C14orf166 antibody (e.g., Proteintech 19848-1-AP) is a rabbit-derived polyclonal IgG antibody that targets the human, mouse, and rat C14orf166 protein . Key attributes include:
The antibody facilitates the study of C14orf166’s role in RNA polymerase II activation, viral replication (e.g., influenza A, hepatitis C), and tumor progression .
C14orf166 antibody is widely used in experimental workflows:
C14orf166 overexpression is strongly linked to aggressive tumor behavior and poor prognosis:
Similar trends are observed in breast, cervical, and nasopharyngeal cancers, where elevated C14orf166 levels predict tumor progression .
Viral Pathogenesis: C14orf166 interacts with influenza A and hepatitis C virus polymerases, enhancing viral RNA replication .
Oncogenic Pathways:
C14orf166 antibody enables the detection of this protein in serum and tissues, showing promise for:
This comprehensive FAQ collection addresses key research questions about C14orf166 antibodies for academic and clinical investigations. C14orf166 (Chromosome 14 Open Reading Frame 166) is a 28-kDa protein that localizes to both the nucleus and cytoplasm, playing crucial roles in RNA metabolism, viral infection responses, and cancer progression. Overexpression of C14orf166 has been associated with poor prognosis in multiple cancers, making it an important research target. This document organizes questions from basic to advanced, providing methodological guidance for researchers utilizing C14orf166 antibodies in their experimental workflows.
C14orf166, also known as CLE, hCLE, CGI-199, or RTRAF, is a highly conserved gene located on chromosome 14 at cytogenetic band 14q22.1. It encodes a 28-kDa protein that localizes to both the nucleus and cytoplasm . This protein serves multiple biological functions:
Acts as a core element of cytosolic RNA granules in neuronal processes
Functions in RNA metabolism as part of the human spliceosome and tRNA-splicing ligase complex
Interacts with 7SK snRNA methylphosphate capping enzyme
Participates in viral RNA replication and transcriptional activation
C14orf166 has gained significant attention in cancer research due to its:
Overexpression in multiple cancer types (NSCLC, cervical cancer, bladder cancer, hepatocellular carcinoma)
Strong correlation with advanced TNM stages, lymph node metastasis, and tumor size
Association with poor prognosis and shorter survival times
These characteristics make C14orf166 a promising biomarker for cancer progression and a potential therapeutic target.
Based on commercially available antibodies, C14orf166 can be detected using multiple techniques:
Researchers should validate these detection methods in their specific experimental systems, as reactivity may vary across species (human, mouse, rat) and sample types (cell lines, tissue sections) .
For optimal immunohistochemical detection of C14orf166 in formalin-fixed paraffin-embedded (FFPE) tissues:
Perform heat-mediated antigen retrieval using Tris/EDTA buffer at pH 9.0
Bring buffer to boiling point, then place sections in the heated buffer
Maintain at sub-boiling temperature for 15-20 minutes
Allow sections to cool gradually in the buffer for 20 minutes
Rinse thoroughly with PBS before proceeding with immunostaining
This protocol has been validated for human tissue samples including astrocytoma, fetal kidney, and various cancer tissues . Alternative approaches using citrate buffer (pH 6.0) may be tested if results are suboptimal, though they appear less effective for C14orf166 detection based on reported literature .
Thorough validation of C14orf166 antibody specificity is crucial for reliable research results:
Western blot analysis:
Knockdown/knockout validation:
Peptide competition assay:
Cross-reactivity assessment:
Immunoprecipitation followed by mass spectrometry:
Understanding the clinical correlations of C14orf166 expression is essential for designing experiments with clinical relevance:
C14orf166 expression correlates with:
These correlations suggest experimental designs should consider:
Stratification of samples based on clinical parameters
Inclusion of follow-up data when evaluating biomarker potential
Comparison of expression in primary tumors vs. metastatic sites
Correlation with other established biomarkers
Multiplexed immunofluorescence with C14orf166 antibodies allows simultaneous detection of multiple markers and provides valuable information about protein co-localization and cell type-specific expression:
Antibody selection and validation:
Optimization steps:
Determine optimal dilution for each antibody (typically 1:50-1:500 for IF)
Test different fixation methods (4% paraformaldehyde recommended)
Optimize blocking solutions to minimize background (5% BSA or normal serum)
Sequence antibody applications based on sensitivity
Multiplexing with markers of interest:
Subcellular markers: Combine with nuclear (DAPI), cytoplasmic (β-tubulin), or centrosomal markers to study C14orf166 localization
Pathway components: Co-stain with JAK2/STAT3 pathway members to study functional interactions
Cell-type markers: Combine with epithelial, stromal, or immune cell markers in tumor samples
Signal detection and controls:
Special considerations:
C14orf166 shuttles between nucleus and cytoplasm; therefore, fixation timing may affect localization patterns
In tumors, expression levels vary significantly across cells, requiring careful analysis of heterogeneity
C14orf166 functions in RNA transcription, splicing, and transport, making it relevant for studies of gene expression regulation:
RNA immunoprecipitation (RIP):
Chromatin immunoprecipitation (ChIP):
Use C14orf166 antibodies to precipitate chromatin fragments
Analyze associated DNA sequences to identify genomic binding sites
Connect to transcriptional regulation of specific genes
Proximity ligation assay (PLA):
Detect interaction between C14orf166 and known partners (e.g., RNA polymerase II, 7SK snRNA methylphosphate capping enzyme)
Visualize interaction events at single-molecule resolution in intact cells
Quantify changes in interaction under different conditions
Mass spectrometry analysis of protein complexes:
Immunoprecipitate C14orf166 and associated proteins
Identify components of C14orf166-containing complexes
Study composition changes during cell cycle or in disease states
Functional validation:
Combine with C14orf166 knockdown/knockout approaches
Assess effects on global RNA synthesis using techniques like EU incorporation
Analyze splicing patterns using exon junction microarrays or RNA-seq
Given the significant associations between C14orf166 overexpression and poor cancer prognosis, several experimental approaches can help understand its mechanistic role:
Analysis of JAK2/STAT3 pathway activation:
C14orf166 interacts with JAK2 as a JH2-interacting protein
Use phospho-specific antibodies to detect STAT3 activation (pSTAT3)
Compare pSTAT3 levels in cells with normal vs. overexpressed vs. depleted C14orf166
In esophageal carcinoma, C14orf166 has been shown to activate the JAK2/STAT3 signaling pathway, potentially initiating carcinogenesis
Cell cycle analysis:
Invasion and migration assays:
Given its association with metastasis, test effects of C14orf166 modulation on:
Transwell migration and invasion
Wound healing
3D spheroid invasion models
In vivo metastasis models:
Generate stable cell lines with C14orf166 overexpression or knockdown
Assess metastatic potential in animal models
Correlate with lymph node involvement observations in clinical samples
Multi-omics integration:
Combine proteomics, transcriptomics, and phosphoproteomics
Identify downstream effectors and signaling networks
Connect to pathways involved in tumorigenesis and metastasis
Developing domain-specific antibodies for C14orf166 requires careful consideration of protein structure, function, and experimental goals:
Domain structure and functional mapping:
C14orf166 has distinct functional domains including:
RNA-binding regions
Nuclear localization signals
JAK2-interacting domains
Regions involved in centrosome architecture regulation
Epitope selection strategy:
Antibody development approaches:
Validation requirements:
Confirm epitope-specific binding using deletion mutants
Verify accessibility of the epitope in native protein conformations
Test in multiple applications (WB, IP, IHC, IF)
Evaluate cross-reactivity with related proteins
Functional blocking potential:
Design antibodies that can inhibit specific interactions:
C14orf166-JAK2 interaction
RNA binding
Nuclear-cytoplasmic shuttling
Test ability to modulate downstream signaling (e.g., STAT3 activation)
Recent advances in computational biology offer powerful tools for antibody design:
Biophysics-informed modeling for antibody specificity:
Epitope prediction algorithms:
Computational tools can identify accessible, immunogenic regions within C14orf166
Structures can be predicted using AlphaFold or similar tools
Molecular dynamics simulations can reveal conformational flexibility
Sequence conservation analysis:
Optimization of antibody properties:
Computational design of complementarity-determining regions (CDRs)
Prediction of physicochemical properties (solubility, stability)
Optimization of binding kinetics and affinity
Integration with experimental validation:
Design-build-test cycles combining computational prediction with experimental validation
Machine learning models improve with additional data from experimental testing
This hybrid approach can efficiently generate antibodies with desired specificities and affinities
Researchers may encounter several challenges when working with C14orf166 antibodies:
Inconsistent detection in Western blotting:
Problem: Variable band intensity or multiple bands
Solution: Optimize lysis conditions to ensure complete protein extraction; fresh samples typically yield better results; use phosphatase inhibitors if phosphorylation affects detection
Background in immunohistochemistry:
Subcellular localization variability:
Problem: Inconsistent nuclear vs. cytoplasmic staining
Solution: C14orf166 shuttles between nucleus and cytoplasm; fixation timing may affect localization patterns; compare multiple fixation methods; examine functional state of cells (proliferation, stress)
Species cross-reactivity issues:
Problem: Unexpected results in non-human samples
Solution: Verify antibody's predicted reactivity (human antibodies show varying homology with other species); validate antibody in the specific species being studied
Batch-to-batch variability:
Problem: Different results with new antibody lots
Solution: Request certificate of analysis; run side-by-side comparisons with previous lots; maintain consistent positive controls across experiments
For reliable quantitative assessment of C14orf166 in clinical samples:
Immunohistochemistry scoring systems:
Implement standardized scoring based on:
Staining intensity (0 = negative, 1 = weak, 2 = moderate, 3 = strong)
Percentage of positive cells (0-100%)
Calculate H-score (intensity × percentage) or use cutoff values
In published studies, high C14orf166 expression was defined based on:
Digital image analysis:
Use software for automated quantification
Segment nuclei and cytoplasm for compartment-specific analysis
Generate continuous data rather than categorical scores
Standardize image acquisition parameters
Western blot densitometry:
Normalize C14orf166 band intensity to loading controls (α-Tubulin, GAPDH)
Include gradient standards for quantification
Use technical and biological replicates
qRT-PCR for mRNA quantification:
Statistical analysis recommendations:
Several innovative applications of C14orf166 antibodies are emerging in cancer research:
Liquid biopsy development:
Detection of C14orf166 in circulating tumor cells (CTCs)
Analysis in exosomes as potential non-invasive biomarker
Correlation with tissue expression patterns and clinical outcomes
Therapeutic targeting approaches:
Antibody-drug conjugates (ADCs) targeting C14orf166-overexpressing cells
Blocking antibodies that inhibit C14orf166-JAK2 interaction
CAR-T cell development using anti-C14orf166 single-chain variable fragments (scFvs)
Multi-parameter tissue analysis:
Multiplexed immunofluorescence combining C14orf166 with tumor microenvironment markers
Spatial transcriptomics integrated with C14orf166 protein detection
Single-cell analysis of heterogeneous C14orf166 expression patterns
Predictive biomarker development:
Functional antibody applications:
Intrabodies targeting specific compartments (nuclear vs. cytoplasmic C14orf166)
Antibody-based protein degradation approaches
Monitoring therapy-induced changes in C14orf166 expression/localization
C14orf166's fundamental role in RNA metabolism offers unique therapeutic opportunities:
Targeting RNA-dependent processes:
C14orf166 is involved in spliceosome function and tRNA-splicing
Potential to disrupt aberrant RNA processing in cancer cells
Small molecule inhibitors of C14orf166-RNA interactions
Disruption of protein complexes:
Modulation of gene expression programs:
C14orf166 affects RNAP II activity
Potential to restore normal transcriptional regulation
Combination with epigenetic modifiers to normalize gene expression
Exploitation of synthetic lethality:
Identify genes/pathways that become essential in C14orf166-overexpressing cells
Develop targeted therapies based on these dependencies
Screen for synthetic lethal interactions using CRISPR-Cas9 libraries
Integration with existing targeted therapies: