CDK5 is a proline-directed serine/threonine kinase belonging to the cyclin-dependent kinase family. Unlike other CDKs, it does not regulate the cell cycle but is critical for neuronal development, synaptic plasticity, and cytoskeletal organization . The "Human, Sf9" designation indicates recombinant human CDK5 expressed in Sf9 insect cells, often co-expressed with activating partners like p25 or p35 to ensure kinase activity .
Molecular Weight: ~34–60 kDa (varies based on tags and co-expressed partners) .
Amino Acid Sequence: 298 residues (1–292) with a C-terminal His tag or N-terminal GST tag .
Formulation: Stabilized in phosphate-buffered saline (pH 7.4) with glycerol and reducing agents .
Cytoskeletal Regulation: Phosphorylates neurofilament heavy chain and CRMP2A to support axon growth .
Tau Phosphorylation: Hyperactivation by p25 (a truncated form of p35) leads to neurofibrillary tangle formation, implicated in Alzheimer’s disease .
Angiogenesis: Upregulates HIF-1α to promote tumor vascularization .
Immune Evasion: Enhances PD-L1 expression, enabling tumors to evade T-cell detection .
Silencing CDK5 in transgenic mice reduced tau phosphorylation by 52.5% and decreased neurofibrillary tangles, suggesting therapeutic potential for Alzheimer’s disease .
Dinaciclib: A CDK5 inhibitor (IC₅₀ = 1–4 nM) reduced tumor growth in medulloblastoma models by blocking PD-L1 upregulation .
Tamoxifen: Repurposed as a CDK5 inhibitor, it reduced tau phosphorylation and tumor proliferation in preclinical trials .
Inhibitor | Targets | Phase | Clinical Trials (Examples) |
---|---|---|---|
Dinaciclib | CDK1/2/5/9 | II | NCT02684617 (multiple myeloma) |
Roscovitine | CDK2/5 | II | NCT00999401 (non-small cell lung cancer) |
Flavopiridol | CDK1/2/4/9 | II | NCT02520011 (leukemia) |
CDK5, Cyclin Dependent Kinase 5, Serine/Threonine-Protein Kinase PSSALRE, Tau Protein Kinase II Catalytic Subunit, Cell Division Protein Kinase 5, TPKII Catalytic Subunit, Cyclin-Dependent-Like Kinase 5, Protein Kinase CDK5 Splicing, EC 2.7.11.22, EC 2.7.11.1, EC 2.7.11, PSSALRE, CDKN5, LIS7.
MQKYEKLEKI GEGTYGTVFK AKNRETHEIV ALKRVRLDDD DEGVPSSALR EICLLKELKH KNIVRLHDVL HSDKKLTLVF EFCDQDLKKY FDSCNGDLDP EIVKSFLFQL LKGLGFCHSR NVLHRDLKPQ NLLINRNGEL KLADFGLARA FGIPVRCYSA EVVTLWYRPP DVLFGAKLYS TSIDMWSAGC IFAELANAGR PLFPGNDVDD QLKRIFRLLG TPTEEQWPSM TKLPDYKPYP MYPATTSLVN VVPKLNATGR DLLQNLLKCN PVQRISAEEA LQHPYFSDFC PPHHHHHH
CDK5 is an atypical cyclin-dependent kinase that, unlike other CDKs, is primarily active in post-mitotic neurons rather than regulating cell cycle progression. It plays critical roles in neuronal migration, axonal guidance, synaptic plasticity, and neurotransmission . CDK5 has more than 30 identified substrates involved in various cellular pathways .
The significance of CDK5 in neuroscience stems from its dual nature - while essential for normal nervous system development and function, its dysregulation (particularly through formation of the CDK5/p25 complex) is implicated in neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis . This makes CDK5 both critical for understanding neuronal physiology and a potential therapeutic target for neurological disorders.
CDK5 is unique among CDKs in several key ways:
Activation mechanism: While most CDKs are activated by cyclins, CDK5 is activated by the non-cyclin proteins p35 and p39, or their proteolytic fragments p25 and p29 .
Expression pattern: Most CDKs are active in dividing cells and regulate cell cycle progression, whereas CDK5 is primarily active in post-mitotic neurons .
Structural differences: The T-loop of CDK5 is highly conserved from mammals to insects but distinct from other CDKs like CDK2, contributing to its unique interactions with activators and substrates .
Substrate repertoire: CDK5 phosphorylates numerous neuronal proteins including microtubule-binding proteins (TAU, MAP2, MAP1B), small GTPases (PAK1, RAC1, RHOA, CDC42), and proteins involved in exocytosis and endocytosis at synaptic terminals .
Physiological roles: CDK5 has specialized functions in the nervous system rather than cell cycle control, including roles in the regulation of the circadian clock through phosphorylation of CLOCK protein .
The CDK5/p35 and CDK5/p25 complexes represent physiological and pathological states of CDK5 activation, respectively:
Features a short half-life (20-30 minutes) enabling tight regulation of CDK5 activity
Contains an N-terminal myristoylation signal targeting the complex to cell membranes
Regulates normal neuronal functions including migration, axon guidance, and synaptic plasticity
p25 is a truncated form of p35 produced by calpain-mediated cleavage under neurotoxic conditions
Lacks the N-terminal myristoylation signal, allowing relocalization away from membranes
Has a longer half-life (2-3 hours), leading to prolonged and dysregulated CDK5 activity
Can hyperphosphorylate substrates like tau protein, contributing to neurofibrillary tangle formation in Alzheimer's disease
Associated with neurodegeneration in various conditions including Alzheimer's and Parkinson's disease
Sf9 cells (derived from Spodoptera frugiperda) combined with baculovirus expression systems offer significant advantages for expressing functional human CDK5 complexes:
Post-translational modifications: Sf9 cells can perform many mammalian post-translational modifications required for kinase functionality
Complex formation: Allows co-expression of CDK5 with its activator (p25 or p35) to form active complexes
Protein yield: Typically produces higher yields of recombinant protein compared to mammalian expression systems
Protein folding: Provides an environment that allows proper folding of complex mammalian proteins
Scalability: The baculovirus/Sf9 system can be scaled up relatively easily for producing larger amounts of protein
Purity: Can achieve >90% purity suitable for structural and functional studies
For CDK5 specifically, expression in Sf9 cells allows researchers to obtain the protein in complex with its activator, which is essential for studying kinase activity and developing inhibitors.
Several methodologies are employed to accurately assess CDK5 kinase activity:
Radioactive assays using [γ-32P]ATP and measuring incorporation into substrates like histone H1 or tau peptides
Non-radioactive assays using phospho-specific antibodies or fluorescent readouts
Kinase activity assessment with purified recombinant CDK5/p25 or CDK5/p35 complexes
Western blotting with phospho-specific antibodies detecting CDK5-mediated phosphorylation of substrates like tau protein
Mass spectrometry-based phosphoproteomics to identify and quantify phosphorylation sites
Microscale thermophoresis (MST) to measure binding affinities between CDK5 complexes and inhibitors
Biolayer interferometry (BLI) for real-time kinetic analysis of protein-inhibitor interactions
Ensuring specificity by using appropriate controls (kinase-dead CDK5 mutants, other kinase inhibitors)
Distinguishing CDK5 activity from other proline-directed kinases with overlapping substrate specificity
The evolution of peptide inhibitors for CDK5/p25 has progressed from longer, less specific peptides to shorter, more selective inhibitors:
CIP (CDK5 inhibitory peptide): A larger peptide derived from p35
p5 peptide: A 24-amino acid fragment from p35 that demonstrated selectivity for CDK5/p25 over CDK5/p35
Systematic amino acid substitutions: Each residue in p5 was systematically replaced with homologous residues to identify critical inhibitory elements
C-terminal modifications: A/V substitution at the C-terminus (creating p5-MT) significantly enhanced inhibitory potency
Length reduction: The 12-amino acid Cdk5i peptide showed greater specificity than longer peptides while being considerably smaller
Targeting unique structural features: The Cdk5i peptide targets the T-loop of CDK5, which is distinct from other CDKs
Focusing on interface residues: Incorporating residues (R149, A150, I153, C157, S159) that directly mediate interactions between CDK5 and p25
Enhancing specificity: The best inhibitors show >40-fold stronger binding to CDK5/p25 than to CDK5 alone or CDK2
Improving delivery: Addition of cell-penetrating sequences (like TAT) and fluorescent tags for tracking in vivo distribution
Two particularly effective biophysical techniques for quantifying CDK5-inhibitor interactions are:
Principle: Measures the directed movement of molecules along microscopic temperature gradients and detects binding-induced changes
Implementation: CDK5/p25 protein is labeled with fluorescent dye (NT647), mixed with inhibitor dilutions, and analyzed using Monolith NT.115 instrument
Advantages: Requires small sample volumes, measures interactions in solution without immobilization, suitable for protein-peptide interactions
Data output: Determines binding affinity (Kd) from concentration-dependent changes in thermophoresis
Principle: Measures changes in light interference patterns as molecules bind to biosensors, allowing real-time monitoring
Implementation: Biotinylated CDK5/p25 is immobilized on streptavidin sensors, exposed to inhibitor concentrations, and binding kinetics measured through association/dissociation phases
Advantages: Label-free detection for the analyte, real-time kinetic measurements (both ka and kd), high-throughput capability
Data output: Provides binding constants (ka, kd, KD) through kinetic analysis
For CDK5 inhibitors specifically, these techniques have been successfully used to demonstrate the selectivity of inhibitors like Cdk5i peptide, showing its substantially higher affinity for CDK5/p25 compared to CDK5 alone or CDK2 .
Evaluating CDK5's role in neurodegeneration employs multiple complementary approaches:
Transgenic mice overexpressing p25 to model CDK5 hyperactivation
Non-cleavable p35 mutant (Δp35KI) animals that prevent pathological p25 formation
Conditional CDK5 knockout models to assess loss-of-function effects
Testing CDK5 inhibitors in these models to assess therapeutic potential
Analysis of brain lysates from disease models versus wild-type to assess CDK5 complex formation
In vitro kinase assays to determine the effect of CDK5 modulators on substrate phosphorylation
Analysis of phosphorylation status of CDK5 substrates like tau in brain tissue
Discrimination between effects on CDK5/p25 (pathological) versus CDK5/p35 (physiological) complexes
Assessment of synaptic and cognitive functions after CDK5 modulation
Investigation of DNA damage as a pathological component related to CDK5 dysregulation
The most informative studies combine multiple approaches, such as demonstrating that the Cdk5i peptide inhibits CDK5/p25 activity in vitro and affects Cdk5 activity in vivo specifically in Tau P301S brain lysates but not in wild-type mice where CDK5/p25 levels are expected to be very low .
When working with recombinant CDK5/p25 for in vitro studies, several critical parameters require careful control:
Purity verification: Ensure >90% purity using SDS-PAGE or other appropriate methods
Activity confirmation: Validate kinase activity with standard substrates before experimental use
Storage conditions: Optimize to maintain stability and avoid repeated freeze-thaw cycles
Buffer optimization: Typically containing 50 mM Tris pH 7.4, 150 mM NaCl, 10 mM MgCl₂, and 0.01% Brij-35 for optimal activity
ATP concentration: Use physiologically relevant concentrations for kinase assays
Temperature effects: Human proteins may have different activities at 37°C versus room temperature
Substrate selection: Choose substrates relevant to research questions (tau peptides, histone H1, etc.)
Specificity controls: Include testing against other kinases (e.g., ERK1, GSK3β) to confirm selectivity
Comparative analysis: Test both CDK5/p25 and CDK5/p35 to distinguish pathological from physiological activity
Reference standards: Use established inhibitors as positive controls
Dose-response relationships: Test multiple concentrations to establish EC50/IC50 values
Statistical validation: Ensure sufficient replicates for reliable analysis
Normalization approaches: Use appropriate internal standards for quantitative comparisons
These parameters are essential for generating reproducible and physiologically relevant results when studying CDK5 activity and inhibition.
Distinguishing CDK5-specific effects from other proline-directed kinases (like GSK3β, ERK1, and other CDKs) requires multiple strategic approaches:
Inhibitor specificity testing: Evaluate inhibitors against panels of related kinases - the most promising CDK5 inhibitors show minimal activity against ERK1 and GSK3β while strongly inhibiting CDK5
Substrate specificity analysis: While CDK5 shares the S/T-P motif with other proline-directed kinases, certain substrates or phosphorylation sites show preference for CDK5
Complex-specific targeting: Target the unique CDK5/p25 or CDK5/p35 complexes rather than the catalytic domain shared with other kinases
Genetic manipulation: Use CDK5 knockout/knockdown models or dominate negative constructs alongside pharmacological inhibition
Activator manipulation: Target p35/p25 (unique to CDK5) rather than CDK5 itself to achieve specificity
Comparative pharmacology: Use multiple inhibitors with different selectivity profiles to triangulate CDK5-specific effects
Phosphoproteomics: Compare phosphorylation patterns after specific CDK5 inhibition versus inhibition of other proline-directed kinases
Temporal dynamics: Exploit differences in the timing of activation between different kinases
Spatial localization: Utilize the distinct subcellular localization patterns of CDK5/p35 versus CDK5/p25 versus other kinases
The p5 peptide studies demonstrate this approach well - they showed strong inhibition of CDK5 but not the related proline-directed kinases ERK1 and GSK3β, demonstrating that targeting specific elements of the CDK5-activator interaction can achieve the selectivity needed for biological studies .
Several innovative therapeutic strategies are being developed to selectively target pathological CDK5/p25 activity while preserving physiological CDK5/p35 function:
Optimized inhibitory peptides: The p5-MT peptide (with A/V substitution at the C-terminus) showed enhanced inhibitory potency compared to the original p5 peptide
Minimalist designs: The 12-amino acid Cdk5i peptide demonstrated >40-fold stronger binding to CDK5/p25 than to CDK5 alone, offering high specificity in a smaller package
Delivery enhancements: Addition of cell-penetrating sequences (TAT) and fluorescent tags improves in vivo delivery and monitoring
Non-cleavable p35: Studies with Δp35KI (non-cleavable mutant p35) demonstrated attenuated pathological features in mouse models of AD and FTD, providing proof-of-concept for preventing p25 generation
Alternative p25 prevention approaches: Targeting calpain or other mechanisms of p25 generation represents another strategy
Structure-guided design: Using the crystal structure of the Cdk5/p25 complex to design small molecules that specifically disrupt pathological interactions
High-throughput screening: Fluorescence polarization assays designed to identify small molecule inhibitors mimicking the effects of inhibitory peptides
Allosteric modulators: Targeting sites unique to the CDK5/p25 interaction rather than the conserved ATP-binding pocket
Dual-target strategies: Simultaneously targeting CDK5 hyperactivation and downstream consequences like tau hyperphosphorylation
Biomarker-guided therapy: Developing diagnostics to identify patients with CDK5 hyperactivation who would benefit most from targeted therapies
These approaches aim to address the limitations of current CDK5 inhibitors, which often lack specificity and may interfere with the essential physiological functions of CDK5/p35 complexes .
Advanced structural biology techniques offer promising avenues to enhance our understanding of CDK5 regulation and inhibition:
Capture CDK5 complexes in different conformational states to understand dynamic regulation
Visualize larger assemblies of CDK5 with multiple binding partners that may be difficult to crystallize
Observe structural changes upon inhibitor binding with minimal perturbation of natural states
Map protein dynamics and conformational changes upon p35 versus p25 binding
Identify regions with differential solvent accessibility in active versus inhibited states
Characterize allosteric networks connecting inhibitor binding sites to the active site
Combine X-ray crystallography, NMR, SAXS, and computational modeling to build complete models
Incorporate molecular dynamics simulations to understand conformational flexibility
Use cross-linking mass spectrometry to identify interaction interfaces in complex assemblies
Fragment-based approaches targeting unique pockets at the CDK5-p25 interface
Computational screening of virtual libraries against multiple conformational states
Application of machine learning to predict effective inhibitor modifications based on structural data
Utilize FRET-based biosensors to monitor CDK5 conformational changes in living cells
Apply cellular thermal shift assays (CETSA) to evaluate target engagement in cellular environments
Develop methods for structural analysis of CDK5 complexes directly in neurons
These approaches could help identify specific structural elements that distinguish CDK5/p25 from CDK5/p35 complexes, providing crucial insights for developing highly selective therapeutic agents that disrupt pathological hyperactivation while preserving essential physiological functions .
Cyclin-Dependent Kinase 5 (CDK5) is a proline-directed serine/threonine protein kinase that belongs to the cyclin-dependent kinase family. Unlike other members of this family, CDK5 does not directly control cell cycle regulation. Instead, it plays a crucial role in the nervous system, particularly in the development and function of neurons .
CDK5 was first discovered due to its sequence homology to the human cell division cycle protein 2 (Cdc2, also known as CDK1), a regulator of cell cycle progression . Despite its similarity to other cyclin-dependent kinases, CDK5’s activity is primarily observed in post-mitotic neurons, where it is essential for neuronal cell cycle arrest and differentiation .
CDK5 requires association with a regulatory subunit to become active. Unlike other CDKs that associate with cyclins, CDK5 is activated by binding to non-cyclin proteins such as p35 and p39 . These regulatory subunits are primarily expressed in the brain and are crucial for the kinase activity of CDK5 .
CDK5 is involved in various cellular processes in neurons, including:
Recombinant CDK5 is produced using the Sf9 insect cell expression system. This system is widely used for the production of recombinant proteins due to its ability to perform post-translational modifications similar to those in mammalian cells . The recombinant CDK5 produced in Sf9 cells retains its kinase activity and can be used for various research applications, including studying its role in neuronal functions and neurodegenerative diseases .