Nik-related kinase (NRK) is a protein coding gene, also known as Nik-related protein kinase . The human NRK gene encodes a protein with 1582 amino acids and a molecular weight of approximately 178 kDa . It belongs to the Ste20-type kinase family . Diseases associated with NRK include hypermobility syndrome, and its related pathways include NAD metabolism .
DKFZp686A17109
HGNC: 25391
NCBI Gene: 203447
Ensembl: ENSG00000123572
OMIM®: 300791
UniProtKB/Swiss-Prot: Q7Z2Y5
NRK is involved in the TNF-alpha-induced signaling pathway and may phosphorylate cofilin-1, inducing actin polymerization during late embryogenesis . The mouse ortholog of this gene encodes a protein kinase required for JNK activation and may be involved in the induction of actin polymerization in late embryogenesis .
NRK is expressed in various tissues, including the ovary and placenta . It is also expressed in cultured vascular smooth muscle cells (VSMC) and mouse arterial intima .
NRK is expressed in normal VSMCs, but its expression is significantly reduced in neointimal and atherosclerotic regions in murine and human arteries . Treatment of mouse VSMCs with lipopolysaccharide (LPS) or platelet-derived growth factor (PDGF) significantly reduces NRK expression . Reduced NRK expression is associated with diabetes, hypertension, ischemic heart disease, and atherosclerosis .
Reduced NRK expression is associated with the induction of matrix metalloproteinases (MMP3, MMP8, and MMP12) and inflammatory cytokines/chemokines (CCL6, CCL8, CCL11, CXCL1, CXCL3, CXCL5, and CXCL9) in VSMCs . Silencing of NRK and LPS treatment synergistically increases the expression of these factors .
Resveratrol significantly impairs LPS- and Nrk siRNA-induced expression of MMP3, CXCL3, CXCL5, CCL8, and CCL11 .
Actin Polymerization: NRK may phosphorylate cofilin-1 and induce actin polymerization through this process during the late stages of embryogenesis .
JNK Activation: The mouse ortholog of NRK encodes a protein kinase required for JNK activation .
Hypermobility Syndrome: Diseases associated with NRK include hypermobility syndrome .
NAD Metabolism: Among its related pathways is NAD metabolism .
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional fees.
The tag type is determined during production. Please specify your desired tag type for preferential development.
NRK (Nik Related Kinase) is a protein-coding gene that encodes a serine/threonine kinase involved in several critical cellular processes. The protein functions primarily as a signaling molecule with roles in the TNF-alpha-induced signaling pathway. One of its most significant functions appears to be phosphorylating cofilin-1, thereby inducing actin polymerization during late embryogenesis . This activity suggests NRK plays an important role in cytoskeletal organization and potentially in cellular morphogenesis. NRK belongs to the protein kinase superfamily and demonstrates transferase activity, specifically transferring phosphorus-containing groups. It also exhibits protein tyrosine kinase activity according to Gene Ontology (GO) annotations .
Research indicates NRK is associated with certain pathological conditions, particularly Hypermobility Syndrome . Its relationship with the NAD metabolism pathway suggests additional roles in cellular energy homeostasis and metabolic regulation. An important paralog of NRK is MAP4K4, which may provide insight into overlapping or complementary functions in cellular signaling cascades .
Based on patterns observed with similar kinases, several expression systems can be employed for recombinant NRK production, each with distinct advantages:
| Expression System | Advantages | Considerations | Recommended For |
|---|---|---|---|
| Bacterial (E. coli) | High yield, cost-effective, rapid | Potential misfolding, lacks post-translational modifications | Initial screening, structural studies |
| Insect cells (Sf9/Sf21) | Proper folding, post-translational modifications | Moderate yield, higher cost | Functional studies, enzyme activity assays |
| Mammalian cells | Native-like processing, appropriate modifications | Lower yield, most expensive, time-consuming | In-depth functional characterization |
| Wheat germ in vitro | Good for difficult-to-express proteins | Moderate yield | Alternative when other systems fail |
For most kinase proteins requiring proper folding and post-translational modifications, insect cell expression systems (such as Sf9 cells) are frequently employed as they provide a balance between yield and functional protein production . This is particularly important for NRK given its role in phosphorylation cascades where proper folding and activation are crucial for downstream functional studies.
Validation of recombinant NRK should follow a multi-method approach:
SDS-PAGE analysis: Evaluating protein size and initial purity assessment through Coomassie Blue staining. For NRK, the expected molecular weight is approximately 54-55 kDa, though this may vary with added tags.
Western blot confirmation: Using specific anti-NRK antibodies to confirm protein identity.
Mass spectrometry analysis: For definitive identification and sequence coverage verification of the recombinant protein.
Kinase activity assay: Functional validation using a substrate such as cofilin-1, with detection of phosphorylation via:
Radiometric assays using [γ-32P]ATP
Phospho-specific antibodies in western blot
Mass spectrometry to identify phosphorylation sites
Purity assessment: Using analytical techniques such as size exclusion chromatography or high-performance liquid chromatography.
A comprehensive validation approach combining at least three of these methods is recommended to ensure both identity and functional integrity of the recombinant NRK preparation.
Optimizing buffer conditions is essential for preserving the functionality of recombinant NRK:
| Buffer Component | Recommended Range | Function | Notes |
|---|---|---|---|
| pH | 7.2-7.8 | Stability | Most kinases maintain optimal activity in this range |
| Salt (NaCl) | 50-150 mM | Ionic strength | Higher concentrations may stabilize but inhibit activity |
| Reducing agent | 1-5 mM DTT or 0.5-2 mM TCEP | Prevent oxidation | TCEP more stable for longer storage |
| Glycerol | 10-25% | Stability | Prevents aggregation and extends shelf-life |
| Divalent cations | 5-10 mM MgCl₂ | Cofactor | Essential for kinase activity |
| ATP | 0.1-1 mM | Substrate | Include in activity assays, not long-term storage |
| Protease inhibitors | Cocktail | Prevent degradation | Essential during initial purification |
Research indicates that most protein kinases, including those in the same family as NRK, show optimal stability when stored at -80°C in small aliquots to avoid repeated freeze-thaw cycles. For working solutions, maintaining the protein on ice and limiting exposure to room temperature is recommended to preserve enzymatic activity.
Investigating the NRK-mediated phosphorylation of cofilin-1 requires thoughtful experimental design:
In vitro kinase assay setup:
Purified recombinant NRK (active)
Purified recombinant cofilin-1 (substrate)
ATP (typically 50-100 μM)
Appropriate kinase buffer with Mg²⁺ or Mn²⁺
Incubation at 30°C for 15-60 minutes
Detection methods:
Western blotting with phospho-specific antibodies against phosphorylated cofilin-1
Mass spectrometry to identify specific phosphorylation sites
Functional actin polymerization assays to correlate phosphorylation with cytoskeletal changes
Controls to include:
Heat-inactivated NRK (negative control)
Known cofilin-1 kinase such as LIMK1 (positive control)
Kinase inhibitor controls
Phosphatase treatment to confirm reversibility
Cellular validation approaches:
Transfection of wild-type vs. kinase-dead NRK mutants
siRNA knockdown of endogenous NRK
Visualization of actin dynamics following NRK manipulation
The interpretation of results should consider the potential influence of NRK on TNF-alpha signaling pathways, as NRK has been implicated in this cascade . Researchers should also investigate the temporal relationship between NRK activation, cofilin-1 phosphorylation, and subsequent actin polymerization changes.
Purification of active NRK presents several challenges that can be addressed through specific strategies:
| Challenge | Recommended Solution | Scientific Rationale |
|---|---|---|
| Protein aggregation | Add 0.05-0.1% non-ionic detergent (e.g., Triton X-100) | Disrupts hydrophobic interactions without denaturing |
| Low solubility | Use fusion tags (GST, MBP) | Enhances solubility and provides purification handle |
| Proteolytic degradation | Add protease inhibitor cocktail and minimize purification time | Reduces exposure to endogenous proteases |
| Loss of activity during purification | Include ATP/ADP during purification | Stabilizes kinase domain conformation |
| Insufficient purity | Implement multi-step purification (affinity + ion exchange + gel filtration) | Removes contaminants with different properties |
| Co-purifying chaperones | Include ATP/Mg²⁺ wash steps | Promotes release of bound chaperones |
When working with recombinant proteins like NRK, incorporating affinity tags such as GST (glutathione S-transferase) or polyhistidine can significantly facilitate purification while potentially enhancing solubility . The choice between N-terminal versus C-terminal tagging should be empirically determined, as the kinase domain structure and function may be differently affected depending on tag placement.
For NRK specifically, consideration should be given to the potential impact of tags on its interaction with cofilin-1 and its ability to participate in TNF-alpha signaling pathways.
Understanding the expression pattern of NRK provides important context for research:
The expression of NRK appears to be developmentally regulated, with significant roles during embryogenesis particularly related to actin polymerization . This suggests temporal specificity in its function. Based on information about similar kinases, researchers should consider:
Tissue-specific expression analysis:
RT-qPCR to quantify mRNA levels across tissues
Western blotting with specific antibodies to detect protein levels
Immunohistochemistry for spatial localization within tissues
Developmental regulation:
Time-course studies during embryonic development
Correlation with actin cytoskeleton remodeling events
Relationship to TNF-alpha signaling pathway maturation
Regulatory mechanisms to investigate:
Transcriptional control elements in the NRK promoter
Post-translational modifications affecting protein stability
Subcellular localization changes during development
When designing experiments to study NRK expression, researchers should be mindful of its X-chromosome location (as indicated by the gene identifiers) , which may result in sex-specific expression patterns that should be accounted for in experimental design.
To elucidate NRK's function in TNF-alpha signaling:
Stimulation experiments:
Treat cells expressing NRK with TNF-alpha at various concentrations (typically 10-50 ng/ml)
Conduct time-course analysis (5 minutes to 24 hours)
Monitor NRK activation (phosphorylation state)
Track downstream signaling components (JNK, p38, NF-κB)
Genetic manipulation strategies:
CRISPR/Cas9-mediated NRK knockout
Doxycycline-inducible NRK expression systems
Domain-specific mutants to identify critical functional regions
Interaction studies:
Co-immunoprecipitation to identify binding partners
Proximity ligation assays for in situ interaction analysis
Yeast two-hybrid screening for novel interactors
Signaling pathway analysis:
Phosphoproteomic profiling before and after TNF-alpha stimulation
Inhibitor studies to delineate pathway dependencies
Comparison with known TNF-alpha response genes
Functional readouts:
Research designs should include appropriate controls such as cells lacking NRK expression, treatment with TNF-alpha blocking antibodies, and comparison with effects of other TNF family cytokines to establish specificity.
Researchers frequently encounter specific challenges when working with kinases like NRK:
| Common Issue | Underlying Cause | Prevention/Solution |
|---|---|---|
| Loss of enzymatic activity | Improper storage, oxidation | Store in small aliquots with reducing agents, avoid freeze-thaw cycles |
| False negative in activity assays | Incorrect buffer conditions, missing cofactors | Optimize Mg²⁺/Mn²⁺ concentration, include fresh ATP |
| Substrate specificity confusion | Promiscuity in vitro | Validate with multiple methods, include specificity controls |
| Inconsistent phosphorylation | ATP limitation, phosphatase contamination | Ensure sufficient ATP, add phosphatase inhibitors |
| Poor reproducibility | Batch-to-batch variation | Standardize production, include internal activity standards |
| Artifactual results | Tag interference | Compare tagged vs. untagged proteins, use cleavable tags |
When designing experiments with recombinant NRK, researchers should include appropriate positive controls (such as a well-characterized kinase with known activity) and negative controls (heat-inactivated NRK or a kinase-dead mutant). Additionally, considering that NRK may phosphorylate cofilin-1 , including this as a substrate in activity assays can serve as a relevant biological readout.
Differentiating direct from indirect effects requires methodical approaches:
In vitro reconstitution:
Purified components only (NRK, potential substrate, ATP)
Minimal system to demonstrate direct phosphorylation
Kinase-dead controls:
K→R or D→N mutations in the catalytic site
Expression of kinase-dead mutant as dominant negative
Temporal resolution:
Rapid kinetics measurements (seconds to minutes)
Direct effects occur faster than indirect ones
Substrate trapping approaches:
ATP-binding pocket mutations that trap substrates
Substrate identification by mass spectrometry
Specific inhibitor studies:
ATP-competitive vs. allosteric inhibitors
Dose-dependent inhibition patterns
Phosphosite mapping:
Identification of consensus motifs for direct phosphorylation
Mutational analysis of potential phosphosites
When investigating NRK's role in TNF-alpha signaling , careful time-course experiments with specific attention to the order of molecular events can help distinguish direct phosphorylation targets from downstream effectors.
To investigate NRK's connection to NAD metabolism :
Metabolic profiling:
Measure NAD⁺/NADH levels in NRK-expressing vs. knockout cells
Monitor NAD⁺ biosynthesis pathway intermediates
Isotope tracing to track NAD⁺ turnover rates
Enzyme activity correlations:
Assess activities of key NAD metabolism enzymes (NAMPT, NMNAT, PARP)
Determine if NRK directly phosphorylates any NAD metabolism enzymes
Genetic interaction studies:
Combinatorial knockdown/knockout of NRK with NAD metabolism enzymes
Complementation assays with NAD precursors
Subcellular localization:
Co-localization analysis of NRK with NAD metabolism enzymes
Compartment-specific NAD⁺ pool measurements
Functional readouts:
Sirtuin activity assays (NAD⁺-dependent deacetylases)
Poly-ADP-ribosylation levels
Mitochondrial function assessments
Research designs should include controls for cellular energy status and stress conditions that might independently affect NAD metabolism. Considering the link between NAD metabolism and various cellular processes, researchers should also examine potential effects on cell survival, DNA repair, and metabolic adaption when studying NRK in this context.
When faced with contradictory results regarding NRK function:
Contextual differences analysis:
Cell/tissue type variations
Experimental conditions (growth factors, stress, cell density)
Expression levels (endogenous vs. overexpression)
Technical considerations:
Antibody specificity issues
Tag interference with function
Detection method sensitivity limits
Isoform-specific effects:
Alternative splicing variants
Post-translational modification states
Complexes with different binding partners
Systematic validation approach:
Multiple independent methods to test the same hypothesis
Genetic rescue experiments (express wild-type in knockout)
Dose-response relationships to establish causality
Reporting framework:
Document all experimental conditions thoroughly
Consider publishing contradictory results as they may reveal unknown regulatory mechanisms
Discuss limitations transparently
When evaluating NRK's role in processes like actin polymerization through cofilin-1 phosphorylation , researchers should be particularly attentive to the cellular context, as cytoskeletal regulation often shows cell type-specific patterns and is influenced by multiple converging pathways.
Researchers can leverage various bioinformatic resources for NRK studies:
For kinase-specific analyses, specialized resources like KinBase and PhosphoSitePlus can provide valuable information on consensus phosphorylation motifs and known substrates of kinases related to NRK. Researchers should also consider using protein domain analysis tools to understand the functional organization of NRK and identify critical regions for mutagenesis studies.
Phosphoproteomics offers powerful insights into NRK biology:
Global phosphoproteome analysis:
Compare phosphorylation landscapes in NRK-expressing vs. knockout cells
Identify direct and indirect targets through temporal dynamics
Quantify changes in phosphorylation at specific sites
Experimental design considerations:
Include multiple time points (5, 15, 30, 60 minutes) after NRK activation
Use both label-free and isotope labeling approaches (SILAC, TMT)
Include phosphatase inhibitors during sample preparation
Data analysis strategies:
Motif analysis to identify consensus phosphorylation sites
Pathway enrichment of differentially phosphorylated proteins
Integration with protein-protein interaction networks
Validation of phosphoproteomic findings:
Targeted validation by phospho-specific antibodies
Functional studies of key phosphorylation events
Mutagenesis of identified phosphosites
Advanced approaches:
Kinase-substrate relationship mapping
Phosphorylation dynamics modeling
Cross-referencing with other post-translational modifications
Phosphoproteomic studies are particularly relevant for investigating NRK's role in the TNF-alpha signaling pathway , as this pathway involves complex phosphorylation cascades that can be comprehensively mapped using these technologies. Additionally, such studies can provide insights into how NRK may influence actin dynamics through cofilin-1 phosphorylation and potentially reveal novel substrates.
Several cutting-edge approaches could accelerate NRK research:
CRISPR-based technologies:
Base editing for precise point mutations in endogenous NRK
CRISPRi/CRISPRa for temporal control of expression
CRISPR screens to identify genetic interactions
Protein engineering approaches:
Engineered allosteric switches for controlled activation
Optogenetic regulation of NRK activity
FRET-based biosensors for real-time activity monitoring
Single-cell technologies:
Single-cell phosphoproteomics
Single-cell RNA-seq for expression heterogeneity
Spatial transcriptomics for tissue context
Advanced imaging:
Super-resolution microscopy for subcellular localization
Live-cell imaging of kinase activity using genetically encoded sensors
Correlative light and electron microscopy for structural context
Structural biology advances:
Cryo-EM for complex structure determination
Hydrogen-deuterium exchange mass spectrometry for conformational dynamics
Integrative structural biology combining multiple data types
These technologies could be particularly valuable for understanding NRK's role during embryogenesis and in TNF-alpha signaling pathways , where temporal and spatial regulation is likely critical to its function.
Critical knowledge gaps in NRK research include:
Substrate specificity:
Regulation mechanisms:
How is NRK itself activated/inactivated?
What upstream signals regulate NRK during embryogenesis?
Are there feedback mechanisms controlling NRK activity?
Structural insights:
What is the three-dimensional structure of NRK?
How does ATP binding and hydrolysis couple to phosphoryl transfer?
What conformational changes occur during activation?
Physiological roles:
Therapeutic relevance:
Could NRK be a target for cytoskeletal regulation disorders?
Is NRK dysregulation involved in inflammatory conditions?
What are the consequences of NRK inhibition in adult tissues?
Addressing these questions will require interdisciplinary approaches combining biochemistry, cell biology, structural biology, and systems biology perspectives.