Recombinant Human Fibroblast growth factor 19 (FGF19), partial (Active)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

FGF19 activates FGFR4 in a βKlotho-dependent manner, triggering downstream pathways such as:

  • Metabolic regulation: Suppresses hepatic bile acid synthesis via CYP7A1 inhibition and improves glucose tolerance by reducing gluconeogenesis (G6pc1) .

  • Proliferative effects: Stimulates hepatocyte and cancer cell proliferation through ERK1/2 and PI3K/AKT pathways .

Notably, FGF19’s proliferative activity can be dissociated from its metabolic effects via mutations in the β1-β2 loop or residues 38–42 .

Metabolic Benefits in Chronic Kidney Disease (CKD)

  • Muscle preservation: FGF19-treated CKD mice exhibited 25% larger skeletal muscles and reduced ectopic lipid accumulation in soleus muscle .

  • Glucose homeostasis: Fasting glucose decreased by 11% (p < 0.01), and glucose tolerance improved (AUC: 12,573 vs. 16,693 mg/dl·min⁻¹, p < 0.0001) .

  • Anti-inflammatory effects: Hepatic Il-6, Tnfα, and Mcp1 expression decreased by 40–60% .

Oncogenic Roles in Hepatocellular Carcinoma (HCC)

  • Proliferation: FGF19+/FGFR4+ HCC cell lines (Huh7, JHH7) showed 2–3× faster growth than FGF19−/FGFR4− lines (HepG2, PLC/PRF/5) .

  • Clinical relevance: FGF19+/FGFR4+ tumors correlated with higher AFP levels (p = 0.001) and poor differentiation (p = 0.003) .

Bile Acid Regulation

  • FGF19 represses CYP7A1 via FXR activation, reducing bile acid synthesis by 70% in murine models .

Applications in Research

ApplicationModel SystemOutcomeSource
Organoid culturePorcine hepatic organoidsEnhanced proliferation and differentiation
Metabolic syndrome studiesHFD-fed miceImproved insulin sensitivity and lipid profiles
Cancer therapeutics screeningHuman HCC xenograftsTumor growth inhibition via FGFR4 knockdown

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS solution, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. For optimal stability, store working aliquots at 4°C for up to one week.
Reconstitution
For optimal reconstitution, we recommend briefly centrifuging the vial prior to opening to collect the contents at the bottom. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
FGF 19; FGF-19; FGF15; FGF19; FGF19_HUMAN; Fibroblast growth factor 15; Fibroblast growth factor 19
Datasheet & Coa
Please contact us to get it.
Expression Region
23-216aa
Mol. Weight
21.8 kDa
Protein Length
Partial
Purity
>95% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor 19 (FGF19) is a key regulator of bile acid biosynthesis, glucose metabolism, and cell growth. It suppresses bile acid synthesis by downregulating CYP7A1 expression, following positive regulation of the JNK and ERK1/2 cascades. FGF19 also stimulates glucose uptake in adipocytes. Its activity requires the presence of KLB and FGFR4.
Gene References Into Functions
  1. A study revealed that FGF19 amplification is a recurrent genetic alteration in Chinese lung squamous cell carcinoma (LSCC) patients, observed in 37.5% of cases. FGF19-amplified LSCC cells exhibit elevated FGF19 mRNA expression, and downregulation of FGF19 expression significantly inhibits cell proliferation both in vitro and in vivo. PMID: 28906590
  2. FGFR4/FGF19 autocrine signaling plays a crucial role in the survival of a specific subtype of basal-like breast cancer cells. PMID: 27192118
  3. FGF19 copy number may increase in hepatocellular carcinoma, often coinciding with a complete response to sorafenib treatment. PMID: 27384874
  4. Research indicates that elevated FGF19 expression or hyperactivation of FGF19/FGFR4 signaling in hepatocellular carcinoma cells is a significant contributor to sorafenib resistance. PMID: 28069043
  5. This study represents the first investigation to elucidate the role of FGF19/FGFR4 signaling in the development of hepatocellular carcinoma cells arising from fatty liver. PMID: 27447573
  6. High FGF19 expression is strongly associated with the presence of hepatocellular carcinoma. PMID: 26498355
  7. Findings demonstrate that FGF19 exerts a cytoprotective effect against ER stress by activating a FGFR4-GSK3beta-Nrf2 signaling cascade. This suggests targeting this signaling pathway as a potential therapeutic strategy for managing hepatocellular carcinoma (HCC). PMID: 28951455
  8. Fibroblast growth factor 19 levels in human portal blood are consistently higher than in arterial blood. Under fasted steady state conditions, fibroblast growth factor 19 is released by the portal-drained viscera. PMID: 28003563
  9. Intestinal sensing of significantly elevated levels of conjugated bile acids in blood triggers FGF15/FGF19 signaling, leading to reduced hepatic bile acid synthesis and modulation of bile acid transporters. PMID: 28498614
  10. Serum FGF19 and FGF21 levels, alongside hepatic Klotho expression, exhibit an inverse association with hepatic damage in children with NAFLD. PMID: 23840612
  11. Administering FGF19, or a suitable mimetic, as a pharmacological intervention to increase circulating levels of FGF19 and suppress BA synthesis by inhibiting CYP7A1 gene expression holds promise for therapeutic benefits in many PBC patients. PMID: 28570655
  12. Amplification of FGF19 was validated in independent LSCC samples. Furthermore, FGF19 stimulated LSCC cell growth in vitro. These data identify FGF19 as a potential driver gene in LSCC, particularly in cases associated with smoking. PMID: 26943773
  13. FGF19 significantly enhances the migratory and invasive capabilities of gastric cancer cells. PMID: 27053348
  14. Bile acid and FGF19 levels increased following Roux-en-Y bypass, but not after intensive medical management, in type 2 diabetic subjects who achieved similar improvements in glycemic control. PMID: 26259981
  15. FGF19 levels correlate with the severity of liver disease and may serve as a valuable indicator of chronic cholestatic liver injury. PMID: 26293907
  16. Research indicates that FGF19 can be secreted and promotes ovarian cancer progression, including proliferation and invasion, by activating FGFR4. PMID: 26323668
  17. Significant mechanistic differences appear to exist between humans and mice regarding the nuclear receptors controlling the VitA-FGF15/19 axis. PMID: 26723851
  18. This study suggests a potential link between gallbladder cholangiocyte-derived FGF19 and bile acid metabolism that could contribute to metabolic dysregulation following cholecystectomy. PMID: 26256900
  19. This review provides an overview of the current understanding of the complex biology of endocrine FGFs. PMID: 26567701
  20. The increase in FGF-19 levels after OGL was positively associated with age and negatively associated with abnormal glucose regulation and statin treatment. PMID: 26343925
  21. KL methylation is a characteristic feature of many breast cancer cases. The resulting or associated perturbation in FGFR4 expression, similar to FGF19, could potentially serve as a biomarker for poor prognosis. PMID: 26152288
  22. While the pathogenesis of intestinal failure-associated liver disease is not fully understood, research has investigated the potential roles of FGF19 and pro-inflammatory cytokines in this disease state. PMID: 25595885
  23. This review summarizes the altered expression of FGF19 in non-alcoholic fatty liver disease and hepatocellular carcinoma, highlighting the limited information available on its role in other liver diseases. PMID: 25547779
  24. In mice with humanized livers, expression of an FGF19 transgene effectively corrects bile acid signaling defects, leading to normalization of bile acid synthesis, the bile acid pool, and liver size. PMID: 26028580
  25. Data suggest that circulating levels of FGF19 and FGF21, along with hepatic gene expression of the associated signaling pathway, are significantly dysregulated in type 2 diabetes. PMID: 25664662
  26. This study describes a non-tumorigenic FGF19 variant, M70, which regulates bile acid metabolism and, through inhibition of bile acid synthesis/reduction of excess hepatic bile acid accumulation, protects mice from cholestatic liver injury. PMID: 25080475
  27. Obesity appears to be the primary determinant of abnormalities in FGF21 and FGF19 levels. Contrasting changes in beta-Klotho expression in fat and liver indicate potential tissue-specific alterations in responsiveness to endocrine FGFs in obesity. PMID: 24813368
  28. FGF19 levels were reduced in non-diabetic obese subjects compared to lean controls and obese type 2 diabetic subjects. PMID: 24841294
  29. Fibroblast growth factor 19 may be associated with biochemical recurrence after radical prostatectomy by promoting cell proliferation and epithelial-mesenchymal transition of prostate cancer. PMID: 25854696
  30. In hepatocellular carcinoma, FGF19 amplifications, known to activate Wnt signaling, were mutually exclusive with CTNNB1 and AXIN1 mutations and significantly associated with cirrhosis. PMID: 24798001
  31. FGF19 expression is not associated with lymph node metastasis or locally invasive characteristics of the tumor in colorectal cancers. PMID: 23803094
  32. Reduced fibroblast growth factor 19 is a characteristic feature of bile acid diarrhea. PMID: 23981126
  33. [review] While FGF19 serves as a negative feedback regulator of bile acid metabolism and can circulate as a hormone, emerging evidence has demonstrated its autocrine or exocrine function. PMID: 24827712
  34. FGF19 stimulates tumor progression by activating the STAT3 pathway. PMID: 24728076
  35. Reduced serum FGF19 levels may play a role in the pathophysiology of gestational diabetes mellitus, while increased serum FGF21 levels might represent a compensatory response to this condition. PMID: 24260557
  36. Quantification of FGF19 expression appears to provide valuable prognostic information in breast cancer. PMID: 24248542
  37. Fasting serum FGF19 levels were decreased in Chinese subjects with IFG and inversely associated with fasting glucose levels. PMID: 23628619
  38. These results suggest that SREBP-2 negatively regulates the FXR-mediated transcriptional activation of the FGF19 gene in human intestinal cells. PMID: 24321096
  39. Serum FGF19 is associated with the presence and severity of coronary artery disease in a Chinese population. PMID: 23940810
  40. The specificity of hFGF19 signaling is significantly altered in a mouse model system. PMID: 23064887
  41. FGF19 protein expression may serve as an effective predictor of early recurrence and a marker for poor prognosis in hepatocellular carcinoma. PMID: 23456506
  42. FGF19 (fibroblast growth factor 19) emerges as a novel target gene for activating transcription factor 4 in response to endoplasmic reticulum stress. PMID: 23205607
  43. A decrease in fasting FGF19 levels is associated with the development of non-alcoholic fatty liver disease in obese adolescents. PMID: 23329754
  44. HNF4alpha and LRH-1 promote active transcription histone marks on the Cyp7a1 promoter, which are reversed by FGF19 in a SHP-dependent manner. PMID: 23038264
  45. These results suggest that FGF19 is transcriptionally activated through multiple Farnesoid X receptor-responsive elements in the promoter region. PMID: 22561792
  46. Endocrine FGF19 and FGF21 exhibit differential specificity towards FGFR1 and FGFR4 when complexed with KLB. PMID: 22442730
  47. The effect of FGF19 on APOA was attenuated by transfection of primary hepatocytes with siRNA against the FGF19 receptor 4 (FGFR4). PMID: 22267484
  48. Baseline serum FGF-19 levels are significantly lower in obese patients with type 2 diabetes and are at least partially dependent upon nutritional status, but not related to glucose metabolism or insulin sensitivity parameters. PMID: 21574752
  49. Mouse Fgf15 and human FGF19 play pivotal roles in enterohepatic signaling, regulation of liver bile acid biosynthesis, gallbladder motility, and metabolic homeostasis. PMID: 22396169
  50. FGF-19 levels are reduced in type 2 diabetic patients with metabolic syndrome. PMID: 22166511

Show More

Hide All

Database Links

HGNC: 3675

OMIM: 603891

KEGG: hsa:9965

STRING: 9606.ENSP00000294312

UniGene: Hs.249200

Protein Families
Heparin-binding growth factors family
Subcellular Location
Secreted.
Tissue Specificity
Expressed in fetal brain, cartilage, retina, and adult gall bladder.

Q&A

What is the structural composition of recombinant human FGF19?

Recombinant human FGF19 is typically derived from E. coli expression systems spanning amino acids Leu25-Lys216 of the native protein. The full-length human FGF19 consists of a 251 amino acid precursor with a 22-amino acid signal peptide and a 229-amino acid secreted mature protein without N-linked glycosylation sites . The protein contains critical structural elements including the N-terminal region (residues 38-42) and heparin-binding regions that significantly influence its receptor specificity and biological activity .

How does FGF19 differ from other members of the FGF family?

FGF19 belongs to the unique FGF19 subfamily that functions as endocrine hormones rather than typical paracrine factors. Unlike most FGFs that activate multiple FGF receptors, FGF19 exhibits remarkable specificity for FGFR4 . While many FGF family members require heparin/heparan sulfate for receptor activation, FGF19 can activate receptors in both heparin-dependent and βKlotho-dependent manners. FGF19 shares approximately 61% amino acid identity with chicken FGF-19 and 51% with murine FGF-15, which is considered its functional ortholog in mice .

What are the primary biological activities of FGF19?

FGF19 demonstrates dual biological activities:

ActivityMechanismPhysiological Effect
MetabolicActivation of FGFR1c/βKlothoReduction of serum glucose and insulin levels, improved glucose tolerance
MitogenicActivation of FGFR4Enhanced hepatocyte proliferation

These distinct activities operate through separate structural elements and receptor activation pathways, enabling researchers to potentially separate these functions through targeted mutations .

How should recombinant FGF19 be reconstituted and stored for optimal activity?

For optimal reconstitution of lyophilized FGF19:

  • For carrier-containing FGF19: Reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin .

  • For carrier-free FGF19: Reconstitute at 100 μg/mL in sterile PBS .

Storage recommendations:

  • Store the reconstituted protein at -20°C to -80°C

  • Use a manual defrost freezer

  • Avoid repeated freeze-thaw cycles as they may compromise protein activity

  • Consider preparing single-use aliquots for long-term experiments

Proper reconstitution and storage are critical for maintaining FGF19 functionality in receptor activation assays, cell proliferation studies, and metabolic experiments.

What are the validated biological assays for measuring FGF19 activity?

Several assays can be employed to assess distinct FGF19 activities:

  • Receptor Activation Assays: ERK phosphorylation in L6 cells transfected with specific FGFRs (with or without βKlotho) provides a direct measure of receptor activation. Western blot analysis can quantify phospho-ERK levels as an indicator of downstream signaling .

  • Metabolic Activity Assays:

    • Glucose uptake assays in adipocytes

    • In vivo glucose tolerance tests in diabetic mouse models (e.g., ob/ob)

    • Measurement of serum glucose and insulin levels after FGF19 administration

  • Mitogenic Activity Assays:

    • BrdU incorporation assays in liver sections to measure hepatocyte proliferation

    • Histopathological examination focusing on pericentral hepatocytes

  • Binding Assays:

    • Solid-phase binding assays to measure interaction with FGFRs and co-receptors

    • Surface Plasmon Resonance (SPR) for quantitative binding kinetics

Each assay should include appropriate controls to distinguish FGF19-specific effects from background activity.

What are appropriate in vivo models for studying FGF19 metabolic functions?

When designing in vivo experiments with FGF19, consider these validated models:

  • Diabetic mouse models:

    • ob/ob mice: Effective for studying glucose homeostasis effects

    • High-fat diet-induced diabetic models: Useful for investigating insulin resistance

  • Administration protocols:

    • Injection routes: Intraperitoneal or intravenous administration is typically used

    • Dosing: 1-5 mg/kg body weight is commonly employed in published studies

    • Timing: Acute (single dose) or chronic (daily dosing for 1-3 weeks) depending on the research question

  • Assessment parameters:

    • Fasting blood glucose and insulin levels

    • Glucose tolerance test (GTT)

    • Insulin tolerance test (ITT)

    • Liver enzyme profiles

    • Changes in gene expression related to glucose metabolism

When conducting these studies, researchers should carefully monitor for potential mitogenic effects on hepatocytes, particularly in chronic administration protocols.

How do carrier proteins affect FGF19 activity in different experimental systems?

The presence of carrier proteins like Bovine Serum Albumin (BSA) has significant implications for FGF19 experiments:

  • Stability considerations:

    • BSA enhances protein stability, increases shelf-life, and allows storage at more dilute concentrations

    • Carrier-free preparations may be less stable and require more careful handling

  • Experimental interference:

    • BSA may interfere with certain assay systems, particularly those involving protein quantification, immunodetection, or where background protein could complicate results

    • For proteomic applications or mass spectrometry, carrier-free versions are strongly recommended

  • Receptor binding studies:

    • For precise binding kinetics measurements, carrier-free preparations provide more accurate data

    • For cell-based assays focusing on physiological responses rather than binding, carrier-containing preparations may provide more consistent results

When reporting research findings, the specific preparation used (carrier-free vs. with carrier) should be clearly documented as it may influence experimental outcomes and reproducibility.

What are the critical quality control parameters for verifying FGF19 functionality before experiments?

Before conducting experiments, verify FGF19 quality through these key assessments:

  • Biological activity testing:

    • ERK phosphorylation assay in receptor-transfected cells (e.g., L6 cells with FGFR1c/βKlotho)

    • Minimum activity threshold should be established for each new lot

  • Protein integrity verification:

    • SDS-PAGE to confirm molecular weight (~24 kDa) and purity

    • Western blotting with specific anti-FGF19 antibodies

  • Endotoxin testing:

    • Limulus Amebocyte Lysate (LAL) assay to ensure endotoxin levels are below 1 EU/μg protein

    • Critical for in vivo applications and primary cell culture experiments

  • Aggregation assessment:

    • Dynamic light scattering or size-exclusion chromatography to detect potential protein aggregation

    • Important after reconstitution and during storage

Documenting these quality control measures enhances experimental reproducibility and facilitates accurate interpretation of results.

How can researchers distinguish between FGF19 and FGF21 effects in experimental systems?

Differentiating the effects of these related proteins requires specific experimental approaches:

  • Receptor specificity exploitation:

    • FGF19 activates FGFR4 strongly, while FGF21 does not

    • Experiments in FGFR4-knockout systems can help distinguish FGF19-specific effects

    • Compare responses in cells with different FGFR expression profiles

  • Structural variant utilization:

    • Use chimeric proteins like those described in research (e.g., FGF19-4, FGF19-5) that specifically lack FGFR4 activation capacity but retain metabolic functions

    • These can help separate metabolic from proliferative effects

  • Downstream signaling analysis:

    • Monitor receptor-specific signaling pathways

    • Quantify differential gene expression profiles induced by each protein

  • Combinatorial approaches:

    • Use specific receptor antagonists alongside FGF19/FGF21 treatment

    • Employ RNA interference to selectively knock down specific receptors

A comprehensive approach combining these methods provides the most reliable differentiation between FGF19 and FGF21 biological activities.

How can researchers separate the metabolic benefits of FGF19 from its potentially harmful mitogenic effects?

Based on structural studies, researchers have successfully developed FGF19 variants with differential activities:

  • Strategic mutagenesis approaches:

    • Modify the N-terminal region (residues 38-42): Replace FGF19's 38WGDPI42 with FGF21's 41GQV43

    • Modify heparin-binding domains: Target β1-β2 loop and the β10–β12 segment

    • Combined mutations in both regions most effectively eliminate mitogenic activity while preserving metabolic benefits

  • Validated chimeric constructs:
    The following engineered variants have shown separated activities:

    VariantModificationsFGFR1c/βKlotho ActivationFGFR4 ActivationMetabolic ActivityMitogenic Activity
    FGF19-4N-terminal + β1-β2 loopPreservedAbolishedPreservedAbolished
    FGF19-5N-terminal + β10–β12PreservedAbolishedPreservedAbolished
    FGF19-6N-terminal + both HBS regionsPreservedAbolishedPreservedAbolished

    These variants maintain glucose-regulating effects without promoting hepatocyte proliferation .

  • Experimental validation:

    • Confirm receptor activation profiles in transfected L6 cells

    • Verify metabolic activity through glucose uptake assays and in vivo glucose measurements

    • Assess proliferative potential using BrdU incorporation in liver sections

This strategic engineering approach demonstrates the feasibility of developing FGF19-based therapeutics with improved safety profiles.

What are the molecular mechanisms underlying FGF19's receptor specificity compared to other FGF family members?

FGF19's unique receptor interactions involve multiple structural elements:

  • Key structural determinants:

    • N-terminal region (residues 38-42): Critical for FGFR4 interaction and specificity

    • Heparin-binding surface (HBS): Contributes to both heparin-dependent and βKlotho-dependent FGFR4 activation

    • These regions work independently but cooperatively

  • Receptor interaction model:

    • The five residues (38WPDPI42) likely directly contact the βC′-βE loop of the receptor D3 domain

    • This region has been associated with receptor specificity determination in paracrine-acting FGFs

    • Structural modeling suggests the extended HBS might interact directly with the receptor

  • Co-receptor dependencies:

    • Unlike most FGFs, FGF19 can activate FGFR4 in either a heparin-dependent or βKlotho-dependent manner

    • The βKlotho co-receptor provides specificity for endocrine FGFs (FGF19, FGF21, FGF23)

    • FGF19/βKlotho interaction involves distinct structural determinants from traditional FGF/heparin interactions

Understanding these molecular mechanisms enables rational design of FGF19 variants with tailored receptor activation profiles for specific research or therapeutic applications.

How do FGF19 mutations impact its functional activities in experimental systems?

Structural studies have revealed how specific mutations alter FGF19 functionality:

These mutation studies provide a structural and functional framework for understanding the molecular basis of FGF19 activity, enabling rational design of variants with specific activity profiles.

What are common pitfalls in FGF19 experimental design and how can they be avoided?

Researchers should be aware of these frequent challenges:

  • Receptor expression variability in cell models:

    • Problem: Inconsistent results due to varying FGFR expression levels between cell lots

    • Solution: Quantify receptor expression in each cell lot; consider using transfected L6 cells with controlled receptor expression for consistent results

  • βKlotho dependency misinterpretation:

    • Problem: Failing to account for βKlotho's role in receptor activation

    • Solution: Include controls with and without βKlotho; verify βKlotho expression in cell models

  • Protein storage and stability issues:

    • Problem: Activity loss due to improper handling

    • Solution: Use carrier proteins for stability; prepare single-use aliquots; verify activity before key experiments

  • Confounding factors in in vivo experiments:

    • Problem: Background metabolic variability masking FGF19 effects

    • Solution: Use age/sex-matched animals; control feeding status; include appropriate vehicle controls

  • Specificity validation:

    • Problem: Attributing non-specific effects to FGF19

    • Solution: Include inactive FGF19 mutants as controls; validate with receptor antagonists or knockdown approaches

Addressing these common pitfalls proactively improves experimental reproducibility and data quality.

How should researchers interpret contradictory results between in vitro and in vivo FGF19 studies?

When facing discrepancies between experimental systems:

  • Physiological complexity considerations:

    • In vivo systems involve multiple cell types, metabolic interactions, and compensatory mechanisms

    • Cell-specific responses may be diluted or enhanced in whole organism context

  • Systematic reconciliation approach:

    • Compare dosing: In vitro concentrations often exceed physiological levels

    • Examine timing differences: Acute vs. chronic exposure effects

    • Consider pharmacokinetics: Protein stability and distribution differ significantly between systems

    • Evaluate model appropriateness: Some cell lines may lack key signaling components

  • Bridging strategies:

    • Use ex vivo approaches (e.g., primary hepatocytes, liver slices) to bridge the gap

    • Employ tissue-specific conditional knockout models to isolate target tissue responses

    • Consider organoid models that better recapitulate tissue architecture

  • Technical validation:

    • Confirm protein activity in both systems using the same lot

    • Verify target engagement through receptor phosphorylation or downstream signaling markers

Careful analysis of these factors can often resolve apparent contradictions between experimental systems.

What advanced analytical approaches can resolve complex FGF19 signaling questions?

For investigating nuanced aspects of FGF19 biology:

  • Receptor-specific signaling discrimination:

    • Phosphoproteomic profiling to identify differential phosphorylation events

    • Temporal signaling analysis to detect differences in activation kinetics

    • CRISPR-based receptor editing to create clean systems for pathway analysis

  • Structural biology approaches:

    • Cryo-electron microscopy of FGF19-FGFR-cofactor complexes

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • Computational modeling to predict effects of mutations on receptor interaction

  • Single-cell analysis methods:

    • Single-cell RNA sequencing to identify cell-specific responses

    • Live-cell imaging with FRET-based sensors for real-time signaling visualization

    • Digital spatial profiling to map tissue-specific responses in heterogeneous samples

  • Multi-omics integration:

    • Combined transcriptomic, proteomic and metabolomic analyses

    • Network biology approaches to map signaling cascades

    • Machine learning algorithms to identify signaling signatures

These advanced approaches can uncover subtle but important aspects of FGF19 biology that conventional methods might miss, providing deeper mechanistic insights.

What are the emerging applications of engineered FGF19 variants in metabolic disease research?

Engineered FGF19 variants offer promising research avenues:

  • Therapeutic potential exploration:

    • Non-mitogenic variants (e.g., FGF19-4, FGF19-5, FGF19-6) provide tools to study metabolic benefits without cancer risk

    • These variants enable longer-term studies of FGF19's effects on obesity, diabetes, and NAFLD

  • Tissue-specific targeting strategies:

    • Engineering receptor-specific variants to target particular tissues

    • Developing tissue-specific delivery methods for FGF19 variants

  • Combination therapy models:

    • Investigating synergistic effects with established anti-diabetic agents

    • Exploring potential in resistant disease models

  • Translational potential assessment:

    • Evaluation in humanized mouse models

    • Comparative studies across species to predict human responses

These applications may lead to both improved research tools and potential therapeutic candidates for metabolic disorders.

How can advanced imaging techniques enhance our understanding of FGF19 biology?

Modern imaging approaches offer unique insights:

  • In vivo molecular imaging applications:

    • PET imaging with labeled FGF19 variants to track tissue distribution

    • Intravital microscopy to observe cellular responses in real time

    • CLARITY-based whole-organ imaging to map receptor distribution

  • Subcellular localization studies:

    • Super-resolution microscopy to track receptor-ligand interactions

    • Live-cell confocal imaging to monitor receptor trafficking

    • FRAP (Fluorescence Recovery After Photobleaching) to study binding dynamics

  • Functional imaging approaches:

    • Calcium imaging to monitor immediate signaling responses

    • Biosensor-based imaging to track metabolic changes in real-time

    • Label-free imaging technologies to study conformational changes

These techniques provide spatial and temporal information about FGF19 activity that complements traditional biochemical and molecular approaches.

What computational approaches can advance FGF19 research?

Computational methods offer powerful tools for FGF19 investigation:

  • Structural prediction and analysis:

    • Molecular dynamics simulations to predict effects of mutations

    • Protein-protein docking to model receptor interactions

    • AlphaFold2 and similar AI approaches to predict structural features

  • Systems biology frameworks:

    • Pathway modeling to predict effects of FGF19 in complex metabolic networks

    • Multi-scale modeling to bridge molecular and physiological effects

    • Agent-based models to simulate tissue-level responses

  • Machine learning applications:

    • Predictive models for FGF19 variant activity based on sequence

    • Pattern recognition in complex datasets to identify response biomarkers

    • Deep learning approaches to integrate multi-omics data

These computational strategies can accelerate hypothesis generation, experimental design, and data interpretation in FGF19 research.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.