CELF6 belongs to the CELF/BRUNOL protein family, characterized by RNA recognition motifs (RRMs) that mediate interactions with RNA molecules. Key findings from recent studies include:
Brain Function: CELF6 binds synaptic protein mRNAs and represses translation via UGU-rich motifs, influencing neuronal gene expression .
Cancer Biology:
In lung cancer, CELF6 overexpression inhibits tumor growth by downregulating immune/inflammation-related genes (e.g., TNFSF10, CCL5) and modulating splicing of apoptosis-related genes (e.g., TP53) .
In colorectal cancer (CRC), CELF6 is downregulated and associated with poor prognosis. Its overexpression reduces CRC cell proliferation and stemness by stabilizing HOXA5 mRNA .
Antibodies targeting CELF6 are primarily used in research to study its expression, localization, and functional interactions. Common applications include:
The development and use of CELF6 antibodies have advanced understanding of its roles in:
Neurological Diseases: CELF6’s repression of synaptic mRNAs suggests its potential involvement in neurodegenerative disorders .
Cancer Diagnostics: CELF6 downregulation in CRC tissues (qRT-PCR, IHC) highlights its utility as a prognostic biomarker .
Therapeutic Targets: Antibodies could aid in validating CELF6 as a target for RNA-based therapies or small-molecule inhibitors.
CELF6 (CUGBP Elav-Like Family Member 6) is an RNA-binding protein that belongs to the cytosine-uridine-guanine-binding protein (CUG-BP), Elav-like family (CELF). It plays critical roles in post-transcriptional regulation of pre-mRNAs through binding to specific RNA sequences . The significance of CELF6 in research stems from its multifunctional roles in:
Regulating mRNA stability and processing in various cellular contexts
Cell cycle regulation through modulation of p21 gene expression and mRNA stability
Cancer development, with evidence suggesting tumor suppressor functions in colorectal and lung cancers
Neurobiological processes, particularly in serotonergic neurons, with implications for neuropsychiatric disorders including autism
Understanding CELF6 biology has become increasingly important due to its downregulation in several cancer types and its association with poor clinical outcomes, positioning it as both a potential biomarker and therapeutic target .
CELF6 antibodies have been validated for multiple research applications, each requiring specific protocols and considerations:
| Application | Validated Methods | Key Considerations |
|---|---|---|
| Western Blot | - 10% SDS-PAGE - 5% non-fat milk in TBST (blocking) - 1:1,000 dilution (primary antibody) - 1:10,000 HRP-conjugated secondary antibody - ECL detection | - Effectively detects CELF6 protein expression - Can monitor protein level changes in response to experimental manipulations |
| Immunofluorescence | - 4% paraformaldehyde fixation - Specific peptide-derived antibodies required | - Visualizes subcellular localization - Can reveal expression patterns in different cell types |
| Immunohistochemistry | - Paraffin-embedded tissue analysis - Specific optimization required | - Allows examination of CELF6 expression in tissue contexts - Enables correlation with pathological features |
Research has demonstrated that antibodies generated against the QPGSDTLYNNGVSPC peptide sequence are effective for both Western blot and immunofluorescence applications, while antibodies against AASEGRGEDRKC peptide are only effective for Western blot, highlighting the importance of selecting appropriate antibodies for specific applications .
Thorough validation of CELF6 antibodies is essential for generating reliable data. A comprehensive validation protocol should include:
Specificity testing:
Application-specific validation:
Experimental controls:
Researchers should be aware that not all CELF6 antibodies work equally well across different applications, necessitating application-specific validation .
Optimizing Western blot protocols for CELF6 detection requires attention to several technical parameters:
Sample preparation:
Use complete lysis buffers containing protease inhibitors
Standard RIPA or NP-40 buffers are generally effective for CELF6 extraction
Consider phosphatase inhibitors if studying post-translational modifications
Gel electrophoresis and transfer:
Antibody incubation:
Detection:
When comparing CELF6 expression between samples, normalization to housekeeping proteins such as GAPDH is essential for accurate quantification .
CELF6 functions as an RNA-binding protein that modulates mRNA stability. To investigate this function, researchers should consider these methodological approaches:
RNA immunoprecipitation (RIP) assays:
mRNA stability assays:
Binding site identification:
In vitro binding assays with recombinant CELF6 and target RNA sequences
Mutational analysis of putative binding sites
Cross-linking immunoprecipitation (CLIP) methods to identify binding sites with high resolution
Functional validation:
CELF6 overexpression or knockdown experiments
Assessment of target mRNA and protein levels
Analysis of downstream cellular phenotypes
Studies have demonstrated that CELF6 binds to the 3′UTR of p21 transcript, increasing its mRNA stability, which contributes to cell cycle regulation and potential tumor suppressor activity .
Selecting appropriate experimental models is crucial for CELF6 research. Current evidence supports the following models:
Cell line models:
Animal models:
Patient-derived samples:
Tissue microarrays comparing tumor vs. normal tissues for CELF6 expression
Cancer patient cohorts for correlation with clinical outcomes
Genetic models:
CRISPR/Cas9-mediated CELF6 knockout or mutation
Inducible overexpression systems for temporal control of CELF6 expression
Each model offers distinct advantages depending on the research question. For cancer-related studies, both in vitro cell culture and in vivo xenograft models have provided valuable insights into CELF6's tumor suppressor functions .
CELF6 has emerged as a potential tumor suppressor in colorectal cancer (CRC). Researchers can employ CELF6 antibodies in several sophisticated approaches:
Expression analysis in clinical samples:
Immunohistochemistry of tissue microarrays to correlate CELF6 expression with clinical stages and outcomes
Western blot analysis comparing normal colonic mucosa versus CRC tissues at various stages
Mechanistic investigations:
Functional analysis:
Immunofluorescence to track changes in CELF6 localization during cell cycle progression
Western blot analysis to monitor CELF6 expression after drug treatments or genetic manipulations
Cancer stem cell studies:
Analysis of CELF6 expression in cancer stem cell populations using flow cytometry and immunofluorescence
Investigation of CELF6's role in regulating stemness-related genes
Research has demonstrated that CELF6 overexpression decreases CRC cell proliferation and stemness in vitro, reduces tumor growth in vivo, and induces G1 phase cell cycle arrest . These findings position CELF6 as a promising target for therapeutic development in CRC.
CELF6 has been identified as a cell cycle-regulated protein that controls cancer cell proliferation through p21 regulation. Researchers should consider these methodological approaches:
Cell cycle synchronization and analysis:
Synchronize cells at different cell cycle phases
Analyze CELF6 protein levels by Western blot across cell cycle stages
Immunofluorescence to visualize CELF6 localization changes during cell cycle progression
Degradation pathway analysis:
Downstream effect analysis:
Flow cytometry for cell cycle distribution after CELF6 overexpression or knockdown
BrdU incorporation assays to measure proliferation
Colony formation assays to assess long-term proliferative capacity
p21-focused experiments:
Luciferase reporter assays with p21 3'UTR constructs
mRNA stability assays comparing p21 mRNA half-life in CELF6-modulated cells
Rescue experiments with p21 knockdown in CELF6-overexpressing cells
Studies have shown that CELF6 overexpression induces G1 phase arrest, while its depletion promotes cell cycle progression, proliferation, and colony formation . This effect appears to be p53 and/or p21 dependent, highlighting the importance of these pathways in CELF6's antiproliferative functions.
Integrating transcriptomic approaches with CELF6 protein analysis provides comprehensive insights into its biological functions:
RNA-seq after CELF6 modulation:
Pathway analysis of CELF6-regulated genes:
Alternative splicing analysis:
Examine exon usage and alternative splicing events after CELF6 modulation
Validate splicing changes using RT-PCR with isoform-specific primers
Correlate splicing changes with functional outcomes
Integrative multi-omics approaches:
Combine transcriptomic data with ChIP-seq, CLIP-seq, and proteomics
Construct regulatory networks to understand CELF6's global impact
Identify direct versus indirect regulatory effects
This integrated approach revealed that CELF6 overexpression in lung cancer cells affects genes involved in TNF signaling pathway and cytokine-cytokine receptor interaction, suggesting broader roles in cancer immunity and progression .
CELF6 has been implicated in autism spectrum disorders (ASDs) through genetic and functional studies. CELF6 antibodies have facilitated several key discoveries:
Neural expression mapping:
CELF6 antibodies enabled characterization of expression patterns in neuronal populations
Identified enrichment in serotonergic neurons, which are implicated in repetitive behaviors and resistance to change that characterize autism
Allowed correlation of CELF6 expression with specific neural circuits
Genetic variant analysis:
Immunoblotting was used to validate CELF6 expression in samples with identified genetic variants
Analysis of common variants near CELF6 in the Autism Genetic Resource Exchange (AGRE) collection implicated CELF6 in autism risk
Screening for rare variants using allele-specific PCR allowed further genetic characterization
Animal model development and validation:
Translational profiling:
These studies highlight CELF6's potential role in neurodevelopmental processes relevant to autism and establish it as a valuable target for further investigation in neuropsychiatric research.
Investigating CELF6 in neuronal contexts requires specialized experimental approaches:
Cell type-specific analysis:
Use of bacTRAP (translating ribosome affinity purification) to isolate cell type-specific mRNAs
Single-cell RNA sequencing combined with CELF6 immunostaining
Patch-seq methods to correlate CELF6 expression with electrophysiological properties
Neural circuit mapping:
Immunohistochemistry with CELF6 antibodies combined with neuronal subtype markers
Triple labeling approaches to identify specific neuronal populations expressing CELF6
Brain region-specific analysis focusing on circuits relevant to behaviors affected in autism
Functional studies:
Celf6 knockout or knockdown in primary neuronal cultures
Analysis of dendritic morphology, synapse formation, and neuronal activity
Electrophysiological assessment of neurons with altered CELF6 expression
Behavioral analysis:
Comprehensive behavioral testing of Celf6-/- mice
Focus on behaviors relevant to autism spectrum disorders
Correlation of behavioral phenotypes with molecular and cellular changes
These approaches have successfully identified CELF6's expression in serotonergic neurons and implicated it in autism risk , providing a foundation for deeper investigation of its neurobiological functions.
Distinguishing CELF6 from other CELF family members is crucial for accurate experimental outcomes:
Antibody selection strategies:
Use antibodies raised against peptides specifically selected for "relative uniqueness across the Celf family"
The peptide sequence QPGSDTLYNNGVSPC has been validated for specificity in both Western blot and immunofluorescence applications
Always validate commercial antibodies using appropriate controls
Expression pattern analysis:
Compare expression patterns of different CELF family members using specific antibodies
RNA-seq or RT-qPCR analysis with isoform-specific primers
Consider tissue and developmental stage-specific expression differences
Functional discrimination:
Design rescue experiments with CELF6 versus other CELF proteins
Analyze binding specificities and RNA targets
Evaluate functional outcomes of overexpressing different family members
Knockout validation:
Use Celf6-/- samples as negative controls for antibody specificity
Ensure that antibodies don't cross-react with other CELF proteins in knockout samples
Consider compensatory changes in other CELF family members after CELF6 manipulation
Researchers successfully generated CELF6-specific antibodies by selecting peptides based on cross-species conservation, uniqueness across the CELF family, and hydrophobicity, providing a model for specific antibody development .
Researchers frequently encounter technical issues when working with CELF6 antibodies. Here are solutions to common problems:
Low signal intensity in Western blots:
Increase protein loading (30-50 μg recommended)
Optimize antibody concentration (try 1:500 instead of 1:1000)
Extend primary antibody incubation (overnight at 4°C)
Use more sensitive detection methods (e.g., chemiluminescent substrates with extended signal duration)
Consider signal amplification systems
High background in immunofluorescence:
Implement more stringent blocking (5% BSA instead of non-fat milk)
Increase washing steps (5-6 washes of 10 minutes each)
Dilute primary antibody further after titration experiments
Pre-absorb antibody with acetone powder from tissues not expressing CELF6
Note that only antibodies against specific peptides (e.g., QPGSDTLYNNGVSPC) have been validated for immunofluorescence
Multiple bands in Western blot:
Determine if bands represent isoforms, post-translational modifications, or degradation products
Use Celf6-/- samples as negative controls
Perform peptide competition assays to identify specific versus non-specific bands
Consider using gradient gels for better resolution
Inconsistent results between experiments:
Standardize lysate preparation procedures
Establish consistent blocking, antibody incubation, and washing protocols
Create detailed standard operating procedures for each application
Use internal controls consistently across experiments
Research has shown that not all CELF6 antibodies work equally well across different applications, highlighting the importance of application-specific validation and optimization .
Immunoprecipitation (IP) of CELF6 requires careful optimization:
Lysis buffer selection:
For RNA-binding proteins like CELF6, use non-denaturing lysis buffers
Consider buffers containing low concentrations of NP-40 or Triton X-100
Include RNase inhibitors if studying RNA-protein complexes
Add protease and phosphatase inhibitors to preserve native protein interactions
Antibody selection and coupling:
Use antibodies validated for IP applications
Consider direct coupling to beads (using crosslinkers like BS3 or DMP) to avoid IgG contamination
For challenging IPs, compare different antibodies targeting distinct CELF6 epitopes
Optimization strategies:
Adjust antibody-to-lysate ratio
Test different incubation times and temperatures
Compare various washing stringencies
Pre-clear lysates thoroughly before adding CELF6 antibody
Controls and validation:
Include isotype control antibodies
Use CELF6 knockout or knockdown samples as negative controls
Verify IP efficiency by Western blotting input, unbound, and eluted fractions
Confirm specificity of co-immunoprecipitated proteins by reciprocal IP
Specialized applications:
For RNA immunoprecipitation (RIP), include crosslinking steps
For chromatin immunoprecipitation (ChIP), optimize crosslinking and sonication conditions
For co-IP studies, consider mild detergent conditions to preserve protein-protein interactions
These optimized IP protocols enable investigation of CELF6's interactions with proteins and RNAs, such as its binding to the 3'UTR of p21 mRNA .
Emerging technologies and approaches promise to enhance CELF6 antibody applications:
Advanced imaging techniques:
Super-resolution microscopy for detailed subcellular localization studies
Multiplexed imaging to simultaneously visualize CELF6 with multiple markers
Live-cell imaging with nanobody-based probes for dynamic studies
FRET-based approaches to study CELF6 interactions in living cells
Single-cell applications:
Integration with single-cell RNA sequencing
Mass cytometry (CyTOF) incorporating CELF6 antibodies
Spatial transcriptomics combined with CELF6 immunostaining
High-throughput screening applications:
CELF6 antibody-based readouts for drug screening
CRISPR screens with CELF6 antibody detection
Automated imaging platforms for phenotypic analysis
Therapeutic development:
Use of CELF6 antibodies for target validation in drug discovery
Development of antibody-drug conjugates if CELF6 shows cell-surface expression in specific contexts
Companion diagnostic development for stratifying patients
Clinical applications:
Development of standardized immunohistochemistry protocols for diagnostic use
Creation of tissue microarrays for large-scale analysis of CELF6 in cancer progression
Correlation of CELF6 expression with treatment response
These future directions will expand our understanding of CELF6's biological functions and potential clinical relevance, particularly in cancer biology and neuropsychiatric disorders where CELF6 has already shown significant promise as a research target .