This conjugate is validated for:
Western blotting (WB): Detects FGF19 at ~25 kDa in human liver and intestinal lysates .
Immunohistochemistry (IHC): Localizes FGF19 in formalin-fixed paraffin-embedded tissues .
Immunofluorescence (IF): Visualizes intracellular FGF19 distribution using fluorescence microscopy .
Flow cytometry (FCM): Quantifies FGF19 expression in cell populations .
Application | Recommended Dilution | Compatible Species |
---|---|---|
WB | 1:300 – 1:5,000 | Human, Rat |
IHC | 1:50 – 1:200 | Human |
IF | 1:100 – 1:500 | Human, Mouse |
FGF19 suppresses hepatic CYP7A1 expression, reducing bile acid synthesis . In adipocytes, it enhances glucose uptake via FGFR4/KLB receptor complexes . Studies show FGF19 inhibits insulin-induced fatty acid synthesis by 30–38% in hepatocytes through SREBP-1c downregulation .
NAFLD/NASH: Reduces hepatic steatosis by inhibiting ACCα and FAS expression .
Cholestasis: Modulates bile acid enterohepatic circulation .
Specificity: Validated using siRNA-mediated FGF19 knockdown in HepG2 cells .
Cross-reactivity: 100% homology with human, 92% with chimpanzee, no reactivity with mouse .
Lot consistency: ≥95% purity by SDS-PAGE, endotoxin levels <1 EU/µg .
FGF19 (Fibroblast Growth Factor 19) serves as a physiological regulator of bile acid homeostasis in humans. It functions primarily by suppressing bile acid biosynthesis through down-regulation of CYP7A1 expression, following positive regulation of the JNK and ERK1/2 signaling cascades. Additionally, FGF19 stimulates glucose uptake in adipocytes and has emerged as a potent insulin sensitizer capable of normalizing plasma glucose concentration, improving lipid profiles, and ameliorating fatty liver disease. The activity of FGF19 requires the presence of two co-receptors: KLB (βklotho) and FGFR4 (Fibroblast Growth Factor Receptor 4) .
To investigate FGF19's dual functions in metabolism and cell proliferation, a comprehensive experimental design should include:
Metabolic assessment:
Measure glucose uptake in adipocytes or hepatocytes using glucose uptake assays
Analyze bile acid synthesis by measuring expression of CYP7A1 using RT-PCR
Perform glucose tolerance tests in animal models
Cell proliferation analysis:
BrdU incorporation assays to measure DNA synthesis rates
Cell cycle analysis by flow cytometry
Assessment of EGFR and Wnt signaling pathway activation
When using FITC-conjugated FGF19 antibodies, these can be employed to track FGF19 localization during these processes using confocal microscopy. Additionally, comparing wild-type FGF19 with engineered variants that show reduced mitogenic potential (such as FGF19 ΔKLB) can help differentiate between metabolic and proliferative signaling pathways .
For comprehensive multi-parameter analysis using FITC-conjugated FGF19 antibodies, researchers can implement the following combinations:
Technique | Compatible Fluorophores | Parameters Measured | Application |
---|---|---|---|
Multi-color Flow Cytometry | PE, APC, PerCP-Cy5.5 | FGF19 expression, receptor binding, cell cycle | Quantitative analysis of cell populations |
Confocal Microscopy | DAPI, TRITC, Cy5 | FGF19 localization, co-localization with FGFR4/KLB | Subcellular distribution studies |
FRET Analysis | CFP (donor) | Protein-protein interactions | FGF19-receptor binding dynamics |
Single-cell RNA-seq with protein detection | Antibody-oligo conjugates | Transcriptome + FGF19 protein | Correlation of gene expression with protein levels |
When designing multi-parameter experiments, ensure spectral compatibility between fluorophores to minimize bleed-through, and include appropriate compensation controls when using flow cytometry .
For optimal flow cytometry results with FITC-conjugated FGF19 antibodies, follow this protocol:
Sample preparation:
Harvest cells (1-5 × 10^6 cells/sample)
Wash twice in cold PBS containing 2% FBS
Fix cells with 2% paraformaldehyde for 15 minutes at room temperature (if intracellular staining is required)
Permeabilize with 0.1% saponin in PBS for 10 minutes (for intracellular targets only)
Staining procedure:
Block with 2% normal serum from the same species as secondary antibody for 30 minutes
Incubate with FITC-conjugated FGF19 antibody at 1-5 μg/mL for 30-45 minutes at 4°C in the dark
Wash three times with PBS containing 2% FBS
Resuspend in PBS with 2% FBS for immediate analysis
Controls:
Include an isotype control conjugated to FITC
Use FGF19-transfected cells as a positive control
Use non-transfected cells as a negative control
Instrument settings:
To determine the optimal antibody concentration for immunofluorescence with FITC-conjugated FGF19 antibodies, perform a titration experiment following these guidelines:
Titration series:
Prepare a serial dilution of the antibody (typically 0.1-10 μg/mL)
Test each concentration on identical samples with known FGF19 expression
Evaluation criteria:
Signal-to-noise ratio (quantify signal intensity vs. background)
Specificity (confirm pattern matches expected subcellular localization)
Reproducibility across replicate samples
Optimization table:
Antibody Concentration (μg/mL) | Signal Intensity | Background | Signal-to-Noise Ratio | Notes |
---|---|---|---|---|
0.1 | Low | Minimal | Low | Insufficient signal |
0.5 | Moderate | Minimal | Good | Potential working dilution |
1.0 | Strong | Low | Excellent | Optimal for most applications |
2.5 | Very Strong | Moderate | Good | May be suitable for low-expressing samples |
5.0 | Very Strong | High | Reduced | Excessive concentration |
10.0 | Very Strong | Very High | Poor | Not recommended |
Validation:
To preserve the fluorescence activity and binding capacity of FITC-conjugated FGF19 antibodies, adhere to these storage guidelines:
Short-term storage (up to 1 month):
Store at 4°C in the dark
Add sodium azide (0.02-0.05%) as a preservative
Protect from light using amber vials or by wrapping in aluminum foil
Long-term storage:
Store at -20°C in small aliquots to avoid repeated freeze-thaw cycles
Add a cryoprotectant such as glycerol (final concentration 30-50%)
Include a protein stabilizer such as BSA (0.1-1%)
Stability considerations:
FITC is pH-sensitive; maintain storage buffer at pH 7.2-7.4
FITC conjugates are photosensitive; minimize exposure to light
Avoid repeated freeze-thaw cycles (limit to <5) which can cause protein aggregation and fluorophore degradation
Quality control:
Distinguishing specific FITC-conjugated FGF19 antibody binding from autofluorescence requires systematic controls and analytical approaches:
Essential controls:
FITC-conjugated isotype control antibody (same species, isotype, and fluorophore:protein ratio)
Unstained samples to establish baseline autofluorescence
FGF19-negative samples (knockout or knockdown) as negative controls
Pre-absorption with recombinant FGF19 protein to confirm specificity
Analytical approaches:
Spectral unmixing to separate FITC signal from autofluorescence
Multi-parameter analysis using antibodies against known FGF19 interacting partners (FGFR4, KLB)
Comparison of fluorescence patterns between fixed and unfixed samples
Technical considerations:
Autofluorescence typically has broader emission spectra than FITC
Cellular autofluorescence often correlates with cell size/granularity
Tissue autofluorescence is often associated with specific structures (lipofuscin, elastin)
Advanced solutions:
When encountering unexpected bands in Western blots using FGF19 antibodies, implement this systematic troubleshooting approach:
Potential causes and solutions:
Issue | Possible Causes | Solutions |
---|---|---|
Higher MW bands (>24 kDa) | Post-translational modifications | Treat with deglycosylation enzymes |
Protein aggregation | Add stronger reducing agents; adjust sample preparation | |
Protein complexes | Use stronger denaturing conditions | |
Lower MW bands (<24 kDa) | Protein degradation | Add protease inhibitors; reduce sample processing time |
Alternative splice variants | Verify with RT-PCR for splice variants | |
Antibody cross-reactivity | Pre-absorb antibody with recombinant FGF19 | |
Multiple bands | Non-specific binding | Optimize blocking conditions; increase wash stringency |
Cross-reactivity with other FGF family members | Validate with FGF19-specific peptide competition |
Validation strategies:
Compare results from multiple anti-FGF19 antibodies targeting different epitopes
Test antibody against recombinant FGF19 protein as positive control
Include FGF19-transfected cell lysate as reference (expected 24 kDa band)
Compare with known FGF19-negative samples
Technical optimization:
When analyzing FGF19 signaling pathway activation from experimental data, follow these systematic approaches:
Key signaling nodes to assess:
FGFR4 and KLB receptor complex formation
JNK and ERK1/2 phosphorylation status (early signaling events)
CYP7A1 expression levels (metabolic endpoint)
EGFR and Wnt/β-catenin pathway activation (proliferative endpoints)
Integrated data analysis framework:
Signaling Component | Metabolic Pathway Activation | Proliferative Pathway Activation | Analytical Method |
---|---|---|---|
FGFR4-KLB dimerization | Required | Required | Proximity ligation assay |
ERK1/2 phosphorylation | Transient, low threshold | Sustained, high threshold | Western blot, phospho-flow cytometry |
CYP7A1 expression | Significant downregulation | Minimal effect | RT-PCR, RNA-seq |
EGFR/TGFα signaling | Minimal activation | Strong activation | Phospho-protein analysis |
Wnt/β-catenin pathway | Minimal activation | Strong activation | TCF reporter assays, β-catenin localization |
Differential activation thresholds:
Metabolic effects require weaker FGFR dimerization and transient signaling
Proliferative effects require stronger FGFR dimerization and sustained signaling
Gene expression changes in BA biosynthesis genes vs. cancer-related genes can distinguish pathway bias
Comparison with engineered FGF19 variants:
Wild-type FGF19 activates both metabolic and proliferative pathways
FGF19 ΔFGFR shows reduced proliferative but maintained metabolic signaling
FGF19 ΔHBS shows further reduced proliferative with maintained metabolic signaling
FGF19 ΔKLB shows minimal proliferative with fully preserved metabolic signaling
FITC-conjugated FGF19 antibodies provide powerful tools for investigating the complex formation between FGF19, FGFR, KLB, and heparan sulfate (HS) through several advanced methodological approaches:
Live-cell imaging techniques:
Use FITC-conjugated FGF19 antibodies to track FGF19 localization in real-time
Combine with differently labeled antibodies against FGFR4 and KLB for co-localization studies
Implement FRAP (Fluorescence Recovery After Photobleaching) to assess complex dynamics and stability
Super-resolution microscopy approaches:
STORM or PALM imaging using photoconvertible fluorophores for nanoscale resolution
Structured illumination microscopy (SIM) for enhanced spatial resolution of complex components
Combination with proximity ligation assays for validation of protein-protein interactions
Quantitative complex analysis:
Implement automated image analysis algorithms to quantify co-localization coefficients
Utilize FLIM-FRET (Fluorescence Lifetime Imaging-Förster Resonance Energy Transfer) to measure molecular distances between complex components
Correlate with functional readouts such as ERK1/2 phosphorylation using multi-parameter imaging
Methodological considerations:
Developing non-mitogenic FGF19 variants while preserving beneficial metabolic effects involves several sophisticated experimental approaches:
Structure-guided engineering strategies:
Targeted mutagenesis of residues involved in FGFR, HS, or KLB binding
Examples include FGF19 ΔFGFR (Y115A mutation), FGF19 ΔHBS (K149A mutation), and FGF19 ΔKLB (D198A mutation)
Engineering chimeric proteins combining regions from FGF19 and FGF21
Functional validation pipeline:
Experimental Approach | Purpose | Key Readouts |
---|---|---|
Surface Plasmon Resonance | Measure binding affinities to FGFR, HS, KLB | Binding constants (KD) |
Proximity Ligation Assay | Assess FGFR dimerization potential in situ | Dimerization signal intensity |
Cell Proliferation Assays | Evaluate mitogenic potential | BrdU incorporation, Ki67 expression |
Glucose Tolerance Tests | Assess metabolic function | Blood glucose levels |
RNA-sequencing | Compare gene expression profiles | Differential regulation of metabolic vs. proliferative genes |
Threshold-based design principles:
Exploit differential signaling thresholds for metabolic vs. proliferative pathways
Target receptor dimerization strength to maintain weak (metabolic) signaling while eliminating strong (proliferative) signaling
Focus on modifications that affect the stability and duration of receptor complexes
Advanced animal model testing:
FITC-conjugated FGF19 antibodies provide valuable tools for dissecting the distinct signaling pathways activated by different FGF19 variants:
Cellular localization and trafficking studies:
Track the subcellular localization of FGF19 variants in real-time
Compare receptor complex formation patterns between variants using multi-color imaging
Assess endocytosis and trafficking rates of different FGF19-receptor complexes
Signaling dynamics assessment:
Correlate FGF19 binding with downstream signaling pathway activation
Measure the duration and intensity of ERK1/2 phosphorylation following stimulation
Implement live-cell reporters for real-time monitoring of signaling pathways
Multi-parameter approach for pathway discrimination:
Parameter | Metabolic Signaling | Proliferative Signaling | Experimental Approach |
---|---|---|---|
Signal duration | Transient | Sustained | Time-course immunofluorescence |
Receptor complex stability | Lower | Higher | FRAP analysis |
Downstream pathway activation | JNK/ERK (transient) | EGFR/Wnt/β-catenin | Multiplexed immunostaining |
Gene expression changes | CYP7A1, CYP8B1 downregulation | EGFR, Axin2, TCF7 upregulation | Single-cell RNA-seq with protein detection |
Advanced methodological considerations:
Rigorous validation of FITC-conjugated FGF19 antibodies requires a comprehensive set of controls:
Positive controls:
FGF19-transfected cell lysates (showing expected 24 kDa band in Western blot)
Recombinant human FGF19 protein
Tissues/cells known to express FGF19 (e.g., ileum, liver)
Negative controls:
Non-transfected cell lysates
FGF19 knockout or knockdown samples
Tissues/cells known not to express FGF19
Specificity controls:
Pre-absorption with recombinant FGF19 protein
Competing peptide controls
Cross-reactivity testing with related FGF family members (FGF21, FGF23)
Technical controls:
FITC-conjugated isotype control antibody
Secondary antibody-only controls (for indirect methods)
Autofluorescence controls (untreated samples)
Validation matrix:
Validation Method | Purpose | Expected Result |
---|---|---|
Western blot | Confirm size specificity | Single band at 24 kDa |
Immunoprecipitation | Verify target capture | Enrichment of 24 kDa protein |
Peptide competition | Confirm epitope specificity | Signal reduction with specific peptide |
Immunofluorescence pattern | Assess subcellular localization | Consistent with known biology |
Multiple antibody comparison | Confirm target recognition | Concordant results with antibodies to different epitopes |
These controls collectively ensure that the observed signals represent genuine FGF19 detection rather than technical artifacts or cross-reactivity .
The choice of fixation and permeabilization methods significantly impacts epitope preservation and accessibility for FGF19 antibody detection:
Fixation methods comparison:
Fixation Method | Advantages | Disadvantages | Impact on FGF19 Detection |
---|---|---|---|
Paraformaldehyde (4%) | Good morphology preservation | May mask some epitopes | Generally suitable for most FGF19 epitopes |
Methanol (-20°C) | Better penetration, no cross-linking | Poor membrane preservation | May better expose some intracellular epitopes |
Glutaraldehyde | Excellent ultrastructure preservation | Significant autofluorescence | Generally not recommended for FITC detection |
Glyoxal | Low autofluorescence | Limited literature | May preserve some conformational epitopes |
Acetone | Good for some nuclear antigens | Poor morphology | Variable results with FGF19 |
Permeabilization method effects:
Permeabilization Agent | Mechanism | Effect on FGF19 Epitopes | Recommended Use |
---|---|---|---|
Triton X-100 (0.1-0.5%) | Dissolves lipids | May disrupt membrane-associated complexes | Intracellular FGF19 detection |
Saponin (0.1%) | Creates pores in membranes | Preserves membrane structures | FGF19-receptor complex studies |
Digitonin (10-50 μg/mL) | Selective cholesterol extraction | Minimal disruption of protein complexes | Receptor-bound FGF19 studies |
Tween-20 (0.2%) | Mild detergent | Gentle permeabilization | General purpose |
No permeabilization | N/A | Access only to extracellular epitopes | Secreted/surface-bound FGF19 |
Optimization strategies:
Several quantitative approaches can be employed with FITC-conjugated FGF19 antibodies for accurate measurement in biological samples:
Flow cytometry-based quantification:
Quantitative flow cytometry using calibration beads
Measurement of molecules of equivalent soluble fluorochrome (MESF)
Single-cell analysis of FGF19 expression levels across populations
Microscopy-based quantification:
Integrated fluorescence intensity measurement
Automated high-content imaging with quantitative analysis
Ratio imaging using internal reference standards
Calibration and standardization approaches:
Method | Principle | Advantages | Considerations |
---|---|---|---|
Standard curve | Serial dilutions of recombinant FGF19 | Direct quantification | Requires identical staining conditions |
Flow cytometry beads | Beads with defined FITC molecules | Absolute quantification | May not account for antibody affinity variations |
Competitive binding assay | Competition with known quantities | High sensitivity | Complex setup |
Reference standards | Known positive controls | Inter-assay normalization | Requires stable reference samples |
Data analysis considerations:
Background subtraction using appropriate negative controls
Correction for autofluorescence contribution
Standardization across different instruments and experiments
Statistical validation using technical and biological replicates
Assay validation parameters:
Analytical sensitivity: lowest detectable concentration
Analytical specificity: lack of interference from related proteins
Precision: intra- and inter-assay coefficients of variation
Linearity: proportional response across concentration range
Recovery: accurate measurement in complex biological matrices
FGF19 antibodies are integral to advancing research on FGF19's therapeutic potential for metabolic diseases through several cutting-edge approaches:
Therapeutic target validation:
Tracking FGF19 distribution and receptor binding in metabolic tissues
Correlating FGF19 levels with disease severity in patient samples
Monitoring changes in FGF19 signaling during disease progression
Development of FGF19-based therapeutics:
Screening engineered FGF19 variants for optimal metabolic/minimal proliferative activities
Comparing wild-type FGF19 with variants like FGF19 ΔKLB that maintain beneficial metabolic effects
Validating efficacy of FGF19 analogs in normalizing blood glucose and regulating bile acid synthesis
Mechanism elucidation:
Dissecting the differential signaling thresholds between metabolic and proliferative pathways
Identifying tissue-specific responses to FGF19 signaling
Characterizing the interplay between FGF19 and other metabolic regulators
Clinical translation research:
Developing companion diagnostics to identify optimal responders to FGF19-based therapies
Monitoring on-target and off-target effects during clinical trials
Assessing long-term safety through detection of proliferative marker changes
Potential therapeutic applications:
Metabolic Condition | FGF19 Therapeutic Approach | Key Endpoints | Antibody Application |
---|---|---|---|
Type 2 diabetes | Non-mitogenic FGF19 analogs | Glucose normalization | Monitoring tissue distribution and target engagement |
NAFLD/NASH | FGF19 ΔKLB or similar variants | Liver fat reduction | Assessing liver uptake and signaling activation |
Cholestatic liver disease | Engineered FGF19 with enhanced CYP7A1 regulation | Bile acid normalization | Quantifying pathway-specific effects |
Obesity | FGF19 with targeted adipose tissue delivery | Weight loss | Tracking tissue-specific accumulation |
Research with these antibodies is revealing that fine-tuning of receptor dimerization and downstream signaling thresholds provides a practical approach for engineering safer FGF19 agonists for treating metabolic diseases .
Recent structural biology advances have significantly enhanced our understanding of FGF19's bifunctional nature:
Quaternary complex structure insights:
Detailed 2:2:2:2 FGF19-FGFR1c-KLB-HS complex model reveals coordination between multiple binding partners
Critical residues that mediate FGF19-FGFR interactions identified (e.g., Tyr-115)
Structure of the atypical heparan sulfate (HS) binding site elucidated (involving Lys-149)
C-terminal tail interactions with KLB mapped (with Asp-198 playing a key role)
Structure-function correlation:
Residues critical for FGFR dimerization identified through mutagenesis and functional studies
Differential binding interfaces associated with metabolic vs. proliferative outcomes characterized
Conformational changes induced by receptor binding correlated with downstream signaling bias
Advanced structural techniques employed:
Technique | Information Provided | Key Findings |
---|---|---|
X-ray crystallography | High-resolution static structures | Binding interfaces between FGF19 and receptors |
Cryo-electron microscopy | Complex assemblies at near-atomic resolution | Quaternary complex architecture |
Surface plasmon resonance | Binding kinetics and affinities | Differential binding strengths of FGF19 variants |
Proximity ligation assays | In situ complex formation | Dimerization capacity in cellular context |
Hydrogen-deuterium exchange MS | Conformational dynamics | Flexible regions involved in binding |
Translational outcomes:
Rational design of FGF19 variants with selective pathway activation
Development of the threshold model for FGF signaling specificity
Creation of variants with progressively reduced dimerization capacity (FGF19 ΔFGFR, FGF19 ΔHBS, FGF19 ΔKLB)
Demonstration that metabolic signaling requires weaker receptor dimerization than proliferative signaling
Emerging single-cell technologies combined with FITC-conjugated FGF19 antibodies offer unprecedented insights into cellular heterogeneity in FGF19 responses:
Advanced single-cell methodologies:
Single-cell RNA-seq combined with protein detection (CITE-seq) to correlate FGF19 binding with transcriptional responses
Mass cytometry (CyTOF) with metal-labeled antibodies for high-parameter analysis of FGF19 signaling
Single-cell Western blotting to detect FGF19-induced signaling in individual cells
Spatial transcriptomics to map FGF19 responses within tissue architecture
Key biological questions addressable:
Cell-to-cell variability in FGF19 receptor expression and signaling responses
Identification of distinct cellular subpopulations with differential sensitivity to FGF19
Transition dynamics between metabolic and proliferative states
Spatial organization of FGF19 responsive cells within complex tissues
Technological integration approaches:
Single-cell Technology | Application with FGF19 Antibodies | Research Insights |
---|---|---|
Single-cell RNA-seq + FITC-Ab | Correlate FGF19 binding with gene expression | Transcriptional signatures of responding vs. non-responding cells |
Imaging mass cytometry | Spatial mapping of FGF19 and downstream signaling | Microenvironmental influences on FGF19 signaling |
Live-cell imaging + microfluidics | Real-time tracking of FGF19 responses | Temporal dynamics and cellular decision-making |
Spatial proteomics | Localization of FGF19-receptor complexes | Subcellular organization of signaling machinery |
Translational implications:
When selecting and validating FGF19 antibodies, researchers should implement these comprehensive best practices:
Application-specific selection criteria:
Western blot: Select antibodies validated for denatured epitopes
Immunofluorescence: Choose antibodies validated for preserved cellular morphology
Flow cytometry: Prefer directly conjugated antibodies for single-step detection
Functional studies: Select non-neutralizing antibodies that don't interfere with FGF19 activity
Validation framework:
Multi-technique validation using orthogonal methods
Testing across multiple biological contexts (different cell types/tissues)
Inclusion of appropriate positive and negative controls
Comparison with alternative antibodies targeting different epitopes
Documentation and reporting standards:
Record complete antibody information (catalog number, lot, concentration used)
Document validation experiments in detail
Include all controls in published research
Share validation data with research community
Decision-making matrix for antibody selection:
Research Question | Recommended Antibody Type | Key Validation Tests | Important Considerations |
---|---|---|---|
FGF19 expression levels | Mono/polyclonal against conserved epitope | Western blot, IHC with knockdown controls | Check cross-reactivity with FGF21/FGF23 |
Receptor complex studies | Non-neutralizing antibodies | Proximity ligation assays, co-IP | Verify antibody doesn't disrupt complex formation |
Signaling pathway activation | Phospho-specific for downstream targets | Stimulation time-course experiments | Include pathway inhibitor controls |
Therapeutic variant testing | Epitope-specific antibodies | Binding assays with wild-type and variants | Ensure epitope is preserved in variants |
Following these best practices ensures reliable, reproducible research outcomes and facilitates comparison across different studies investigating FGF19 biology and therapeutic applications .
Researchers investigating FGF19 signaling specificity should incorporate these critical experimental design elements:
Receptor complex considerations:
Account for the quaternary complex (FGF19-FGFR-KLB-HS) in experimental design
Verify expression levels of all components in model systems
Consider tissue-specific variations in receptor/co-receptor distribution
Address the impact of receptor dimerization strength on signaling outcomes
Signaling dynamics approach:
Implement time-course experiments to distinguish transient from sustained signaling
Measure both early (minutes to hours) and late (hours to days) signaling events
Compare dose-response relationships across different pathways
Correlate receptor dimerization strength with pathway activation thresholds
Pathway discrimination strategy:
Pathway | Key Markers | Optimal Detection Method | Temporal Characteristics |
---|---|---|---|
Metabolic signaling | CYP7A1 suppression, JNK/ERK activation | qRT-PCR, phospho-specific Western blot | Rapid onset, transient |
Proliferative signaling | EGFR/TGFα, Wnt/β-catenin activation | RNA-seq, reporter assays | Delayed onset, sustained |
Bile acid metabolism | CYP7A1, CYP8B1, CYP27A1 | Metabolomics, qRT-PCR | Hours to days timeframe |
Glucose metabolism | Glucose uptake, glycolysis | Metabolic flux analysis | Rapid onset (minutes) |
Comparative approach using FGF19 variants:
Include wild-type FGF19 as reference standard
Test engineered variants with defined receptor binding characteristics
Compare variants with selective metabolic vs. proliferative activities
Implement parallel in vitro and in vivo experimental systems
Technical and biological controls:
Include receptor/co-receptor knockdown controls
Implement pathway-specific inhibitors as reference points
Use FGF19 ΔKLB as a metabolic-only positive control
Include FGF19 ΔC-tail as negative control unable to bind KLB