Recombinant Mouse Protein Aurora Borealis, commonly referred to as Bora, is a crucial protein involved in the regulation of cell cycle progression, particularly during mitosis. Bora plays a pivotal role in activating Polo-like kinase 1 (Plk1) by interacting with Aurora A kinase, thereby facilitating mitotic entry and progression . This article will delve into the structure, function, and research findings related to Recombinant Mouse Protein Aurora Borealis (Bora).
Bora is an intrinsically disordered protein that lacks a defined three-dimensional structure under physiological conditions. Its flexibility allows it to interact with various proteins, including Aurora A and Plk1, facilitating their activation and subsequent roles in mitosis . Bora's interaction with Aurora A enhances the phosphorylation of Plk1 at its T-loop, which is essential for Plk1's activation .
Phosphorylation Sites: Bora is phosphorylated by Cyclin-Cdk at multiple sites, including a polo-docking site (S252), which enhances its binding to Plk1 .
Tpx2-like Motifs: Bora contains motifs similar to those found in Tpx2, which are crucial for its function in promoting mitotic entry .
Bora's primary role is to facilitate the activation of Plk1 by Aurora A, which is critical for mitotic entry and progression. The absence or malfunction of Bora can lead to defects in mitotic spindle formation and cell cycle progression .
Bora Phosphorylation: Bora is phosphorylated by Cyclin-Cdk, which enhances its ability to interact with Aurora A and Plk1.
Interaction with Aurora A: Phosphorylated Bora binds to Aurora A, promoting its activation and subsequent phosphorylation of Plk1.
Plk1 Activation: The interaction between Bora and Aurora A facilitates the phosphorylation of Plk1 at its T-loop, activating Plk1 and enabling mitotic progression .
Recent studies have highlighted the importance of Bora in mitotic regulation. For instance, mutations in Bora's Tpx2-like motifs (M1 and M2) impair its ability to support mitotic entry in Xenopus egg extracts . Additionally, Bora's phosphorylation by Cyclin-Cdk is essential for its function, as mutations at these sites significantly reduce its ability to rescue mitotic delays .
Xenopus Egg Extracts: Studies using Xenopus egg extracts have shown that Bora is crucial for mitotic entry, as its depletion prevents Plx1 activation by Aurora A .
Cell Culture Models: In mammalian cells, Bora mutants lead to mitotic defects similar to those observed when Aurora A function is impaired .
While specific data on Recombinant Mouse Protein Aurora Borealis (Bora) is limited, recombinant proteins are typically used in research to study protein function and interactions. Recombinant Bora would be expected to mimic the native protein's behavior, allowing researchers to investigate its role in mitosis and its interactions with other proteins like Aurora A and Plk1.
Cell Cycle Research: Recombinant Bora can be used to study cell cycle regulation and the mechanisms underlying mitotic entry.
Cancer Research: Given Bora's role in promoting mitotic progression, it may be implicated in cancer development, making it a potential target for therapeutic interventions.
| Feature | Description |
|---|---|
| Phosphorylation Sites | Multiple sites, including a polo-docking site (S252) |
| Tpx2-like Motifs | Essential for promoting mitotic entry |
| Interaction with Aurora A | Enhances Aurora A's ability to phosphorylate Plk1 |
| Role in Mitosis | Facilitates mitotic entry and progression by activating Plk1 |
Aurora Borealis (Bora) is a conserved mitotic protein that functions as a critical co-activator of Aurora A kinase and plays a key role in activating Polo-like kinase 1 (PLK1). First identified in Drosophila melanogaster, Bora controls the proper timing of mitosis onset by enabling Aurora A to activate PLK1 . In mammals, this protein is essential for cell cycle progression, particularly during the G2/M transition. Bora's activity is tightly regulated during the cell cycle, with its function being particularly crucial during mitotic entry. Mechanistically, Bora binds to Aurora A and enhances its kinase activity, which subsequently leads to proper centrosome maturation, spindle assembly, and asymmetric protein localization during mitosis .
Bora exhibits remarkable evolutionary conservation from C. elegans to humans, indicating its fundamental role in eukaryotic cell division . The functional domains of Bora, particularly those involved in Aurora A binding and activation, show high sequence similarity across species. Both Drosophila and human Bora can bind to Aurora-A and activate the kinase in vitro, demonstrating functional conservation . The conserved region in the N-terminal portion is critical for Aurora-A binding, while the non-conserved C-terminus appears dispensable for this interaction. This evolutionary conservation suggests that Bora-mediated regulation of Aurora A represents a fundamental mechanism in mitotic control that has been maintained throughout eukaryotic evolution.
Bora exhibits a dynamic subcellular localization pattern that changes throughout the cell cycle:
In interphase cells: Bora is predominantly a nuclear protein
Upon entry into mitosis: Bora is excluded from the nucleus and translocates to the cytoplasm
This localization pattern is functionally significant as it provides a spatial regulatory mechanism for Aurora A activation. During interphase, nuclear retention of Bora helps keep Aurora A inactive. When Cdc2 (CDK1) becomes activated at the G2/M transition, Bora is released into the cytoplasm where it can bind and activate Aurora A . This mechanism elegantly explains how sequential activation of Cdc2 leads to Aurora A activation, establishing a temporal coordination of mitotic events.
Bora serves as a critical intermediary in the activation cascade of PLK1 during mitotic entry through several coordinated mechanisms:
CDK1-mediated phosphorylation: Under recovery conditions from G2/M checkpoint, CDK1 (Cdc2) phosphorylates Bora on its N-terminal domain, which is essential for PLK1 re-activation and subsequent mitotic commitment .
Aurora A facilitation: Bora binds to Aurora A and enhances its kinase activity, which then phosphorylates PLK1 at threonine 210 in its activation loop.
Conformational changes: The binding of Bora to the kinase domain of Aurora A may induce conformational changes that facilitate PLK1 access to the Aurora A active site.
Cytoplasmic translocation: As Bora translocates from the nucleus to the cytoplasm during prophase (in a Cdc2-dependent manner), it enables spatial coordination of PLK1 activation at specific subcellular locations .
This multilayered activation mechanism ensures proper timing of PLK1 activity, which is crucial for centrosome maturation, bipolar spindle formation, and chromosome segregation during mitosis.
Effects of Bora depletion:
Cell cycle arrest or delay at G2/M transition
Reduced PLK1 activation and decreased phosphorylation of its downstream targets
Apoptosis-consistent pattern of condensed and/or fragmented chromatin
Reduction in the surrogate marker of PLK1 activity (pTCTP-Ser46)
Defects in asymmetric cell division similar to those observed in Aurora-A mutants
Impaired centrosome maturation and spindle assembly
Effects of Bora overexpression:
Increased capacity to form colonies in soft agar, indicating malignant transformation potential
Loss of contact inhibition, with cells forming multiple layers after reaching confluence
Accelerated migration capacity (approximately two-fold increase)
These opposing phenotypes highlight Bora's central role in regulating mitotic progression and cellular transformation, making it a potential target for anticancer therapeutic strategies.
For the production of functional recombinant mouse Bora protein, several expression systems have been validated with varying advantages:
| Expression System | Advantages | Limitations | Yield | Recommended Use |
|---|---|---|---|---|
| E. coli (BL21-DE3) | - Cost-effective - High yield - Simple purification using His or GST tags | - Lack of post-translational modifications - Potential inclusion body formation | 5-10 mg/L | Binding assays, in vitro kinase assays |
| Insect cells (Sf9, Hi5) | - Proper folding - Mammalian-like post-translational modifications - Suitable for functional studies | - Higher cost - Longer production time - More complex purification | 1-3 mg/L | Functional assays, structural studies |
| Mammalian cells (HEK293, CHO) | - Most authentic post-translational modifications - Native conformation - Optimal for activity assays | - Lower yields - Highest cost - Complex purification protocols | 0.5-1 mg/L | Activity assays, interaction studies with mammalian partners |
For most research applications requiring high functional activity, the baculovirus-insect cell system offers the best balance between yield and proper protein processing. For in vitro binding assays where post-translational modifications are less critical, bacterial expression using MBP or GST fusion tags has been successfully employed to produce Bora that can bind and activate Aurora A .
Several robust methodologies have been established to assess Bora-Aurora A interactions:
Co-immunoprecipitation (Co-IP):
GST/MBP pull-down assays:
In vitro kinase assays:
Surface Plasmon Resonance (SPR):
Immobilize purified Bora or Aurora A on sensor chips
Measure real-time binding kinetics with the partner protein
Determine association/dissociation constants
Fluorescence Resonance Energy Transfer (FRET):
Tag Bora and Aurora A with compatible fluorophores
Measure energy transfer upon interaction in vitro or in live cells
The optimal method should be selected based on the specific research question, with a combination of approaches providing the most robust validation of interaction.
Bora has been identified as having a significant oncogenic role in ovarian cancer (OC) through multiple lines of evidence:
Ovarian Cancer Implications:
Experimental Models for Studying Bora in Ovarian Cancer:
These models collectively provide complementary approaches to investigate Bora's role in ovarian cancer, with the combination of in vitro, in vivo, and clinical specimen analysis offering the most comprehensive understanding of its oncogenic functions.
Several promising therapeutic strategies targeting Bora or its associated pathways have emerged from recent research:
Direct Bora Inhibition:
Targeting Downstream Effectors:
Aurora A Inhibitors:
Since Bora activates Aurora A, Aurora A inhibitors (several in clinical trials) may indirectly block Bora-dependent pathways
This approach may be particularly effective in cancers with Bora overexpression
PLK1 Inhibitors:
As Bora is critical for PLK1 activation, PLK1 inhibitors represent another strategy to block the Bora-dependent oncogenic pathway
Examples include volasertib and BI2536
Cell Cycle Checkpoint Modulators:
Targeting the G2/M checkpoint in combination with Bora pathway inhibition may create synthetic lethality in cancer cells
Combination Approaches:
These therapeutic strategies highlight the potential of targeting the Bora pathway as a novel approach for cancer treatment, particularly in tumors showing high Bora expression, such as advanced ovarian cancers.
Bora's activity and stability are tightly regulated through multiple post-translational modifications (PTMs) that ensure precise temporal control of its function during cell cycle progression:
CDK1-mediated phosphorylation:
ATR-mediated phosphorylation:
Ubiquitin-dependent degradation:
Bora levels are regulated through the ubiquitin-proteasome system
SCF-β-TrCP E3 ubiquitin ligase complex has been implicated in Bora degradation
Degradation timing is critical for proper mitotic progression
Aurora A-mediated phosphorylation:
Creates a feedback loop where Aurora A may phosphorylate Bora
This may affect Bora's activity or stability
These modification events form an intricate regulatory network that coordinates Bora function with cell cycle progression and DNA damage responses. Understanding these PTMs provides potential intervention points for therapeutic development targeting Bora-dependent pathways in cancer.
The structural basis for Bora-mediated activation of Aurora A involves specific domain interactions and conformational changes:
Critical Binding Regions:
The N-terminal region of Bora is essential for Aurora A binding
Deletion studies have shown that removing either the conserved region (BoraΔ2) or a region N-terminal to the conserved part (BoraΔ1) abrogates the interaction with Aurora A
The C-terminus of Bora (beyond amino acid 404 in Drosophila Bora) is dispensable for Aurora A binding and activation
Activation Mechanism:
Bora binding likely induces conformational changes in Aurora A's activation loop
This conformational change enhances Aurora A's catalytic activity toward substrates like PLK1
The interaction may also protect the activating phosphorylation of Aurora A (Thr288 in humans) from dephosphorylation
Species Conservation:
Comparison with Other Activators:
Unlike TPX2 (another Aurora A activator), which prevents PP1-dependent dephosphorylation, Bora appears to have a more direct activation mechanism
Bora may represent a more universal activation pathway for Aurora A that is conserved across species
While detailed structural data from crystallography or cryo-EM studies of the Bora-Aurora A complex are still pending, these biochemical insights provide a framework for understanding the molecular basis of this critical regulatory interaction in mitotic progression.
For robust and reproducible in vitro kinase assays with recombinant Bora and Aurora A, the following optimized conditions are recommended:
Reagent Preparation:
Recombinant Aurora A: Best expressed in insect cells with a removable His-tag
Recombinant Bora: Can be expressed as MBP-fusion protein from bacteria or insect cells
Both proteins should be purified to >90% homogeneity by affinity chromatography followed by size exclusion
Reaction Components:
| Component | Optimal Concentration | Notes |
|---|---|---|
| Kinase buffer | 50 mM Tris-HCl (pH 7.5), 10 mM MgCl₂, 1 mM DTT | pH is critical for activity |
| ATP | 50-100 μM | Can use [γ-³²P]ATP for radioactive assays |
| Aurora A | 50-100 ng per reaction | Activity may vary between preparations |
| Bora | 100-500 ng per reaction | 2-5 fold molar excess over Aurora A |
| Substrate | 1-2 μg (MBP or histone H3) | For measuring Aurora A activity |
| Reaction volume | 20-25 μl total | |
| Temperature | 30°C | Optimal for balancing activity and stability |
| Incubation time | 20-30 minutes | Kinetics should remain linear |
Controls to Include:
Aurora A alone without Bora (baseline activity)
Heat-inactivated Aurora A (negative control)
Known Aurora A activator such as TPX2 (positive control)
Catalytically inactive Aurora A mutant (e.g., K162R)
Detection Methods:
Radioactive assay: Use [γ-³²P]ATP and detect incorporation by autoradiography or scintillation counting
Western blot: Use phospho-specific antibodies against Aurora A (pT288) or substrate
ELISA-based: For higher throughput quantification
ADP-Glo™: Luminescence-based detection of ADP production
These optimized conditions should provide a reliable system to assess Bora's ability to activate Aurora A and can be adapted for inhibitor screening or mechanistic studies.
3D cell culture systems offer significant advantages over traditional 2D cultures for studying Bora-dependent pathways in a more physiologically relevant context. Here are methodological approaches for effectively modeling these pathways:
Recommended 3D Culture Systems:
| Culture System | Methodology | Advantages | Applications |
|---|---|---|---|
| Spheroid Culture | - Single cell suspensions in ultra-low attachment plates - Hanging drop method - Rotating bioreactors | - Simple setup - Good reproducibility - Compatible with high-throughput screening | - Assessing effects of Bora modulation on growth - Drug sensitivity testing |
| Matrigel/ECM Overlay | - Cells cultured on thin layer of Matrigel with ECM overlay - Allow formation of acini-like structures | - Recapitulates basement membrane interactions - Enables polarity studies | - Investigating Bora's role in cell polarity - Asymmetric division analysis |
| Organoid Culture | - Stem cell-derived 3D structures - Patient-derived tissue fragments | - Closest to in vivo architecture - Maintains tissue-specific differentiation | - Long-term effects of Bora manipulation - Tissue-specific functions |
| Scaffold-Based Systems | - Synthetic or natural polymers (e.g., PEG, alginate) - 3D-printed matrices | - Controllable matrix properties - Customizable geometries | - Studying mechanical aspects of Bora function - Migration/invasion in defined architecture |
Analytical Methods for 3D Systems:
Live Imaging Approaches:
Confocal microscopy with Bora-fluorescent protein fusions
Time-lapse imaging to track mitotic progression
Light-sheet microscopy for deeper tissue penetration
Molecular Analysis:
RNA isolation from 3D cultures using specialized extraction protocols
Protein extraction with enhanced lysis buffers containing stronger detergents
Single-cell sequencing for heterogeneity assessment
Functional Assays:
Growth curve analysis using 3D image quantification
Invasion/migration assays in 3D matrices
Cell cycle analysis by EdU incorporation and flow cytometry
Apoptosis detection using cleaved caspase-3 staining
Genetic Manipulation Strategies:
Inducible shRNA or CRISPR systems for temporal control
Viral transduction protocols optimized for 3D structures
Region-specific genetic manipulation for spatial control
These methodologies enable researchers to investigate Bora's functions in contexts that better recapitulate the in vivo environment, particularly important for understanding its roles in processes like asymmetric cell division, which are highly dependent on three-dimensional cellular organization.
Several cutting-edge technologies are poised to significantly advance our understanding of Bora's functions:
CRISPR-based Technologies:
Base editing and prime editing: For introducing precise mutations in Bora to study structure-function relationships
CRISPRi/CRISPRa systems: For temporal control of Bora expression in specific tissues
CRISPR screens: To identify synthetic lethal interactions with Bora in cancer cells
Advanced Imaging Technologies:
Super-resolution microscopy: To visualize Bora-Aurora A interactions at nanometer resolution
Lattice light-sheet microscopy: For long-term imaging of Bora dynamics during development
FRET/FLIM techniques: To measure real-time protein interactions in living cells and tissues
Single-cell Multi-omics:
Single-cell RNA-seq with CITE-seq: To correlate Bora expression with protein markers across cell populations
Single-cell proteomics: To analyze Bora pathway activity at individual cell resolution
Spatial transcriptomics: To map Bora expression patterns in intact tissues
Structural Biology Approaches:
Cryo-EM: To determine the structure of Bora-Aurora A complexes
Hydrogen-deuterium exchange mass spectrometry: To map dynamic conformational changes
AlphaFold2/RoseTTAFold: For computational prediction of Bora structure and interactions
Organoid and In Vivo Technologies:
Patient-derived organoids: To study Bora function in personalized cancer models
Tissue-specific conditional knockouts: For developmental studies in mice
In vivo CRISPR screens: To identify context-dependent functions
Optical Control Technologies:
Optogenetics: For spatial and temporal control of Bora activity
Photo-caged compounds: To activate or inhibit Bora function with light
Opto-FGFR: For light-controlled activation of signaling pathways upstream of Bora
These emerging technologies, especially when used in combination, have the potential to provide unprecedented insights into Bora's functions in development, cell cycle regulation, and cancer biology.
The growing understanding of Bora's functions presents several promising avenues for precision medicine approaches in cancer therapy:
Biomarker Development:
Bora expression levels correlate with aggressive disease in ovarian cancer
The human Bora homolog is located on chromosome 13 in a region associated with breast cancer susceptibility
These observations suggest Bora could serve as a prognostic biomarker
Quantitative assessment of Bora protein or mRNA in biopsies could help stratify patients for treatment selection
Patient Stratification Strategies:
Patients with high Bora expression may benefit from targeted therapies against the Bora-Aurora A-PLK1 axis
Transcriptome analysis could identify tumors with activated Bora-dependent pathways
Combining Bora expression with other cell cycle markers could create treatment-predictive signatures
Novel Therapeutic Targets:
Downstream effectors of Bora (BCL2, CDK6) represent actionable targets with existing FDA-approved inhibitors
The synergistic effect observed when targeting these effectors offers promising combination therapy approaches
Rational drug combinations based on Bora pathway activation could maximize efficacy while minimizing toxicity
Combination Therapy Design:
Transcriptome analysis of Bora-depleted cells revealed modulated genes involved in survival, dissemination, and inflammation-related pathways
This knowledge enables rational design of combination therapies targeting multiple aspects of Bora's oncogenic functions
Example combinations include Aurora A inhibitors with anti-apoptotic protein inhibitors or immune modulators
Drug Resistance Mechanisms:
Understanding how Bora-dependent pathways contribute to therapy resistance
Developing strategies to overcome resistance by targeting Bora or its downstream effectors
Monitoring Bora pathway activation as a mechanism of acquired resistance
Innovative Therapeutic Approaches:
Protein-protein interaction disruptors targeting the Bora-Aurora A interface
Degraders (PROTACs) targeting Bora for selective degradation
Cell cycle phase-specific delivery systems that exploit Bora's cell cycle-dependent regulation
As research advances, these precision medicine approaches could significantly improve outcomes for patients with cancers driven by dysregulation of the Bora-Aurora A-PLK1 axis, particularly in ovarian and potentially breast cancers where Bora's oncogenic role has been established.