FGFR1OP is located on chromosome 6q27 and encodes a largely hydrophilic centrosomal protein . The gene product contains functional domains including a leucine-rich region at the N-terminus that becomes part of fusion proteins in chromosomal translocations . When studying FGFR1OP, researchers should consider both the wild-type protein structure and the chimeric variants that arise from gene fusion events. The most common fusion involves the N-terminal leucine-rich region of FGFR1OP joined to the catalytic domain of FGFR1, creating an oncogenic protein with constitutive tyrosine kinase activity.
FGFR1OP expression can be evaluated through multiple complementary techniques:
Northern blot analysis: Can be performed using 32P-labeled PCR products of FGFR1OP as probes, with primers such as 5′-TAATAGTACCAGCCATCGCTCAG-3′ and 5′-ATCCTACGGCTTTATTGACACCT-3′ .
Immunohistochemistry: Requires specific antibodies such as rabbit polyclonal anti-human FGFR1OP antibodies. Tissue microarrays are particularly effective for analyzing multiple samples simultaneously .
Western blot analysis: Can be performed using antibodies raised against histidine-tagged human FGFR1OP protein (codons 7–173; accession No. NM_007045) .
For reliable quantification, it's advisable to compare expression levels across multiple normal tissues and cancer samples, as was done in studies examining 23 normal tissues and 372 non-small cell lung cancer specimens .
FGFR1OP functions primarily as a centrosomal protein required for anchoring microtubules to subcellular structures . Its role in cellular physiology includes:
Cell cycle regulation: FGFR1OP shows cell cycle-dependent localization, which can be studied by synchronizing cells at the G1–S boundary using aphidicolin block (1 μg/mL for 24 hours) .
Microtubule organization: As a centrosomal protein, it participates in maintaining cellular architecture and facilitating cellular division.
Protein-protein interactions: FGFR1OP interacts with proteins such as ABL1 and WRNIP1, which are involved in DNA replication and cell cycle progression .
To study these functions, researchers typically use confocal microscopy at wavelengths of 488 and 594 nm to visualize the protein's cellular localization throughout the cell cycle .
FGFR1OP contributes to lung carcinogenesis through multiple mechanisms:
Cell growth promotion: Overexpression of FGFR1OP increases cellular growth, while its suppression using siRNA inhibits growth of lung cancer cells .
Enhanced cellular motility: Induction of FGFR1OP increases cell motility, potentially contributing to metastatic capacity .
Modulation of ABL1-dependent signaling: FGFR1OP significantly reduces ABL1-dependent phosphorylation of WRNIP1, resulting in promotion of cell cycle progression .
Experimental approaches to investigate these mechanisms include:
RNA interference assays using vector-based RNAi systems such as psiH1BX3.0
Cell proliferation assays following FGFR1OP knockdown or overexpression
Cell cycle analysis using flow cytometry after synchronization with aphidicolin
Co-immunoprecipitation studies to identify and validate protein interaction partners
Immunohistochemical analysis has revealed positive FGFR1OP staining in 334 (89.8%) of 372 NSCLC specimens, with high expression levels significantly associated with shorter tumor-specific survival times (P < 0.0001) .
FGFR1OP has significant prognostic implications for lung cancer patients:
Survival correlation: High levels of FGFR1OP expression are significantly associated with shorter tumor-specific survival times (P < 0.0001 by log-rank test) .
Independent prognostic factor: Multivariate Cox analysis has determined that FGFR1OP is an independent prognostic factor for surgically treated NSCLC patients (P < 0.0001) .
Methodologically, researchers should:
Utilize tissue microarrays for high-throughput analysis
Apply multivariate Cox analysis on backward (stepwise) procedures
Include variables that satisfy an entry level of P < 0.05
Establish exit criteria of P < 0.05 for independent factors
Perform Kaplan-Meier survival analysis with log-rank tests for statistical validation
When designing prognostic studies, it's crucial to include adequate sample sizes and appropriate clinical follow-up periods to ensure robust statistical power for survival analyses.
FGFR1OP participates in oncogenic fusion events in hematological malignancies:
FGFR1OP-FGFR1 fusion: Resulting from t(6;8)(q27;p11) chromosomal translocation, found in myeloproliferative disorders .
FGFR1OP-RET fusion: A novel fusion identified in chronic myelomonocytic leukemia (CMML) and primary myelofibrosis (PMF) with secondary acute myeloid leukemia (AML) .
The FGFR1OP-RET fusion protein demonstrates:
Constitutive tyrosine kinase activity
Transforming potential in NIH3T3 fibroblasts
Ability to induce IL3-independent growth in hematopoietic Ba/F3 cells
Activation of PI3K/STAT signaling pathways
Support of cytokine-independent growth and enhanced self-renewal of hematopoietic progenitor cells
In vivo studies have shown that FGFR1OP-RET causes a spectrum of disease phenotypes, with >50% of mice developing fatal myeloproliferative disorder (MPD) . Other observed phenotypes include transplantable leukemia, expansion of the mast cell lineage, and reduced repopulating activity following lethal irradiation .
Several experimental approaches have proven effective for investigating FGFR1OP functions:
RNA interference:
Protein interaction studies:
Cell cycle analysis:
In vivo models:
Clinical correlation:
Based on current research, several therapeutic approaches could target FGFR1OP:
RNA interference strategies:
Inhibition of downstream signaling pathways:
Targeting FGFR1OP fusion proteins:
For FGFR1OP-RET fusions, RET tyrosine kinase inhibitors might prove effective
For FGFR1OP-FGFR1 fusions, FGFR inhibitors could potentially block constitutive signaling
When designing therapeutic targeting studies, researchers should consider:
Cell line models with varying FGFR1OP expression levels
Patient-derived xenografts to better recapitulate the tumor microenvironment
Combination approaches targeting multiple nodes in FGFR1OP-related pathways
Biomarker development to identify patients most likely to respond to FGFR1OP-targeted therapies
For optimal immunohistochemical detection of FGFR1OP:
Antibody preparation:
Sample preparation:
Staining protocol:
Evaluation criteria:
Assess staining intensity (negative, weak, moderate, strong)
Determine percentage of positive cells
Consider both cytoplasmic and nuclear staining patterns
Validation of antibody specificity should be performed using Western blot analysis with lysates from NSCLC tissues and cell lines as well as normal lung tissues .
To study FGFR1OP protein interactions and functional consequences:
Identification of interaction partners:
Yeast two-hybrid screening
Co-immunoprecipitation followed by mass spectrometry
Protein array approaches
Validation of interactions:
Reciprocal co-immunoprecipitation experiments
GST pull-down assays
Proximity ligation assays for in situ detection
Functional analysis:
Phosphorylation status assessment using phospho-specific antibodies
Domain mapping through truncation or deletion constructs
Site-directed mutagenesis to identify critical interaction residues
Cellular consequences:
Cell cycle analysis following disruption of specific interactions
Assessment of localization changes during cell cycle progression
Evaluation of effects on cell growth, motility, and survival
For studying FGFR1OP's interaction with ABL1 and WRNIP1 specifically, researchers should examine how FGFR1OP influences ABL1-dependent phosphorylation of WRNIP1 and the subsequent effects on cell cycle progression .
While direct evidence linking FGFR1OP to drug resistance is limited in the provided search results, several avenues warrant investigation:
Cell survival pathways: Given FGFR1OP's role in promoting cell growth and its interaction with signaling proteins like ABL1, it may contribute to resistance by activating survival pathways.
Cell cycle regulation: FGFR1OP's influence on cell cycle progression through WRNIP1 could potentially affect the efficacy of cell cycle-targeting therapies.
Fusion proteins and resistance: FGFR1OP-containing fusion proteins (like FGFR1OP-RET) activate multiple signaling pathways including PI3K/STAT , which are known to contribute to therapy resistance in various cancers.
Methodological approaches to study FGFR1OP in drug resistance:
Generate resistant cell lines through chronic drug exposure
Compare FGFR1OP expression and function between sensitive and resistant populations
Assess whether FGFR1OP knockdown resensitizes resistant cells to therapy
Investigate combinations of FGFR1OP targeting with standard therapies
While the search results primarily focus on FGFR1OP in lung cancer and hematological malignancies, researchers should consider:
Multi-cancer analysis:
Comprehensive immunohistochemical analysis across tumor tissue microarrays representing diverse cancer types
Mining of cancer genomics databases (TCGA, ICGC) for FGFR1OP expression patterns
Correlation with clinical outcomes across different malignancies
Cancer subtype specificity:
Comparison between NSCLC subtypes (adenocarcinoma vs. squamous cell carcinoma)
Analysis in different molecular subtypes of lung adenocarcinoma (EGFR-mutant, ALK-rearranged, KRAS-mutant, etc.)
Assessment in various subtypes of hematological malignancies beyond CMML and PMF/AML
Correlation with other biomarkers:
Integration with established prognostic and predictive biomarkers
Potential for creating composite biomarker panels including FGFR1OP
Current data shows that FGFR1OP is overexpressed in 89.8% (334/372) of NSCLC specimens , suggesting its widespread relevance in lung cancer. Similar comprehensive analyses across other tumor types would be valuable for expanding the therapeutic potential of FGFR1OP targeting.
The Fibroblast Growth Factor Receptor 1 (FGFR1) is a member of the receptor tyrosine kinase (RTK) family, which plays a crucial role in various cellular processes, including proliferation, differentiation, and survival. FGFR1 is involved in the signaling pathways that regulate embryonic development, tissue repair, and angiogenesis .
The term “FGFR1 Oncogene Partner” refers to the various genes that can fuse with FGFR1 due to chromosomal translocations. These fusion events lead to the creation of chimeric proteins that possess oncogenic properties. One well-known example is the FGFR1-TACC1 fusion, which has been identified in glioblastoma and squamous cell carcinoma (SqCC) .
The fusion proteins resulting from FGFR1 translocations often lead to the constitutive activation of FGFR1 signaling pathways. This activation occurs independently of ligand binding and results in uncontrolled cellular proliferation and inhibition of apoptosis . The FGFR1-TACC1 fusion, for instance, promotes hyperactivation of FGFR1, contributing to tumorigenesis .
Targeting FGFR1 and its fusion proteins has become a promising approach in cancer therapy. Several FGFR inhibitors have been developed and are currently undergoing clinical trials. These inhibitors aim to block the aberrant signaling pathways activated by FGFR1 fusions, thereby inhibiting tumor growth and progression .