FAM110C antibody enabled critical discoveries in GBM research:
Prognostic Significance:
Elevated FAM110C expression correlates with poor survival in wild-type IDH1 GBM patients:
Functional Validation:
Knockdown experiments in GBM cell lines (Ln229, U87):
Epigenetic Regulation Findings:
Methylation status analysis in PDAC:
| Lesion Type | Methylation Frequency |
|---|---|
| IPMN | 41.18% (14/34) |
| MCN | 46.67% (7/15) |
| PDAC | 72.89% (207/284) |
Methylated FAM110C associated with:
Drug Development Applications:
Identified through CMap analysis:
| Compound | Mean Connectivity Score | Mechanism |
|---|---|---|
| Felbinac | -0.554 | COX-2 pathway modulation |
| Fludrocortisone | -0.561 | Glucocorticoid receptor activation |
Dose Response Data:
| Cell Line | Felbinac IC₅₀ (μM) | Fludrocortisone IC₅₀ (μM) |
|---|---|---|
| Ln229 | 4.48 | 0.31 |
| U87 | 17.37 | 16.26 |
Standard Protocols:
Western Blot: Primary antibody dilution 1:1000
Immunohistochemistry: Antigen retrieval with citrate buffer (pH 6.0)
Cell Migration Assay: 24-well transwell system, 12h incubation
FAM110C (Family with Sequence Similarity 110 Member C) is a protein-coding gene that has been implicated in several critical cellular processes. Recent studies have identified FAM110C as a potential biomarker in various cancers, particularly glioblastoma (GBM) and pancreatic ductal adenocarcinoma (PDAC). In wild-type GBM, FAM110C has been shown to contribute to cell growth and migration . Interestingly, the expression levels of FAM110C differ significantly between IDH1 mutant and wild-type GBM, with higher expression observed in the wild-type variant . In contrast, in pancreatic cancer, FAM110C has been identified as a potential tumor suppressor that can activate ATM and NHEJ signaling pathways through its interaction with HMGB1 . This dual role highlights the context-dependent function of FAM110C across different cancer types.
FAM110C antibodies are primarily utilized in several key research applications:
Western blot (WB) analysis to detect and quantify FAM110C protein expression in cell lysates and tissue samples
Immunohistochemistry (IHC) to visualize FAM110C expression patterns in tissue sections
Immunoprecipitation (IP) to isolate FAM110C and study its protein-protein interactions
Immunofluorescence (IF) to determine the subcellular localization of FAM110C
These applications are critical for investigating FAM110C's role in disease progression, particularly in cancer research where expression levels may correlate with prognosis and treatment response .
Proper validation of FAM110C antibodies is essential for obtaining reliable experimental results. Recommended validation approaches include:
Positive and negative controls: Use cell lines with known FAM110C expression levels. Based on published data, MIAPaCa-2 and JF-305 cells show minimal expression, while Panc3.11, Panc5.04, and Panc10.05 cells demonstrate higher expression levels .
siRNA/shRNA knockdown: Compare FAM110C detection in cells with and without gene silencing.
Overexpression validation: Test the antibody in cells transfected with FAM110C expression vectors, similar to the approach described in the literature using pCDH-CMV-MCS-puro plasmid .
Multiple antibody comparison: Use antibodies that recognize different epitopes of FAM110C to confirm specificity.
Peptide competition assays: Pre-incubate the antibody with the immunizing peptide to confirm signal specificity.
These validation steps are crucial for ensuring that experimental observations truly reflect FAM110C biology rather than antibody cross-reactivity.
Expression patterns of FAM110C vary significantly across different cell types and disease states:
This expression pattern data is valuable for selecting appropriate positive and negative controls when validating antibodies and designing experiments .
FAM110C expression and its epigenetic regulation show significant correlations with cancer progression and patient outcomes:
In GBM:
FAM110C is highly expressed in wild-type GBM compared to IDH1-mutant GBM (P = 0.0053)
Higher FAM110C expression correlates with poorer prognosis in wild-type GBM patients (P = 0.03, HR = 1.55 [95% CI 1.04, 2.31])
FAM110C expression can predict survival in wild-type GBM patients at 1, 3, and 5 years (ROC-1 year: 0.647; ROC-3 years: 0.709; ROC-5 years: 0.932)
In pancreatic lesions:
These findings highlight the potential value of FAM110C as a diagnostic and prognostic biomarker in multiple cancer types .
FAM110C interacts with several important signaling pathways that influence cancer cell behavior:
In GBM:
Gene Set Enrichment Analysis (GSEA) revealed that FAM110C expression is associated with:
These pathways align with the observed role of FAM110C in promoting glioma cell migration and invasion
In PDAC:
FAM110C activates ATM and Non-Homologous End Joining (NHEJ) signaling pathways by interacting with HMGB1
Loss of FAM110C sensitizes PDAC cells to DNA damage response inhibitors:
These differential pathway interactions may explain the context-dependent roles of FAM110C in different cancer types and suggest potential therapeutic strategies targeting these pathways .
To investigate FAM110C methylation status and its functional consequences, researchers should consider the following methodological approaches:
Methylation status assessment:
Functional consequences investigation:
Re-expression experiments in methylated cell lines using expression vectors
RNA interference to silence FAM110C in unmethylated cell lines
Phenotypic assays (proliferation, migration, apoptosis) to assess functional impact
Cell cycle analysis using flow cytometry following synchronization by serum withdrawal
Correlation with clinical parameters:
This multi-layered approach provides comprehensive insights into how FAM110C methylation affects cellular function and patient outcomes .
Research has uncovered a synthetic lethal relationship between FAM110C expression and sensitivity to DNA damage response (DDR) inhibitors:
Loss of FAM110C expression sensitizes PDAC cells to:
The mechanistic basis for this synthetic lethality involves:
FAM110C's role in activating ATM and NHEJ signaling pathways through HMGB1 interaction
When FAM110C is silenced (by methylation or other mechanisms), cells become more dependent on ATR/CHK1 pathways for DNA damage repair
Inhibiting these compensatory pathways with targeted inhibitors leads to synthetic lethality
This relationship suggests a potential therapeutic strategy:
Identify patients with FAM110C methylation/silencing
Target these tumors with ATR/CHK1 inhibitors
Exploit the synthetic lethal interaction for improved therapeutic efficacy
For experimental validation, researchers can use half-inhibitory concentration analysis with MTT assays after treating cells with gradient dilutions of inhibitors .
For optimal detection of FAM110C via Western blot, researchers should consider the following protocol recommendations:
Sample preparation:
Extract total protein from cells using RIPA buffer with protease inhibitors
Determine protein concentration using BCA or Bradford assay
Load 20-40 μg of protein per lane for cell lysates
Gel electrophoresis and transfer:
Use 10-12% SDS-PAGE gels (FAM110C is approximately 33 kDa)
Transfer to PVDF membrane at 100V for 90 minutes in cold transfer buffer
Antibody selection and dilution:
Controls and validation:
Detection:
Use enhanced chemiluminescence (ECL) detection reagents
Optimize exposure time to avoid signal saturation
Following these guidelines will help ensure specific and reliable detection of FAM110C protein in experimental samples .
To effectively investigate FAM110C's role in cell migration, researchers should design comprehensive experiments based on established protocols:
Experimental models:
Manipulation of FAM110C expression:
Migration assays:
Transwell migration assay: Assess directional cell migration through membrane
Wound healing assay: Evaluate collective cell migration by creating a "scratch"
Time-lapse microscopy: Monitor cell movement in real-time
Analysis and quantification:
For transwell assays: Count migrated cells in multiple fields
For wound healing: Measure wound closure over time
Statistical analysis: Compare migration between FAM110C-manipulated and control cells
Pathway analysis:
This experimental approach will provide comprehensive insights into FAM110C's functional role in cell migration .
To investigate FAM110C's protein interactions, researchers should employ multiple complementary approaches:
Co-immunoprecipitation (Co-IP):
Proximity ligation assay (PLA):
Visualize protein-protein interactions in situ
Provides spatial information about where interactions occur within cells
Use specific antibodies against FAM110C and potential interacting partners
Bimolecular fluorescence complementation (BiFC):
Express FAM110C and candidate interactors as fusion proteins with split fluorescent protein fragments
Interaction brings fragments together, restoring fluorescence
Enables live-cell visualization of interactions
Yeast two-hybrid screening:
Systematic approach to identify novel interacting partners
Use FAM110C as bait to screen a library of prey proteins
Validate hits with orthogonal methods
Bioinformatic analysis:
These approaches provide complementary data on FAM110C's interactome, offering insights into its molecular functions and mechanism of action .
Researchers can utilize FAM110C expression or methylation status for patient stratification in clinical studies using the following approaches:
Several promising therapeutic strategies targeting FAM110C-related pathways warrant further investigation:
Synthetic lethality approaches:
Methylation-based therapies:
In cancers where FAM110C acts as a tumor suppressor (PDAC), demethylating agents could restore expression
Testing combinations of demethylating agents with conventional chemotherapy
Small molecule inhibitors:
The Connectivity Map (CMap) analysis identified felibinac and fludrocortisone as potential targeted drugs for FAM110C in wild-type GBM
These compounds showed mean connective scores of −0.554 and −0.561, respectively (P = 0.001)
Further validation and optimization of these compounds could yield effective therapies
Pathway-based approaches:
Biomarker-guided therapy selection:
These approaches represent promising avenues for translating FAM110C research into clinical applications .
Several technological advances would significantly enhance FAM110C research and accelerate discoveries:
Improved antibody development:
Generation of monoclonal antibodies with higher specificity for different FAM110C isoforms
Development of phospho-specific antibodies to detect post-translational modifications
Creation of antibodies suitable for chromatin immunoprecipitation (ChIP) applications
Advanced imaging techniques:
Super-resolution microscopy to visualize FAM110C localization at nanoscale resolution
Live-cell imaging systems to track FAM110C dynamics during cell cycle and migration
Multiplexed imaging to simultaneously visualize FAM110C and its interaction partners
Single-cell analysis technologies:
Single-cell RNA-seq to characterize heterogeneity in FAM110C expression within tumors
Single-cell proteomics to quantify FAM110C protein levels at cellular resolution
Spatial transcriptomics to map FAM110C expression patterns within tissue architecture
CRISPR-based functional genomics:
CRISPR activation/inhibition systems for precise modulation of FAM110C expression
CRISPR screens to identify synthetic lethal interactions with FAM110C
Base editing to introduce or correct specific mutations in FAM110C
Computational approaches:
Machine learning algorithms to predict FAM110C interactions and functional effects
Integrative multi-omics analysis to place FAM110C in broader cellular networks
Structural biology predictions to model FAM110C protein structure and interactions
These technological advances would address current limitations in FAM110C research and facilitate more comprehensive understanding of its biological functions and clinical applications.