FN3K (Fructosamine-3-kinase) is an enzyme that catalyzes ATP-dependent phosphorylation of fructosamines, which form when glucose reacts with amines through glycation. Its primary function is to destabilize and remove potentially harmful fructosamines from proteins, protecting them from the detrimental effects of nonenzymatic glycation . FN3K mediates phosphorylation of fructoselysine residues on glycated proteins to generate fructoselysine-3 phosphate adducts, which are unstable and decompose under physiological conditions . This enzymatic process effectively reverses protein glycation, making FN3K a critical component of the cellular protein quality control system . Additionally, FN3K is involved in the response to oxidative stress by mediating deglycation of NFE2L2/NRF2, as glycation impairs NFE2L2/NRF2 function .
Several validated antibodies are available for FN3K detection in Western blotting applications:
| Antibody | Type | Source | Catalog Number | Recommended Dilution |
|---|---|---|---|---|
| FN3K Antibody (E-9) | Mouse monoclonal IgG1 | Santa Cruz Biotechnology | sc-271503 | Not specified |
| FN3K antibody | Rabbit polyclonal | Proteintech | 14293-1-AP | 1:1000-1:4000 |
| FN3K Antibody (N-term)(Ascites) | Mouse monoclonal IgG1 | Abcepta | AM2193a | 1:1000-8000 |
| FN3K Polyclonal Antibody | Rabbit polyclonal | Thermo Fisher Scientific | PA5-66239 | Not specified |
When selecting an antibody for Western blotting, researchers should consider specificity, sensitivity, and validated applications reported by the manufacturer . The Proteintech antibody (14293-1-AP) has been extensively validated in multiple sample types including HeLa cells, human brain tissue, human heart tissue, human kidney tissue, and mouse skeletal muscle tissue .
Optimal antibody dilutions vary by application type and specific antibody:
| Application | Antibody (Catalog #) | Recommended Dilution |
|---|---|---|
| Western Blot (WB) | Proteintech (14293-1-AP) | 1:1000-1:4000 |
| Western Blot (WB) | Abcepta (AM2193a) | 1:1000-8000 |
| Immunofluorescence (IF/ICC) | Proteintech (14293-1-AP) | 1:50-1:500 |
| Immunoprecipitation (IP) | Proteintech (14293-1-AP) | 0.5-4.0 μg for 1.0-3.0 mg of total protein lysate |
| ELISA | Santa Cruz (E-9) | Not specified |
Manufacturers consistently recommend titrating each antibody in specific testing systems to obtain optimal results . For instance, Proteintech notes that sample-dependent optimization may be necessary and encourages checking their validation data gallery for application-specific guidance .
The calculated and observed molecular weights for human FN3K in Western blot analysis are:
| Source | Calculated MW | Observed MW |
|---|---|---|
| Proteintech (14293-1-AP) | 34 kDa | 35 kDa |
| Abcepta (AM2193a) | 35171 Da | Not specified |
| Thermo Fisher (PA5-66239) | Not specified | Not specified |
| FN3KRP antibody (Proteintech) | 34 kDa | 30-34 kDa |
When running Western blots, researchers should expect to see bands in the 30-35 kDa range, although post-translational modifications may cause slight variations in the observed molecular weight . It's important to note that FN3KRP (FN3K-related protein) has a similar molecular weight range (30-34 kDa), which should be considered when analyzing samples that might express both proteins .
FN3K plays a central role in protein deglycation through a specific enzymatic mechanism:
It catalyzes the phosphorylation of fructosamines formed by glycation (nonenzymatic reaction of glucose with primary amines followed by Amadori rearrangement)
This phosphorylation generates fructoselysine-3 phosphate adducts, which are unstable and decompose under physiological conditions
The decomposition effectively removes the glycation modification, resulting in deglycation of the protein
This deglycation process is particularly important for maintaining protein function and preventing the accumulation of advanced glycation end products (AGEs)
FN3K is considered one of the first examples of deglycating enzymes acting on the Amadori product to reverse the early stages of the Maillard reaction in vivo . This function is critical in conditions like diabetes, where abnormal FN3K expression can contribute to complications due to increased protein glycation .
FN3K is involved in the deglycation of Nrf2 (Nuclear factor erythroid 2-related factor 2, also known as NFE2L2), a significant regulator of oxidative stress in cancer cells . The relationship functions through the following mechanism:
Glycation of Nrf2 impairs its function as a transcription factor that regulates antioxidant responses
FN3K mediates deglycation of Nrf2, which restores its function
Deglycated Nrf2 predominantly induces antioxidant responses by binding to small MAF proteins (sMAF)
In cancer cells, this deglycation activity can offer protection against oxidative stress
For researchers studying this pathway, several approaches are recommended:
Using FN3K inhibitors to maintain Nrf2 in a glycated state, potentially reducing cancer cell protection against oxidative stress
Implementing combinatorial approaches using phytochemicals like brusatol, topotecan, and platinum derivatives along with kinase modulators
Monitoring the expression patterns of downstream targets of Nrf2, such as NQO1, HO-1, and their relationship with Keap1
Western blot analysis using antibodies against both FN3K and Nrf2 pathway proteins (Nrf2, NQO1, HO-1, Keap1) can help elucidate this relationship. Primary antibodies for these targets are commercially available: FN3K (Invitrogen, Catalog # PA5-28603), Nrf2 (cell signaling, cat#: 12721), NQO1 (cat#: 62262), HO-1 (Abcam, cat#: ab13248), and Keap1 (Abcam, cat#: ab227828) .
Based on the research data, FN3K knockout alters redox-sensitive cellular metabolites . To study this relationship, researchers can employ multiple complementary approaches:
CRISPR/Cas9-mediated knockout of FN3K in appropriate cell lines (HepG2 cells have been successfully used)
siRNA or shRNA knockdown for transient reduction of FN3K expression
Overexpression systems to examine the effects of increased FN3K activity
Treatment with 1-deoxy-1-morpholinofructose (DMF), a competitive inhibitor of FN3K that can inhibit ~10% of total FN3K activity
Screening and utilizing anticancer molecules that interact with the catalytic domain of FN3K using computational approaches such as molecular docking
¹H NMR metabolomics to measure changes in metabolite abundance, as described in published research
Focus on measuring redox-sensitive metabolites including:
Monitor glycation status of key redox-regulating proteins
Assess Nrf2 pathway activation through Western blotting of Nrf2, NQO1, HO-1, and Keap1
Evaluate nuclear translocation of Nrf2 using immunofluorescence or subcellular fractionation techniques
These approaches provide complementary data on how FN3K modulates the cellular redox state, potentially through its deglycating activity on key proteins involved in oxidative stress responses.
To accurately quantify FN3K-mediated deglycation of specific protein targets, researchers should employ a multi-faceted approach:
LC-MS/MS analysis to identify and quantify glycated vs. deglycated peptides
MALDI-TOF MS to assess mass shifts associated with glycation/deglycation
Multiple reaction monitoring (MRM) for targeted analysis of specific glycation sites
Western blotting using specific antibodies against FN3K (such as Proteintech's 14293-1-AP)
ELISA-based quantification using sandwich assays like ELK Biotechnology's Human FN3K ELISA Kit (detection range: 0.32-20 ng/mL)
Immunoprecipitation to isolate specific glycated proteins followed by deglycation assays
Comparison between wild-type and FN3K knockout/knockdown cells
Pulse-chase experiments to track glycation/deglycation dynamics
Focus on known FN3K substrates such as Nrf2
Monitor functional changes in target proteins (e.g., transcriptional activity of Nrf2)
Correlate deglycation with functional outcomes through reporter assays
For optimal quantification, researchers should consider combining these approaches to provide complementary data on both the extent of deglycation and its functional significance.
When designing FN3K inhibitor screening experiments, researchers should consider several critical factors based on published methodologies:
The 3-dimensional structure of Human FN3K should be modeled using homology modeling techniques, as there is no available crystal structure in PDB
The amino acid FASTA sequences (Uniprot, 309 AA, accession number: Q9H479) can be used as the starting point
Specific templates like FN3K from Arabidopsis thaliana (PDB: 6OID) can be used with 'SWISS EXPASY Tools'
Start with computational approaches:
Validated compounds to consider including:
Molecular dynamics simulations using tools like Gromacs 5.0
ATP binding loop interactions analysis
In vitro enzymatic assays to confirm computational predictions
Cell-based assays measuring FN3K activity in the presence of inhibitors
Western blotting to assess effects on FN3K protein expression and downstream targets
Direct measurement of FN3K enzymatic activity
Assessment of target protein deglycation efficiency
Effects on Nrf2 pathway activation
Metabolomic analysis to detect changes in redox-sensitive metabolites
Cytotoxicity assays in relevant cell lines (BT-474, T-47D have been used in published research)
By incorporating these considerations, researchers can develop robust screening platforms for identifying and validating novel FN3K inhibitors with potential therapeutic applications.
¹H NMR metabolomics analysis has revealed significant differences in metabolite abundance between FN3K knockout and wild-type cells . These changes and their implications include:
| Metabolite | Change in FN3K-KO | Biological Significance |
|---|---|---|
| Glutathione | Increased | Major cellular redox regulator |
| Lactate | Increased | Controls glycolysis |
| Pantothenate | Decreased | Reactive to cellular redox status |
| Phosphocreatine/creatine ratio | Decreased | Controls ATP production |
| Aspartate | Decreased | Reactive to cellular redox status |
| Glycine | Decreased | Reactive to cellular redox status |
| Serine | Decreased | Reactive to cellular redox status |
Redox Homeostasis: The enrichment of glutathione (a major cellular redox regulator) suggests FN3K plays a role in maintaining redox balance . Increased glutathione in FN3K-KO cells may represent a compensatory mechanism to manage altered redox states.
Energy Metabolism: Decreased phosphocreatine/creatine ratio indicates potential alterations in ATP production mechanisms . Combined with increased lactate, this suggests a shift in energy metabolism, possibly toward enhanced glycolysis.
Amino Acid Metabolism: Reductions in aspartate, glycine, and serine, all known to be reactive to cellular redox status, further support FN3K's involvement in redox regulation .
Potential Therapeutic Implications: These metabolic changes suggest targeting FN3K could simultaneously affect cancer cell metabolism and redox defenses, representing a potential dual-action therapeutic strategy.
Biomarker Development: The consistent metabolic signature in FN3K-deficient cells suggests these metabolites could serve as biomarkers for effective FN3K inhibition in therapeutic settings.
To fully understand these implications, researchers should consider integrating metabolomic data with other omics approaches (transcriptomics, proteomics) and validate findings across multiple cell types and in vivo models.
For optimal Western blotting detection of FN3K, researchers should follow these validated protocols:
Use validated positive control samples: HeLa cells, human brain/heart/kidney tissues, or mouse skeletal muscle tissue
Prepare whole cell lysates using standard lysis buffers containing protease inhibitors
Include phosphatase inhibitors if phosphorylation status is important
Load 20-40 μg of total protein per lane
Consider using pre-cast gradient gels (4-20%) for optimal resolution around this molecular weight
Primary antibodies:
Secondary antibodies:
Anti-rabbit or anti-mouse HRP-conjugated antibodies depending on primary
Dilute according to manufacturer's recommendations
Blocking: Use 5% non-fat dry milk or BSA in TBST
Primary antibody incubation: Overnight at 4°C or 1.5 hours at room temperature
Include positive and negative controls to validate specificity
For challenging samples, consider using an m-IgG Fc BP-HRP Bundle (like sc-527252) to reduce background
Enhanced chemiluminescence (ECL) works well for FN3K detection
For quantitative analysis, consider fluorescent secondary antibodies and imaging systems
Exposure times may need optimization depending on expression levels
Properly executed Western blotting can reliably detect FN3K in various human and mouse samples, with validation data showing clear bands at the expected molecular weight .
FN3K and FN3KRP (FN3K-related protein) share structural and functional similarities that can create challenges in experimental differentiation:
Molecular Weight: FN3K is observed at 35 kDa while FN3KRP is observed at 30-34 kDa
Substrate Specificity: Both phosphorylate ketosamines but with different preferences
Use specific antibodies validated for each protein:
Verify specificity through knockout/knockdown validation
Consider using epitope-tagged constructs for overexpression studies
Run both proteins on the same gel for direct comparison
Include appropriate positive controls for each protein
Consider immunoprecipitation followed by mass spectrometry for definitive identification
When possible, assess enzyme activity with substrate specificity assays
Compare expression across tissues (FN3K is prominently expressed in erythrocytes)
Use RT-PCR with specific primers to distinguish between their mRNA expression
Consider single-cell analysis techniques to map expression at cellular resolution
By carefully selecting antibodies and experimental approaches that exploit the known differences between these related proteins, researchers can accurately distinguish between FN3K and FN3KRP in their studies.
For accurate ELISA-based quantification of FN3K in biological samples, researchers should consider the following validated approaches:
ELK Biotechnology's Human FN3K ELISA Kit offers:
Proper sample collection and processing is critical:
Serum: Allow blood to clot, centrifuge and collect serum
EDTA/Heparin plasma: Collect blood with anticoagulant, centrifuge promptly
Tissue homogenates: Homogenize in PBS (pH 7.2-7.4), centrifuge to remove debris
Cell culture supernatants: Collect and centrifuge to remove particulates
Sample dilution is often necessary:
Run samples in duplicate or triplicate
Include known positive and negative controls
Consider spike-recovery experiments to validate accuracy in specific sample types
Recovery ranges:
By following these guidelines, researchers can achieve reliable quantification of FN3K in various biological samples, enabling accurate comparison between experimental conditions and contributing to reproducible research outcomes.
Emerging research reveals important connections between FN3K and cancer biology, suggesting several promising therapeutic avenues:
FN3K is involved in the deglycation of Nrf2, a significant regulator of oxidative stress in cancer cells
Deglycated Nrf2 predominantly induces antioxidant responses by binding to sMAF proteins
This activation offers cancer cell protection against oxidative stress
Targeting FN3K could maintain Nrf2 in a glycated state, potentially sensitizing cancer cells to oxidative stress-inducing therapies
FN3K inhibition could represent a novel approach to modulate cancer cell redox homeostasis
Combinatorial approaches using:
Molecular docking studies have identified several potential FN3K inhibitors with anticancer potential, including:
FN3K knockout alters metabolites critical for cancer cell survival:
These metabolic vulnerabilities could be exploited alongside existing cancer therapies
Breast cancer cell lines (BT-474, T-47D) have been successfully used in FN3K studies
These models provide platforms for screening therapeutic compounds and studying resistance mechanisms
The emerging understanding of FN3K's role in cancer biology highlights its potential as a therapeutic target, particularly through its influence on Nrf2-mediated antioxidant responses and cancer cell metabolism.
While the search results focus primarily on cancer research, FN3K plays a crucial role in diabetes complications through its protein deglycation function:
FN3K catalyzes phosphorylation of fructosamines formed by glycation, which is accelerated in hyperglycemic conditions
This enzymatic action helps destabilize and ultimately remove potentially harmful fructosamines from proteins
In diabetes, abnormal FN3K expression or activity can contribute to complications through accumulation of glycated proteins
FN3K antibodies enable researchers to:
Quantify FN3K expression in diabetic vs. non-diabetic tissues
Correlate FN3K levels with markers of glycation damage
Evaluate potential therapeutic interventions targeting the glycation pathway
ELISA kits with sensitivity of 0.119 ng/mL allow precise quantification of FN3K in patient samples
Western blotting protocols using validated antibodies permit assessment of FN3K expression in various tissues
Immunohistochemistry can reveal tissue-specific changes in FN3K distribution in diabetic complications
Modulating FN3K activity could potentially reduce protein glycation damage
1-deoxy-1-morpholinofructose (DMF), a competitive inhibitor of FN3K, has been reported to inhibit approximately 10% of total FN3K activity
FN3K activation might represent a protective mechanism against glycation-induced protein damage
As research progresses, FN3K antibodies will continue to be essential tools for investigating the role of this enzyme in diabetes pathophysiology and developing targeted interventions to mitigate glycation-related complications.