Aurora Kinase B (AURKB) is a serine/threonine kinase critical for mitotic regulation, including chromosome alignment, spindle assembly checkpoint control, and cytokinesis. It is encoded by the AURKB gene (ENSG00000178999) on human chromosome 17 and plays a key role in maintaining genomic stability . Dysregulation of AURKB is strongly linked to cancer progression, making it a prominent therapeutic target .
Domain architecture: The kinase domain (residues 55–344) forms a dimeric structure when bound to its regulatory partner INCENP (inner centromere protein) .
Activation loop dynamics: A unique domain swap in the activation loop (residues 227–245) enables dimerization, altering the DFG motif (Asp-Phe-Gly) conformation compared to Aurora A or Xenopus laevis Aurora B .
Key interactions: Hydrophobic residues (Y239, L237, Pro242) stabilize the dimer interface (buried surface area: 3,460 Ų) .
AURKB phosphorylates histone H3 at Ser10 (H3S10ph) to promote chromosome condensation and regulates:
Overexpression: Observed in retinoblastoma (RB), rhabdomyosarcoma (RMS), and renal cell carcinoma (KIRC), correlating with poor prognosis .
Mechanisms: Drives genomic instability by inducing aneuploidy and suppressing apoptosis .
RB: AURKB inhibition reduced cell viability by 60% in primary tumors .
RMS: AZD1152 enhanced vincristine efficacy, reducing tumor growth by 70% in xenografts .
Expression system: Escherichia coli with His-tag purification .
Protein specifications: 41.4 kDa, 364 amino acids (residues 1–344), >90% purity by SDS-PAGE .
Applications: Structural studies, kinase activity assays, and inhibitor screening .
Immune pathways: AURKB correlates with tumor mutational burden (TMB) in 18 cancers and microsatellite instability (MSI) in 7 cancers .
miRNA regulation: Targeted by hsa-let-7e-5p and hsa-miR-5088-3p, potential biomarkers for therapy .
AURKB functions as the catalytic component of the Chromosomal Passenger Complex (CPC), which orchestrates key mitotic events. Methodologically, researchers studying AURKB function should employ:
Time-lapse microscopy with fluorescently-tagged AURKB to track dynamic localization (chromosomes in prophase → centromeres in metaphase → central spindle in anaphase → midbody during cytokinesis)
Phospho-specific antibodies against validated substrates (particularly Histone H3 at Ser10)
Selective inhibitors (such as AZD1152/Barasertib) with appropriate controls to dissect AURKB-specific functions from other Aurora kinases
Co-immunoprecipitation studies to analyze interactions with other CPC components (INCENP, Survivin, Borealin)
Cell-type specific expression patterns should be considered when studying AURKB function, as its expression correlates with proliferative capacity, showing highest levels in rapidly dividing cells and tissues.
Robust detection of AURKB requires complementary approaches:
Immunostaining: Co-stain with cell-type specific markers (as seen with AURKB+/cTnT+ cardiomyocytes in cardiac research)
Flow cytometry: For quantitative assessment across large cell populations
Western blotting: Use validated antibodies with appropriate positive controls (mitotic cell extracts)
mRNA detection: RT-qPCR with intron-spanning primers or RNA-seq analysis
For accurate quantification of AURKB+ cells, researchers should:
Include cell cycle markers to differentiate proliferating cells
Perform statistical analysis across multiple tissue sections
Use automated imaging software to reduce observer bias
Validate findings with multiple detection methods
In cardiac research, increased percentages of AURKB+ cardiomyocytes were detected in specific experimental conditions, such as Mettl3-deficient neonatal mice, indicating enhanced proliferative capacity .
AURKB phosphorylates numerous substrates critical for mitotic progression. Methodologically, researchers should:
Validation approaches:
In vitro kinase assays with purified components
Phospho-specific antibodies to monitor modification status
Site-directed mutagenesis (phospho-mimetic/dead mutations)
Functional assays to assess phenotypic consequences
Key validated substrates include:
Histone H3 (Ser10, Ser28): Controls chromosome condensation
CENP-A (Ser7): Regulates kinetochore assembly
MCAK: Controls microtubule depolymerase activity
MgcRacGAP: Regulates cytokinesis completion
Vimentin: Coordinates intermediate filament dynamics during division
For comprehensive substrate identification, researchers should combine candidate approaches with unbiased phosphoproteomic screens comparing wild-type versus AURKB-inhibited conditions.
AURKB expression and activity are tightly regulated throughout the cell cycle. To properly study this:
Synchronization techniques: Use double thymidine block, nocodazole arrest, or mitotic shake-off to obtain cell populations at specific cycle phases
Live-cell imaging: Employ fluorescent reporters to track AURKB expression/localization in real-time
Western blotting: Analyze protein levels across synchronized populations
Flow cytometry: Combine with DNA content analysis to correlate expression with cell cycle phase
Key findings include:
Low expression in G1 phase
Increasing levels through S phase
Peak expression during G2/M transition
Protein degradation after mitotic exit via APC/C-mediated ubiquitination
Researchers should distinguish between mRNA expression, protein abundance, and kinase activity, as these parameters may not directly correlate throughout the cell cycle.
Researchers have multiple options for modulating AURKB function:
Chemical tools:
Small molecule inhibitors: AZD1152/Barasertib (AURKB-selective), VX-680/MK-0457 (pan-Aurora)
Degraders: PROTACs targeting AURKB for proteasomal degradation
Activity-based probes: For monitoring AURKB activity in situ
Genetic approaches:
siRNA/shRNA: For transient or stable knockdown
CRISPR-Cas9: For knockout or endogenous tagging
Overexpression systems: Wild-type or mutant variants
Advanced technologies:
Analog-sensitive alleles: For selective inhibition of engineered AURKB
Optogenetic tools: Light-inducible activation/inhibition
Degron systems: For rapid protein depletion
When selecting tools, researchers should consider temporal dynamics, as complete AURKB inhibition may prevent mitosis, complicating interpretation of results.
Distinguishing AURKB from AURKA and AURKC remains challenging due to sequence similarity. Methodological approaches include:
Pharmacological strategies:
Careful titration of inhibitor concentrations
Use of selective inhibitors with verification of target engagement
Correlation with substrate phosphorylation patterns
Genetic approaches:
Selective knockdown with validation of specificity
Rescue experiments with inhibitor-resistant mutants
CRISPR knockout with complementation studies
Localization-based discrimination:
AURKB: Chromosomes → centromeres → midbody
AURKA: Centrosomes → spindle poles
AURKC: Primarily in meiotic cells
Substrate specificity:
AURKB preferentially phosphorylates H3S10, CENP-A
AURKA primarily targets TPX2, TACC3
Researchers should employ multiple complementary approaches and include appropriate controls to confidently attribute observed effects to specific Aurora kinases.
The search results indicate AURKB serves as a marker for proliferating cardiomyocytes during heart regeneration . To investigate this:
Detection strategies:
Co-immunostaining for AURKB and cardiac markers (cTnT)
Quantification of AURKB+ cardiomyocytes in different experimental conditions
Correlation with other proliferation markers (pH3, Ki67)
Functional approaches:
Cardiac-specific AURKB manipulation (knockout/overexpression)
Administration of selective inhibitors in regeneration models
Lineage tracing of AURKB+ cardiomyocytes during regeneration
Molecular mechanism investigation:
Analysis of AURKB-mediated phosphorylation events in cardiomyocytes
Integration with regulatory pathways (such as Mettl3-mediated m6A modification)
Identification of cardiomyocyte-specific AURKB substrates
Research has shown increased percentages of AURKB+ cardiomyocytes in Mettl3-deficient neonatal mice and in models with mutated m6A consensus sequences in Fgf16, corresponding with enhanced regenerative capacity .
AURKB aberrations promote chromosomal instability through multiple mechanisms. Methodological approaches include:
Quantifying chromosomal instability parameters:
Micronuclei formation assays
Fluorescence in situ hybridization for aneuploidy detection
Live-cell imaging of chromosome segregation errors
Single-cell sequencing for copy number variation analysis
Mechanistic investigations:
Assessment of kinetochore-microtubule attachment stability
Measurement of spindle assembly checkpoint function
Analysis of sister chromatid cohesion timing
Quantification of cytokinesis failure rates
Expression modulation studies:
Titrated overexpression to determine threshold effects
Dominant-negative approaches to disrupt function
Correlation of expression levels with CIN markers in patient samples
AURKB overexpression can override spindle assembly checkpoint function, while reduced activity impairs error correction mechanisms. Both scenarios promote genomic instability, highlighting the requirement for precise AURKB regulation.
State-of-the-art phosphoproteomic approaches include:
Experimental design considerations:
Acute vs. sustained AURKB inhibition comparisons
Cell synchronization to capture mitotic phosphorylation events
Subcellular fractionation to enrich for relevant compartments
Advanced techniques:
Multiplexed quantitative phosphoproteomics (TMT, iTRAQ)
Complementary enrichment strategies (IMAC, TiO2, phospho-antibodies)
Data-independent acquisition mass spectrometry
Parallel reaction monitoring for targeted analysis
Bioinformatic analysis:
Motif analysis for AURKB consensus sequences (R/K-X-S/T)
Integration with protein-protein interaction networks
Kinase-substrate enrichment analysis (KSEA)
Temporal clustering of phosphorylation events
Validation approaches:
In vitro kinase assays with purified components
Phospho-specific antibodies for key substrates
Mutational analysis of identified phosphosites
Correlation with AURKB localization patterns
Integration of phosphoproteomic data with other omics approaches provides comprehensive insights into AURKB-regulated processes across different cellular contexts.
CRISPR-Cas9 optimization for AURKB studies requires:
Guide RNA design considerations:
Target functional domains (kinase domain, activation loop)
Use algorithms that minimize off-target effects
Design multiple guides and validate independently
Consider exon essentiality and potential for compensatory splicing
Editing strategies:
Complete knockout vs. domain-specific mutations
Homology-directed repair for precise modifications
Endogenous tagging for visualization/purification
Base or prime editing for specific amino acid changes
Temporal control systems:
Inducible Cas9 expression
Conditional guide RNA expression
Degron-tagging for rapid protein depletion
Auxin-inducible degron systems
Validation requirements:
Deep sequencing to confirm edits and assess off-targets
Rescue experiments with wild-type or mutant AURKB
Phenotypic analysis across multiple independent clones
Evaluation of compensatory mechanisms
As AURKB is essential for cell division, researchers should implement conditional approaches rather than constitutive knockout strategies to study dynamic functions.
Developing selective AURKB inhibitors presents several challenges:
Structural similarity obstacles:
70% sequence identity in ATP-binding pockets across Aurora kinases
Limited structural differences for selective targeting
Shared binding modes with many kinase inhibitors
Selectivity assessment approaches:
Kinome-wide profiling against 300+ kinases
Cellular thermal shift assays to confirm target engagement
NanoBRET target engagement in live cells
Correlation of cellular effects with biochemical potency
Structure-based design strategies:
Fragment-based screening for unique binding modes
Targeting allosteric sites rather than ATP-binding pocket
Exploiting unique features in activation loop conformations
Development of covalent inhibitors for specific cysteines
Alternative approaches:
PROTAC-based degraders with AURKB-selective binding moieties
Targeting protein-protein interactions specific to AURKB
Substrate-competitive inhibitors
Conformation-selective inhibitors
Researchers must balance selectivity with pharmacokinetic properties and cellular penetration to develop effective tools for biological studies and potential therapeutic applications.
AURKB undergoes multiple post-translational modifications that regulate its function. Methodologically:
Key modifications to study:
Autophosphorylation at T232 (activation loop): Essential for kinase activity
Phosphorylation by CDK1 (S331): Primes for activation
Ubiquitination: Controls protein stability and degradation
SUMOylation: Affects CPC assembly and chromosome targeting
Acetylation: Influences chromatin interactions
Detection strategies:
Phospho-specific antibodies for known sites
Mass spectrometry for unbiased PTM mapping
Western blotting with mobility shift analysis
Immunoprecipitation with modification-specific antibodies
Functional analysis approaches:
Site-directed mutagenesis (phospho-mimetic/dead mutations)
In vitro reconstitution with modified components
Pharmacological inhibition of modifying enzymes
Proteomic analysis following perturbation of PTM pathways
Temporal dynamics assessment:
Synchronization methods to capture cell cycle-dependent changes
Live-cell imaging with PTM-specific biosensors
Rapid isolation techniques to preserve labile modifications
These approaches provide insights into how PTMs collectively regulate AURKB localization, activity, substrate specificity, and protein-protein interactions throughout the cell cycle.
Aurora Kinase B (AURKB) is a member of the Aurora kinase family, which plays a crucial role in cell division by regulating chromosome segregation and cytokinesis. This serine/threonine kinase is essential for maintaining genomic stability and is a key player in mitosis.
Aurora Kinase B is involved in several critical processes during mitosis:
Aurora Kinase B functions by phosphorylating various substrates involved in mitosis. One of its key targets is the mitotic centromere-associated kinesin (MCAK), which it phosphorylates on Ser92. This phosphorylation event inhibits MCAK’s microtubule depolymerizing activity, thereby stabilizing the spindle microtubules .
Overexpression of Aurora Kinase B has been linked to various cancers, as it can lead to aneuploidy and genomic instability. Consequently, AURKB is considered a potential target for anticancer therapies. Inhibitors of Aurora Kinase B are being explored as therapeutic agents to treat cancers characterized by high levels of this kinase .