CCDC170 antibody (e.g., ab97814 from Abcam) is a rabbit polyclonal antibody developed against a recombinant fragment of human CCDC170 (amino acids 400 to C-terminus) . Key features include:
Applications: Validated for Western blot (WB) and unspecified applications
Immunogen: Recombinant fragment within residues 400–C-terminus of human CCDC170
CCDC170 plays critical roles in:
Golgi-microtubule stabilization: CCDC170 localizes to the Golgi apparatus, organizing perinuclear microtubules essential for cell polarity and migration . Dysregulation is linked to breast cancer progression .
Apoptosis regulation: Higher CCDC170 expression correlates with better prognosis in breast cancer patients by promoting apoptosis via the IRE1α pathway .
Endocrine resistance: Truncated CCDC170 variants (e.g., ESR1-CCDC170 fusions) drive tamoxifen resistance by activating HER2/HER3/SRC signaling .
Osteogenesis: CCDC170 promotes bone formation by upregulating Runx2, Osterix, and Wnt signaling .
Osteoclast inhibition: Knockdown reduces osteogenic markers (e.g., ALP, Col1a1) and increases osteoclast activity (e.g., TRACP, CTSK) .
Parameter | Detail | Source |
---|---|---|
Specificity | Detects endogenous CCDC170 at ~82 kDa in WB | |
Species reactivity | Human (validated); predicted homology in other species | |
Storage | Stable at −20°C in glycerol-based buffer |
Biomarker potential: CCDC170 expression stratifies luminal breast cancer subtypes and predicts chemotherapy/radiation response .
Therapeutic targeting: ESR1-CCDC170 fusion cells show sensitivity to HER2/SRC inhibitors (e.g., lapatinib, dasatinib) combined with tamoxifen .
CCDC170 is a protein that has gained considerable attention due to its involvement in breast cancer pathogenesis. Its significance stems from the discovery of recurrent rearrangements between the estrogen receptor gene (ESR1) and CCDC170, which have been identified in 6-8% of luminal B breast cancers . These ESR1-CCDC170 gene fusions are associated with more aggressive forms of estrogen receptor-positive (ER+) breast cancer and potentially reduced endocrine therapy responsiveness . Additionally, CCDC170 affects breast cancer apoptosis through modulation of the IRE1 pathway, suggesting its importance in cancer cell survival mechanisms .
CCDC170 expression shows significant variation across breast cancer molecular subtypes. Analysis of data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases reveals that CCDC170 mRNA levels are higher in luminal A and luminal B subtypes, while they are considerably lower in HER2-positive and basal-like breast cancer subtypes . This subtype-specific expression pattern suggests that CCDC170 may play different roles depending on the molecular context and could potentially serve as a biomarker for certain breast cancer subtypes.
The subcellular localization of CCDC170 has been investigated using electron microscopy in HeLa cells transiently expressing GFP-CCDC170. Researchers have used immunogold labeling with anti-GFP antibodies to track the protein's location within cellular compartments . Understanding the subcellular distribution pattern is crucial for interpreting immunofluorescence results and provides insights into potential protein functions. The protein's localization pattern suggests possible associations with specific cellular structures, which may inform hypotheses about its biological role.
CCDC170 antibodies have been validated for several research applications:
Western blotting: For detecting both wild-type CCDC170 and truncated variants resulting from ESR1-CCDC170 gene fusions
Immunohistochemistry (IHC): For analyzing CCDC170 expression in patient tissue samples and xenograft tumors
Immunofluorescence: For examining subcellular localization and co-localization with other proteins
Immunoprecipitation: For protein interaction studies and complex isolation
Proximity labeling experiments: When combined with techniques like BioID for identifying protein interaction networks
Each application requires specific optimization and validation strategies to ensure reliable results.
ESR1-CCDC170 fusion proteins (ΔCCDC170) can be detected using antibodies targeting the C-terminal region of CCDC170, as this portion is retained in the fusion products. Western blot analysis using commercial polyclonal antibodies against the C-terminus of CCDC170 has successfully detected various fusion variants with predicted molecular weights of 41kDa (E2-E6), 39kDa (E2-E7), 30kDa (E2-E8), and 14kDa (E2-E10) in cells ectopically expressing these variants . When designing experiments to detect fusion proteins, researchers should:
Select antibodies that recognize epitopes present in the truncated fusion products
Include appropriate positive controls (cells expressing known fusion variants)
Use negative controls (fusion-negative cell lines)
Optimize protein extraction methods, particularly if the proteins associate with cellular structures
Comprehensive validation of CCDC170 antibodies is crucial for generating reliable research data. Key validation strategies include:
When investigating ESR1-CCDC170 fusion variants, researchers should consider this comprehensive experimental approach:
Detection and characterization:
RT-PCR with primers spanning the fusion junction
Western blotting with C-terminal CCDC170 antibodies to identify fusion proteins
DNA sequencing to confirm fusion breakpoints
Functional analysis:
In vivo studies:
Mechanistic investigations:
Researchers have successfully employed multiple strategies to manipulate CCDC170 expression:
CRISPR/Cas9-mediated knockout:
Overexpression systems:
RNA interference:
Fusion protein expression:
To investigate the relationship between CCDC170 and the IRE1 pathway, researchers should consider these experimental approaches:
Expression correlation analysis:
Protein expression manipulation:
Functional assays:
Assessing the impact of CCDC170 manipulation on cell survival under ER stress conditions
Measuring XBP1 splicing as a readout of IRE1 activity
Evaluating apoptosis rates in relation to CCDC170 and IRE1 expression
Mechanistic studies:
Investigating direct or indirect interactions between CCDC170 and IRE1
Examining effects on downstream IRE1 signaling components
Exploring potential regulatory mechanisms
When encountering conflicting CCDC170 expression data across studies, researchers should consider several factors:
Methodological differences:
Antibody epitopes and specificity
Detection techniques (Western blot, IHC, qPCR)
Sample preparation and fixation methods
Biological variables:
Quantification approaches:
Threshold definitions for "high" versus "low" expression
Normalization methods
Statistical analyses and significance thresholds
Validation strategies:
Correlation between protein and mRNA data
Use of multiple antibodies targeting different epitopes
Inclusion of appropriate positive and negative controls
ESR1-CCDC170 fusions appear to significantly impact endocrine therapy response in breast cancer:
Clinical observations:
Experimental evidence:
T47D xenograft tumors expressing ESR1-CCDC170 fusion variants (E2-E7, E2-E10) show significantly reduced sensitivity to tamoxifen treatment
Kaplan-Meier analysis reveals significantly worse regression-free survival for both E2-E7 (P < 0.01) and E2-E10 (P < 0.001) fusion-expressing tumors treated with tamoxifen
The E2-E10 variant confers a more pronounced growth advantage (P = 0.000002) compared to the E2-E7 variant
Potential mechanisms:
Therapeutic implications:
The prognostic significance of CCDC170 expression shows some data source-dependent variation:
This complex picture suggests that CCDC170's impact on patient outcomes is context-dependent and warrants careful interpretation in relation to other clinicopathological factors.
Distinguishing between wild-type CCDC170 and fusion variants in patient samples requires a multi-modal approach:
Nucleic acid-based detection:
Protein-based detection:
Tissue analysis:
Validation approaches:
Based on current research, several therapeutic strategies could potentially target CCDC170 or its fusion variants:
Targeting downstream signaling pathways:
Fusion-specific approaches:
Development of small molecules targeting the unique protein interfaces created by fusion events
Peptide-based inhibitors designed to disrupt fusion protein interactions
Gene therapy approaches:
siRNA or antisense oligonucleotides targeting fusion-specific junctions
CRISPR/Cas9-based strategies to disrupt fusion genes
Biomarker-guided therapy:
Using ESR1-CCDC170 status to guide treatment decisions
Monitoring fusion status during treatment to detect resistance mechanisms
Combination strategies:
Integrating endocrine therapy with targeted agents based on fusion status
Personalizing treatment regimens according to molecular profiles
While these approaches show promise in experimental settings, clinical translation requires further validation in appropriate models and eventually in clinical trials.