modification status .
Investigating C1orf144 gene regulation requires a comprehensive approach:
Promoter analysis: Conduct reporter gene assays using luciferase or fluorescent proteins to identify key regulatory elements in the C1orf144 promoter and enhancer regions.
Chromatin immunoprecipitation (ChIP): Identify transcription factors binding to the C1orf144 regulatory regions through ChIP-seq or ChIP-qPCR approaches.
CRISPR activation/interference: Employ CRISPRa or CRISPRi systems to specifically activate or repress transcription from the C1orf144 locus without altering the genetic sequence.
DNA methylation analysis: Use bisulfite sequencing or methylation-specific PCR to assess the role of DNA methylation in C1orf144 expression regulation.
Chromosome conformation capture: Implement 3C, 4C, or Hi-C techniques to identify long-range chromatin interactions affecting C1orf144 expression.
RNA stability assessment: Measure C1orf144 mRNA half-life using actinomycin D chase experiments coupled with qRT-PCR to determine post-transcriptional regulation mechanisms .
Antibody validation for C1orf144 research requires rigorous testing:
Specificity validation:
Western blot comparing wild-type to knockout/knockdown samples
Peptide competition assays to confirm epitope specificity
Immunoprecipitation followed by mass spectrometry to confirm target identity
Application-specific validation:
For immunohistochemistry: Tissue microarrays with positive and negative controls
For flow cytometry: Comparison with isotype controls and blocking experiments
For ChIP: Analysis of known non-target regions as negative controls
Cross-reactivity assessment: Test against related proteins, particularly other chromosome 1 open reading frame products, to ensure specificity.
Lot-to-lot consistency: Establish standard operating procedures for validating new antibody lots to ensure experimental reproducibility.
Reporting standards: Document validation results comprehensively according to the Antibody Validation Guidelines established by the International Working Group for Antibody Validation .
Selection of appropriate cell models depends on research objectives:
Expression profiling: Prior to model selection, analyze C1orf144 expression across tissue types using publicly available RNA-seq datasets to identify physiologically relevant models.
Primary cells vs. cell lines:
Primary cells offer physiological relevance but limited lifespan
Immortalized cell lines provide experimental convenience but may have altered signaling networks
Disease models: If investigating potential disease associations, select models that recapitulate relevant pathological features.
3D culture systems: Consider organoids or spheroids for more physiologically relevant microenvironments compared to traditional 2D cultures.
Genetic background considerations: Utilize isogenic cell lines for precise functional studies, particularly when conducting genetic manipulation experiments.
Validation across multiple models: Confirm key findings in at least two independent cell models to enhance result reliability .
Analysis of C1orf144 expression data requires a systematic approach:
Dataset selection and quality control:
Utilize multiple independent datasets (e.g., TCGA, GTEx, GEO)
Implement rigorous quality control measures for RNA integrity and sequencing depth
Account for batch effects using appropriate statistical methods
Differential expression analysis:
Apply appropriate statistical frameworks (DESeq2, edgeR, limma)
Use false discovery rate (FDR) correction for multiple testing
Establish biologically meaningful thresholds for fold change interpretation
Correlation analysis:
Identify genes with expression patterns correlated with C1orf144
Perform gene set enrichment analysis to identify associated pathways
Survival analysis:
Assess the prognostic significance of C1orf144 expression using Kaplan-Meier plots and Cox regression
Consider multivariate models incorporating clinical variables
Validation with quantitative PCR:
Confirm key expression differences using independent sample sets
Normalize to multiple reference genes for reliable quantification
| Data Analysis Method | Application to C1orf144 Research | Statistical Considerations |
|---|---|---|
| Differential Expression | Comparing C1orf144 levels between normal and disease tissues | FDR < 0.05, absolute log2FC > 1.0 |
| Co-expression Networks | Identifying genes functionally related to C1orf144 | WGCNA, PCC > 0.7 |
| Pathway Enrichment | Determining biological processes involving C1orf144 | GSEA, Reactome, KEGG analysis |
| Survival Analysis | Associating C1orf144 expression with clinical outcomes | Cox proportional hazards model |
| eQTL Analysis | Identifying genetic variants affecting C1orf144 expression | Linear regression, FDR < 0.05 |
This approach mirrors successful analyses performed for other chromosome 1 genes such as C1orf74, which demonstrated significant associations with clinical parameters in cancer contexts .
Elucidating structure-function relationships for poorly characterized proteins involves:
Sequence-based predictions:
Employ multiple alignment tools to identify conserved domains
Use structure prediction algorithms (AlphaFold2, RoseTTAFold) to generate structural models
Identify potential functional motifs through specialized databases (Prosite, PFAM)
Experimental structure determination:
Pursue X-ray crystallography for high-resolution static structures
Consider NMR spectroscopy for dynamic information and intrinsically disordered regions
Implement hydrogen-deuterium exchange mass spectrometry for conformational dynamics
Functional mapping:
Create systematic alanine scanning or domain deletion mutants
Assess effects on identified functions through relevant assays
Correlate structural features with functional outcomes
Molecular dynamics simulations:
Simulate protein dynamics under physiological conditions
Identify potential binding pockets or interaction surfaces
Validate computational predictions experimentally
Integrative approaches:
Investigating protein interactions and pathway connections requires integrated approaches:
Affinity purification-mass spectrometry (AP-MS):
Express tagged C1orf144 in relevant cell types
Perform immunoprecipitation under physiological conditions
Identify co-precipitating proteins through mass spectrometry
Validate key interactions through reciprocal co-IP or proximity ligation assay
Yeast two-hybrid and mammalian two-hybrid screening:
Identify direct protein-protein interactions
Validate positive hits with orthogonal methods
Map interaction domains through truncation constructs
Proximity labeling approaches:
Employ BioID or APEX2 fusion proteins to identify proximal proteins
Distinguish between stable and transient interactions
Map subcellular interaction networks
Phosphoproteomic analysis:
Compare global phosphorylation patterns between control and C1orf144-perturbed systems
Identify altered phosphosites to map affected signaling pathways
Validate through kinase inhibition studies
Transcriptomic response profiling:
Analyze gene expression changes following C1orf144 manipulation
Apply pathway enrichment analysis to identify regulated processes
Validate through reporter assays for key pathways
Based on research on related proteins, potential signaling connections may include MAPK pathway components, as demonstrated for other chromosome 1 proteins in cancer contexts .
Therapeutic targeting strategies for C1orf144 should consider:
Target validation:
Establish clear disease association through genetic evidence
Demonstrate modifiable phenotypes in disease-relevant models
Identify specific molecular mechanisms amenable to intervention
Small molecule modulators:
Screen for compounds that specifically activate or inhibit C1orf144
Optimize lead compounds for potency, selectivity, and pharmacokinetic properties
Evaluate compounds like Troglitazone that may indirectly modulate C1orf144 through nuclear receptor activation
RNA-based therapeutics:
Design antisense oligonucleotides or siRNAs for specific C1orf144 downregulation
Develop mRNA therapeutics for controlled expression in deficiency states
Address delivery challenges through lipid nanoparticles or conjugated targeting moieties
Protein-protein interaction disruptors:
Identify critical interaction surfaces through structural studies
Design peptide mimetics or small molecules to specifically disrupt pathological interactions
Validate target engagement in cellular and animal models
Gene therapy approaches:
Develop viral vectors for C1orf144 delivery in deficiency states
Consider gene editing for correction of pathogenic variants
Explore controlled expression systems for context-specific activation
Research on related chromosome 1 proteins suggests potential applications in cancer contexts, where altered expression has demonstrated significant associations with clinical outcomes and therapeutic responses .
Developing effective high-throughput screening assays requires:
Assay design considerations:
Select appropriate readouts based on known or predicted functions
Establish robust positive and negative controls
Optimize signal-to-background ratio and Z' factor (aim for Z' > 0.5)
Primary screening approaches:
Reporter gene assays for transcriptional effects
AlphaScreen or FRET assays for protein-protein interactions
Phenotypic screens in disease-relevant cell models
Confirmatory and counter-screening cascades:
Implement dose-response confirmations for active compounds
Conduct counter-screens to eliminate assay-specific artifacts
Assess specificity against related proteins in the same family
Mechanism of action determination:
Thermal shift assays to confirm direct binding
Competitive binding assays to identify binding sites
Cellular target engagement assays (CETSA, DARTS)
Data analysis and hit prioritization: