AURKA regulates G2/M-phase transition, spindle assembly, and cytokinesis . Dysregulation causes:
Centrosome aberrations: Aneuploidy and chromosomal instability linked to colorectal and ovarian cancers .
Oncogenic signaling: Overexpression activates mTOR/p70S6K/4E-BP1 pathways, promoting tumor proliferation .
AURKA is a synthetic lethal target in cancers with SWI/SNF complex mutations (e.g., ARID1A, SMARCA4) . Key inhibitors include:
BRD-7880: Binds AURKA’s kinase domain (PDB ID: 6GRA), disrupting ATP binding .
Lysine-reactive probes: Covalently target catalytic lysines (e.g., F8Z), enhancing drug residence time .
High AURKA expression correlates with immunosuppressive tumor microenvironments:
Neuroblastoma: Reduced immune/stromal scores, decreased cytokine signaling (e.g., chemokines, interferons) .
Ewing’s Sarcoma: Attenuated immune infiltration linked to YAP1-mediated apoptosis resistance .
Biomarker potential: IHC staining in NB and ES predicts MYCN amplification, INSS stage, and COG risk .
Drug development: Co-crystallization studies (e.g., 8JF4, 6GRA) inform structure-based inhibitor design .
AURKA (also known as Aurora-2/BTAK/STK15) is a serine/threonine kinase that regulates multiple cell cycle events occurring from late S-phase through the M phase. Its primary functions include centrosome maturation, mitotic entry, centrosome separation, bipolar spindle assembly, chromosome alignment, cytokinesis, and mitotic exit .
AURKA activity and protein levels both increase from late G2 through the M phase, with peak activity in pro-metaphase . Beyond its well-established mitotic roles, more recent research has identified additional non-mitotic functions during interphase, including roles in neurite elongation and ciliary resorption .
AURKA regulation follows a complex pattern of activation and inactivation through multiple mechanisms:
Phosphorylation activation: AURKA is activated through phosphorylation of Threonine 288 (T288) on its activation loop .
Co-factor dependent activation: AURKA binds to and phosphorylates the LIM domain-containing Ajuba protein during G2 phase, resulting in autophosphorylation of Aurora-A in its activating loop .
TPX2-mediated protection: The microtubule-associated protein TPX2, when released from importins by GTPase Ran, binds to AURKA and protects it from inactivation by protein phosphatase 1 (PP1) .
Inactivation mechanism: AURKA is inactivated through dephosphorylation of T288 by protein phosphatase 1 (PP1) or protein phosphatase 2A (PP2A) .
The mode of binding between TPX2 and AURKA induces conformational changes that resemble the intramolecular binding and activation of cAMP-dependent kinase .
AURKA's structure consists of distinct domains that contribute to its function and regulation:
N-terminal intrinsically disordered region (IDR): Contains the A-box degron motif that is critical for FZR1-dependent mitotic degradation .
Kinase domain: Contains the activation loop with the critical T288 phosphorylation site required for kinase activity .
C-terminal domain: Previously thought to contain a D-box degron, but recent research indicates it instead mediates essential structural features of the protein rather than acting as a true degron .
The full-length AURKA's degradation depends on both an intact N-terminal A-box region and proper conformational parameters provided by the C-terminal domain .
Recent research has revised our understanding of AURKA degradation mechanisms:
APC/C^FZR1-mediated degradation: AURKA destruction is critically dependent on the anaphase-promoting complex (APC/C^FZR1) during mitotic exit and G1 phase .
A-box degron function: Contrary to earlier models, the N-terminal intrinsically disordered region containing the A-box is sufficient to confer FZR1-dependent mitotic degradation, while the C-terminal domain does not function as a canonical D-box degron .
QRVL motif: Both computational prediction and cellular assays suggest that the QRVL short linear interacting motif (SLiM) within the A-box functions as a phospho-regulated D-box .
Conformational requirements: Degradation of full-length AURKA depends on an intact C-terminal domain because it provides critical conformational parameters that permit both activity and mitotic degradation .
This updated model explains why earlier studies identified both N- and C-terminal regions as important for degradation, even though only the N-terminal region contains a true degron motif.
AURKA's activity is highly dependent on protein-protein interactions that affect its localization, activation state, and substrate selection:
TPX2 activation mechanism: TPX2 binding has no effect on the turnover number of AURKA and does not change its reaction mechanism, but rather protects it from dephosphorylation by PP1 . This binding triggers conformational changes in AURKA.
Targeting to spindle poles: The interaction with TPX2 targets AURKA to spindle microtubules at the poles, ensuring proper spatial regulation .
Synergistic activation: In vivo, AURKA activation synergistically depends on (auto)phosphorylation within its activation segment (on T288) and TPX2 binding, potentially in combination with microtubule binding .
Substrate specificity: Different cofactors may alter AURKA's substrate specificity, allowing it to function in multiple cellular contexts and pathways.
Understanding these cofactor relationships is critical for developing more specific inhibitors that could target particular AURKA functions while sparing others.
AURKA interacts with tumor suppressor pathways through multiple mechanisms:
p53 regulation: AURKA interferes with p53 suppressor function through at least three mechanisms:
TAp73 suppression: AURKA overexpression suppresses TAp73 in p53-deficient cancer cells. TAp73 has significant homology with p53 and plays an essential role in apoptosis induced by cytotoxic agents .
Pro-survival pathway activation: AURKA up-regulates AKT phosphorylation at Ser473, promoting cell survival and preventing apoptosis .
GSK-3β and β-catenin regulation: AURKA regulates GSK-3β and β-catenin in gastric cancer cells, potentially affecting Wnt signaling .
These interactions create a network where AURKA overexpression can simultaneously suppress multiple tumor suppressor functions while activating pro-survival pathways.
AURKA's role in cancer development involves multiple mechanisms:
Oncogenic transformation: AURKA activation can transform rodent fibroblast cells, establishing it as a bona fide oncogene .
Genomic instability: AURKA overexpression leads to formation of multipolar mitotic spindles, inducing genome instability .
Aneuploidy induction: A correlation exists between AURKA overexpression and aneuploidy in gastric cancer; clinical samples with AURKA amplification and overexpression showed aneuploidy and poor prognosis .
Centrosome abnormalities: AURKA plays an important role in centrosome maturation, and centrosomal anomalies arise at early stages of tumor formation and expand with tumor progression .
Cell survival promotion: Through regulation of AKT, AURKA supports cancer cell survival and prevents apoptosis .
Checkpoint override: AURKA has been reported to override the spindle checkpoint activated by paclitaxel (Taxol) and nocodazole, contributing to drug resistance .
AURKA gene amplification and/or overexpression patterns vary across cancer types:
Cancer Type | AURKA Alteration | Associated Features | Prognosis Correlation |
---|---|---|---|
Breast | Amplification/overexpression | Centrosome abnormalities | Poor prognosis |
Colon | Amplification/overexpression | Aneuploidy | Poor prognosis |
Pancreas | Amplification/overexpression | Not specified | Poor prognosis |
Ovarian | Amplification/overexpression | Telomerase regulation | Poor prognosis |
Bladder | Amplification/overexpression | Not specified | Poor prognosis |
Liver | Amplification/overexpression | Not specified | Poor prognosis |
Gastric | Amplification/overexpression | Aneuploidy | Poor prognosis |
Brain | Not specified | Centrosomal abnormalities | Not specified |
Prostate | Not specified | Centrosomal abnormalities | Not specified |
Lung | Not specified | Centrosomal abnormalities | Not specified |
AURKA overexpression has been reported to be significantly associated with higher grade tumors and poor prognosis across multiple cancer types .
When analyzing AURKA in tumor contexts, researchers should consider multiple complementary approaches:
Gene amplification analysis: FISH (Fluorescence In Situ Hybridization) to detect AURKA gene copy number changes at chromosome 20q13.2 .
Expression analysis:
RT-qPCR for mRNA expression levels
Immunohistochemistry (IHC) for protein expression and localization in tissue samples
Western blotting for total protein levels in cell lysates
Activity assessment:
Phospho-specific antibodies to detect activated AURKA (pT288)
Kinase activity assays using immunoprecipitated AURKA
Evaluation of phosphorylation status of downstream targets
Functional analysis:
Centrosome counting to assess centrosome amplification
Spindle morphology analysis
Chromosome segregation error assessment
Aneuploidy measurement through karyotyping or flow cytometry
Correlating these measurements with clinical outcomes and tumor characteristics provides the most comprehensive picture of AURKA's role in a specific cancer context.
Optimal approaches for studying AURKA kinase activity include:
Recombinant protein production: Expression and purification of wild-type and mutant AURKA proteins in bacterial or insect cell systems.
Kinase assays: Using substrate peptides containing the consensus motif (R/K)X(S/T)(I/L/V) and measuring phosphorylation through:
Radioactive ATP incorporation
Phospho-specific antibody detection
Mass spectrometry analysis
Co-factor effects: Testing activity with and without TPX2 or other known binding partners to assess co-factor dependence.
FRET-based biosensors: For real-time monitoring of AURKA activity in living cells.
Phospho-specific antibodies: Detecting T288 phosphorylation or substrate phosphorylation by immunofluorescence or western blotting.
Genetic manipulation:
Conditional knockout models
Expression of kinase-dead mutants
RNAi-mediated knockdown
CRISPR/Cas9 genome editing
Drug inhibition studies: Using specific AURKA inhibitors with appropriate controls to distinguish from effects on related kinases.
These approaches should be combined for comprehensive characterization of AURKA activity in different biological contexts.
Distinguishing between AURKA and AURKB functions requires careful experimental design:
Subcellular localization analysis:
AURKA primarily localizes to centrosomes and spindle poles
AURKB is a chromosomal passenger protein with dynamic localization
Specific inhibitors:
MLN8054 and MLN8237 show higher specificity for AURKA
ZM-447439 shows greater specificity for AURKB
Titration studies with concentration ranges that selectively inhibit one kinase
Genetic approaches:
Selective knockdown with validated siRNAs targeting unique regions
Rescue experiments with kinase-specific mutants resistant to siRNA
CRISPR/Cas9 genome editing with careful validation
Substrate analysis:
Focus on known specific substrates (e.g., TACC3 for AURKA)
Evaluate localization-dependent phosphorylation events
Timing analysis:
Examine effects during specific cell cycle phases when one kinase is predominantly active
Researchers should always use multiple approaches and appropriate controls to confirm kinase-specific effects.
When investigating AURKA degradation, researchers should consider:
Cell cycle synchronization:
Methods to precisely arrest cells at specific phases
Time-course analysis following release from synchronization
Protein stability measurements:
Cycloheximide chase assays with western blot analysis
Pulse-chase experiments with metabolic labeling
Live-cell imaging with fluorescently tagged AURKA
Degron analysis:
Mutation of key residues in the A-box region (especially the QRVL motif)
Chimeric proteins with degron transfers to stable proteins
Structure-function analysis of the C-terminal domain
APC/C^FZR1 manipulation:
FZR1 knockdown or knockout approaches
APC/C inhibitor studies (e.g., proTAME)
Co-immunoprecipitation to assess physical interaction
Phosphorylation state:
Analysis of phospho-regulation of the A-box
Phosphomimetic and phospho-resistant mutations
Kinase and phosphatase inhibitor treatments
The study should account for the recent finding that the N-terminal IDR containing the A-box is sufficient for FZR1-dependent mitotic degradation, while the C-terminal domain provides essential structural features rather than acting as a canonical degron .
Several AURKA inhibitors have been developed with varying specificity profiles:
Inhibitor | Specificity | IC50 for AURKA | Mechanism | Stage of Development | Key Features |
---|---|---|---|---|---|
MLN8054 | AURKA > AURKB | ~4-8 nM | ATP-competitive | Preclinical/Early clinical | Higher specificity for AURKA |
MLN8237 (Alisertib) | AURKA > AURKB | ~1.2 nM | ATP-competitive | Clinical trials | Improved pharmacokinetics over MLN8054 |
VX-680 (MK-0457) | Pan-Aurora | ~0.6 nM | ATP-competitive | Clinical trials | Also inhibits FLT3 |
ZM-447439 | AURKB > AURKA | ~110 nM | ATP-competitive | Preclinical | Primarily AURKB inhibitor |
Hesperadin | AURKB > AURKA | ~250 nM | ATP-competitive | Preclinical | Higher selectivity for AURKB |
When selecting an inhibitor for research purposes, consider:
The desired specificity for AURKA vs. AURKB
Off-target effects on other kinases
Compatibility with your experimental system
Dose-dependent effects that may change selectivity
To investigate and overcome AURKA inhibitor resistance:
Resistance mechanism characterization:
Genomic analysis for mutations in the AURKA ATP-binding pocket
Phosphoproteomics to identify compensatory signaling pathways
Expression analysis of AURKA binding partners that may affect inhibitor efficacy
Combination strategies:
Target parallel pathways that may compensate for AURKA inhibition
Combine with cell cycle checkpoint inhibitors
Pair with inhibitors of AURKA-interacting proteins
Alternative inhibition approaches:
Protein-protein interaction disruptors targeting AURKA-TPX2 binding
Degradation-inducing chimeric molecules (PROTACs) targeting AURKA
Allosteric inhibitors targeting sites outside the ATP-binding pocket
Genetic verification:
CRISPR/Cas9 validation of AURKA dependency in resistant models
Overexpression of wild-type vs. mutant AURKA to confirm mechanism
Researchers should design resistance studies with clinically relevant concentrations and exposure times to maximize translational relevance.
A comprehensive evaluation of AURKA inhibitor efficacy should include:
Biochemical confirmation:
In vitro kinase assays with purified AURKA protein
Cellular target engagement assays (e.g., cellular thermal shift assay)
Phosphorylation status of direct AURKA substrates
Phenotypic validation:
Mitotic index measurements
Spindle pole defect quantification
Mitotic delay assessment by time-lapse microscopy
Cell cycle profile analysis by flow cytometry
Efficacy in cellular models:
Proliferation/viability assays in cancer cell line panels
Colony formation assays for long-term effects
3D spheroid growth inhibition
Apoptosis measurements
In vivo evaluation:
Pharmacokinetic and pharmacodynamic relationship
Tumor growth inhibition in xenograft models
Patient-derived xenograft responses
Biomarker modulation correlating with efficacy
Attention should be paid to dose scheduling, potential off-target effects, and biomarkers of response that could translate to clinical applications.
Emerging non-mitotic functions of AURKA represent exciting research frontiers:
Neurite elongation and neural development:
AURKA's role in microtubule dynamics during neurite outgrowth
Potential implications for neurodevelopmental disorders
Therapeutic relevance for neurodegenerative conditions
Ciliary resorption and ciliopathies:
Mechanisms of AURKA activation in ciliary disassembly
Connection to ciliopathies and related disorders
Drug targeting potential for ciliary dysfunction diseases
Metabolic regulation:
AURKA's potential roles in controlling cellular metabolism
Links to metabolic reprogramming in cancer
Relationship with nutrient sensing pathways
DNA damage response:
Emerging functions in DNA repair mechanisms
Synthetic lethal interactions with DNA damage response pathways
Implications for combination therapies with DNA-damaging agents
Immune regulation:
Potential roles in immune cell function
Implications for immuno-oncology approaches
Cross-talk with inflammatory signaling pathways
Investigating these non-canonical functions may reveal new therapeutic approaches and explain clinical observations not accounted for by mitotic-only models .
Advanced structural approaches offer significant potential to resolve remaining questions about AURKA:
Cryo-electron microscopy:
Structure determination of full-length AURKA including the IDR regions
Visualization of AURKA in complex with mitotic regulators
Structural basis for multiple active conformations
Hydrogen-deuterium exchange mass spectrometry:
Mapping conformational changes upon activation
Identification of allosteric regulatory sites
Characterization of protein-protein interaction interfaces
Single-molecule FRET:
Real-time analysis of AURKA conformational dynamics
Effects of phosphorylation on structural transitions
Influence of binding partners on conformational ensemble
Molecular dynamics simulations:
Prediction of conformational changes upon binding
Analysis of intrinsically disordered region dynamics
Virtual screening for novel binding sites
AlphaFold and related AI approaches:
Prediction of full-length AURKA structure including disordered regions
Modeling of protein-protein complexes
Structure-based drug design for novel inhibitors
These approaches could reveal how AURKA's multiple active conformations relate to its diverse functions and guide more specific therapeutic targeting strategies .
The development of predictive biomarkers for AURKA inhibitor response faces several challenges and opportunities:
Functional redundancy: Aurora kinases have overlapping functions that may compensate for targeted inhibition.
Context-dependent roles: AURKA's functions vary across tissue types and genetic backgrounds.
Multiple conformations: Different active conformations of AURKA may respond differently to inhibitors.
Technical limitations: Measuring kinase activity directly in patient samples is challenging.
Genetic biomarkers:
AURKA amplification status as a stratification factor
Co-occurring genetic alterations that sensitize to AURKA inhibition
Synthetic lethal genetic interactions
Protein biomarkers:
AURKA expression levels by IHC
Expression of critical cofactors like TPX2
Phosphorylation status of downstream targets
Functional assays:
Ex vivo drug sensitivity testing of patient-derived cells
Organoid-based response prediction
Phosphoproteomics signatures of AURKA pathway activation
Imaging biomarkers:
Centrosome abnormalities quantification
Mitotic index in pre- and post-treatment biopsies
PET imaging with selective AURKA inhibitor radiotracers
Integrating multiple biomarker approaches into early-phase clinical trials would accelerate clinical development and patient selection strategies.
Aurora Kinase A (AURKA) is a serine/threonine kinase that plays a crucial role in cell division by regulating various processes during mitosis. It is a member of the Aurora kinase family, which also includes Aurora Kinase B and C. The recombinant form of Aurora Kinase A is often used in research to study its function and potential as a therapeutic target.
Aurora Kinase A is encoded by the AURKA gene located on chromosome 20q13.2. The protein is approximately 403 amino acids long and has a molecular weight of around 46 kDa. It is predominantly expressed in tissues with high mitotic activity, such as the testes and thymus, and its expression is tightly regulated during the cell cycle .
Aurora Kinase A is essential for several key processes during mitosis:
Aurora Kinase A exerts its effects by phosphorylating various substrates involved in mitosis. One of its well-known substrates is the microtubule-associated protein TPX2, which is essential for spindle assembly. The kinase activity of Aurora Kinase A is regulated by autophosphorylation and interaction with other proteins, such as Bora and Ajuba .
Aurora Kinase A has been implicated in cancer development and progression. Overexpression of AURKA is often observed in various cancers, including breast, colorectal, and pancreatic cancers. This overexpression is associated with poor prognosis and increased tumor aggressiveness. As a result, Aurora Kinase A is considered a potential therapeutic target, and several inhibitors are currently being developed and tested in clinical trials.
Recombinant Aurora Kinase A is produced using various expression systems, such as baculovirus-insect cells or E. coli. The recombinant protein is used in biochemical assays to study its kinase activity, substrate specificity, and inhibition by potential therapeutic compounds. It is also used in structural studies to understand the molecular basis of its function and regulation.