C17orf110 belongs to the growing class of microproteins encoded by small open reading frames (smORFs). Emerging evidence highlights its role in calcium homeostasis and mitochondrial interactions:
C17orf110 (ELN) is identified as a SERCA pump inhibitor in nonmuscle cells, modulating calcium reuptake into the sarcoplasmic reticulum (SR). This activity mirrors other SERCA-regulating microproteins like phospholamban (PLN) and sarcolipin (SLN), though its tissue specificity differs .
C17orf110’s role in calcium regulation positions it as a potential therapeutic target for diseases involving dysregulated SERCA activity, such as muscular dystrophy or cardiac myopathies. Its recombinant availability facilitates:
Antibody Validation: Thermo Fisher’s control fragment (aa 2–22) enables neutralization experiments to confirm antibody specificity .
Interaction Studies: Co-immunoprecipitation or proximity ligation assays to map binding partners (e.g., SERCA pumps).
Functional Assays: Measurement of Ca²⁺ flux or mitochondrial bioenergetics in cellular models.
Despite progress, key gaps remain:
Tissue-Specific Roles: Limited data on its expression in human tissues beyond nonmuscle cells.
Mitochondrial Function: No direct evidence links C17orf110 to mtDNA maintenance or nucleoid dynamics.
Pathological Relevance: Association with diseases (e.g., cancer, neurodegeneration) requires validation.
C17orf110 (chromosome 17 open reading frame 110) is a protein-coding gene located on chromosome 17. This gene is also known as SMIM6 (small integral membrane protein 6) and ELN according to current annotations . The protein is classified among uncharacterized or poorly characterized proteins, with relatively limited functional data available in standard databases. According to Pharos classification, it belongs to the "dark genome" category of proteins about which relatively little is known .
Based on the validation data available, the following methodological approach is recommended for C17orf110 detection:
RT-qPCR methodology:
Use exonic primers for C17orf110 detection to avoid genomic DNA contamination
Implement SYBR Green-based detection with the validated primers showing 99% efficiency and 100% specificity
Include appropriate housekeeping genes for normalization
Follow standard melt curve analysis to confirm amplification specificity
Expression profiling by microarray:
When conducting broader expression studies:
Extract total RNA using TRI-REAGENT followed by RNeasy kit purification
Verify RNA quality using Agilent 2100 Bioanalyzer
Convert 10 μg of total RNA to double-stranded cDNA using oligo-dT primers containing T7 RNA polymerase promoter
Purify double-stranded cDNA by phenol/chloroform extraction
Follow standard hybridization protocols for microarray-based detection
Detection Method | Advantages | Limitations | Technical Considerations |
---|---|---|---|
RT-qPCR | High sensitivity, specific quantification | Limited to known transcript variants | Use validated primers with 99% efficiency |
Microarray | Genome-wide expression context | Lower sensitivity than RT-qPCR | Requires high-quality RNA (RIN >8) |
RNA-Seq | Detects novel transcripts, splice variants | Higher cost, complex analysis | Minimum 20M reads per sample recommended |
Western Blot | Protein-level validation | Requires specific antibodies | Limited commercial antibodies available |
Current knowledge about C17orf110/SMIM6 tissue distribution is limited, but preliminary data suggests:
Knowledge scores by tissue relevance:
Researchers investigating tissue expression should implement a systematic approach using:
Multi-tissue qPCR panels
Immunohistochemistry (if antibodies are available)
Mining of public RNA-seq datasets such as GTEx and TCGA
Single-cell RNA-seq data where available to identify cell-type specific expression
When producing recombinant C17orf110, researchers should consider the following methodological approach based on established protocols for small membrane proteins:
Expression vector selection:
For small membrane proteins like C17orf110/SMIM6, use vectors with strong promoters (T7, CMV) for sufficient expression
Consider ORF vectors with restriction enzyme-independent cloning methods between appropriate cut sites (similar to approaches used for other ORF vectors)
Include appropriate tags for detection and purification (His-tag, FLAG-tag)
Expression system recommendations:
For functional studies: Mammalian expression systems (HEK293, CHO) to ensure proper folding and post-translational modifications
For structural studies: E. coli systems with codon optimization or cell-free systems
For difficult-to-express membrane proteins: Consider insect cell (baculovirus) systems
Purification strategy:
A systematic approach to functional characterization should include:
Computational prediction:
Perform comparative sequence analysis (BLAST, HMM)
Utilize protein structure prediction (AlphaFold2)
Identify conserved domains and motifs
Analyze phylogenetic relationships
Localization studies:
Express fluorescently-tagged C17orf110 to determine subcellular localization
Verify with cell fractionation and Western blotting
Consider co-localization studies with organelle markers
Interaction studies:
Conduct yeast two-hybrid or BioID proximity labeling
Perform co-immunoprecipitation followed by mass spectrometry
Consider FRET/BRET for dynamic interaction studies
Loss-of-function studies:
Implement CRISPR-Cas9 knockout/knockdown
Analyze resulting phenotypes across multiple cell types
Perform transcriptomic and proteomic profiling of knockout models
Given that C17orf110/SMIM6 is annotated as a small integral membrane protein, researchers should consider methodologies specific to micropeptides:
Ribosome profiling:
Implement ribosome footprinting to verify translation
Use harringtonine or lactimidomycin to capture translation initiation sites
Analyze data with specialized tools for smORF detection
Mass spectrometry validation:
Employ targeted proteomics approaches (PRM/MRM)
Consider specialized sample preparation methods for small proteins
Use synthetic peptide standards for validation
Functional characterization specific to micropeptides:
When investigating potential disease relevance:
Gene expression analysis in disease cohorts:
Genetic variant analysis:
Analyze WES/WGS data for potentially pathogenic variants
Consider population-specific variant frequencies
Assess potential impact using predictive algorithms and functional validation
Model systems:
Develop appropriate cell and animal models (knockout, knockin)
Consider tissue-specific conditional models if global knockout is lethal
Implement phenotypic analysis across multiple physiological parameters
Researchers face several methodological challenges that can be addressed through specific approaches:
Limited prior knowledge:
Challenge: Absence of functional annotations and validated reagents
Solution: Implement systematic multi-omics approaches, starting with in silico predictions followed by experimental validation
Protein detection issues:
Challenge: Limited availability of specific antibodies
Solution: Generate epitope-tagged recombinant proteins; develop custom antibodies against synthetic peptides based on predicted epitopes
Functional redundancy:
Challenge: Potential compensation by related proteins masking phenotypes
Solution: Implement combinatorial knockouts; use acute depletion systems (AID, dTAG)
Publication challenges:
Challenge: Difficulty publishing on uncharacterized proteins
Solution: Focus on methodological rigor; present comprehensive characterization data; emphasize novelty aspects
Following best practices for scientific data presentation:
Table design principles for uncharacterized protein research:
Effective figure design:
Data presentation decision matrix:
Data Type | Best Presentation Format | Rationale |
---|---|---|
Precise numerical values | Tables | To show many numerical values in compact space |
Expression patterns across tissues | Heatmaps | To visualize patterns across multiple samples |
Protein localization | Fluorescence microscopy images | To document subcellular distribution |
Multiple experimental comparisons | Bar/line graphs with error bars | To show statistical significance across conditions |
Sequence features | Annotated sequence diagrams | To highlight functional domains and motifs |
Researchers should consider these cutting-edge approaches:
Spatial transcriptomics/proteomics:
Apply single-cell spatial technologies to map expression in tissue context
Implement multiplexed immunofluorescence to correlate with other markers
AlphaFold2/RoseTTAFold structure prediction:
Utilize AI-based structure prediction to inform functional hypotheses
Design validation experiments based on structural features
High-throughput CRISPR screens:
Implement genome-wide or focused CRISPR screens to identify genetic interactions
Use CRISPRi/CRISPRa approaches for reversible modulation
Organoid technologies:
Develop relevant organoid systems to study function in tissue-like context
Implement gene editing in organoids to assess phenotypic consequences
To advance collective understanding:
Data sharing recommendations:
Deposit all raw data in appropriate repositories (GEO, PRIDE)
Contribute to protein databases with experimental evidence
Consider preprints for rapid dissemination of findings
Collaborative approaches:
Engage with consortia focused on uncharacterized proteins
Implement standardized protocols for cross-laboratory validation
Consider multi-laboratory replication studies for key findings
Methodological transparency:
Document detailed protocols in repositories like protocols.io
Ensure complete reporting of negative results
Provide comprehensive methods sections with validation metrics