Recombinant Pongo abelii Protein BEX4 (BEX4)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes. We will fulfill your request to the best of our ability.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice and incurs an additional fee.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
BEX4Protein BEX4; Brain-expressed X-linked protein 4 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-119
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Pongo abelii (Sumatran orangutan) (Pongo pygmaeus abelii)
Target Names
BEX4
Target Protein Sequence
MESKEELAAN NLNGENAQQE NEGREQAPTQ NEETRHLGGG EGQKPGGNIR RGRVRRLVPN FRWAIPNRHI EHNEARDDVE RFVGQMMEIK RKTREQQMRH YMRFQTPEPD NHYDFCLIP
Uniprot No.

Target Background

Function

BEX4 may play a role in microtubule deacetylation by negatively regulating SIRT2 deacetylase activity toward α-tubulin. This suggests involvement in cell cycle progression and genomic stability regulation.

Database Links
Protein Families
BEX family
Subcellular Location
Cytoplasm, cytoskeleton, spindle pole. Nucleus. Cytoplasm.

Q&A

What is BEX4 and what are its primary functions in cellular processes?

BEX4 (Brain-Expressed X-linked 4) is a protein that localizes at microtubules, spindle poles, and midbodies and interacts with α-tubulin throughout mitosis. It functions as a mediator of microtubule hyperacetylation through the inhibition of sirtuin 2 (SIRT2) deacetylase. BEX4 plays a critical role in determining whether cells undergo apoptosis or adapt to aneuploidy, particularly when exposed to mitotic stress. Current research indicates that BEX4 acts as a novel oncogene by deregulating microtubule dynamics and chromosome integrity, contributing to cancer development when overexpressed .

What experimental models are currently available for studying Pongo abelii BEX4?

The most promising experimental model appears to be iPSCs derived from Sumatran orangutans (Pongo abelii). These stem cell platforms allow researchers to study protein expression and function in a controlled environment. The methodology involves Sendai virus-mediated Yamanaka factor-based reprogramming of peripheral blood mononuclear cells to generate iPSCs, which can then be used to study various proteins including BEX4. These models enable evaluation of pluripotent markers, chromosome activation status, and transcriptomic profiles that may influence or be influenced by BEX4 expression .

What are the recommended protocols for expressing and purifying recombinant Pongo abelii BEX4?

Based on established protocols for recombinant protein production, a methodological approach for Pongo abelii BEX4 would include:

  • Gene cloning into a suitable expression vector (e.g., GST-tagged or His-tagged)

  • Expression in an E. coli system (similar to the GST-BEX4 production mentioned in the literature)

  • Induction of protein expression using IPTG

  • Cell lysis under native conditions

  • Affinity purification using glutathione-agarose (for GST-tagged proteins) or nickel columns (for His-tagged proteins)

  • Validation of purified protein using Western blotting with anti-BEX4 antibodies

This approach aligns with the methodologies used for human BEX4 studies, where GST-BEX4 was successfully expressed in E. coli and purified for functional assays .

How can I establish a reliable phosphorylation assay for Pongo abelii BEX4?

To establish a phosphorylation assay for Pongo abelii BEX4, researchers should:

  • Express and purify recombinant BEX4 (wild-type and potential mutant versions such as T107A)

  • Incubate purified BEX4 with recombinant kinases (particularly PLK1) in the presence of [γ-³²P]-ATP

  • Analyze phosphorylation by autoradiography and/or immunoblotting using anti-phospho threonine antibodies

  • Validate results using kinase inhibitors (such as BI2536 for PLK1)

  • Compare phosphorylation patterns between wild-type and mutant proteins

This approach mirrors the methodology used for human BEX4, where PLK1 was shown to efficiently phosphorylate wild-type BEX4 but exhibited significantly reduced phosphorylation of the T107A mutant .

What are the best approaches for studying BEX4 localization in orangutan cells?

The recommended approach for studying BEX4 localization would include:

  • Generation of GFP-tagged BEX4 constructs or development of specific antibodies against Pongo abelii BEX4

  • Transfection of tagged constructs into orangutan iPSCs or derived cell lines

  • Immunofluorescence microscopy during different cell cycle stages, with particular attention to mitosis

  • Co-staining with markers for cellular structures (centrosomes, spindle poles, midbodies)

  • Treatment with cell cycle synchronizing agents (e.g., nocodazole) to enrich for mitotic cells

  • Comparative analysis with human cells to identify species-specific localization patterns

This methodology builds on approaches used for human BEX4, which has been observed to colocalize with PLK1 at centrosomes, spindle poles, and midbodies, particularly during mitosis .

What protein-protein interactions are critical for BEX4 function in primates?

The most well-documented interaction for BEX4 is with Polo-like kinase 1 (PLK1). This interaction is particularly prominent during mitosis and has significant functional consequences. Additional interactions include:

  • PLK1: BEX4 colocalizes and interacts with PLK1 at centrosomes, spindle poles, and midbodies during mitosis

  • α-tubulin: BEX4 interacts with α-tubulin throughout mitosis

  • SIRT2: BEX4 inhibits SIRT2 deacetylase, leading to α-tubulin hyperacetylation

  • CDK1: BEX4 forms a complex with CDK1, particularly in mitotically synchronized cells

The PLK1–BEX4 interaction appears to be a novel oncogenic signal that enables the acquisition of chromosomal aneuploidy in human cells, and would be worth investigating in Pongo abelii to determine conservation of this mechanism .

How is BEX4 regulated post-translationally and what are the implications?

BEX4 undergoes significant post-translational modification, primarily through phosphorylation. Key aspects include:

  • PLK1-mediated phosphorylation upregulates BEX4 protein stability

  • T107 is a critical phosphorylation site by PLK1

  • Phosphorylation is important for proper BEX4 localization

  • Inhibition of PLK1 alters the centrosomal localization of BEX4

  • Phosphorylation status may determine BEX4's ability to inhibit apoptosis

The stabilization of BEX4 through PLK1-mediated phosphorylation appears to be a key regulatory mechanism that contributes to its oncogenic potential in human cells. Comparative studies in Pongo abelii would help determine if this regulatory mechanism is conserved across primates .

How might BEX4 contribute to understanding species-specific differences in cell cycle regulation?

BEX4's role in cell cycle regulation, particularly in mitosis and response to spindle damage, makes it an excellent candidate for studying species-specific differences in cell cycle control mechanisms. Considerations for such research include:

  • Comparing BEX4 sequence and structural conservation between humans and Pongo abelii

  • Analyzing species-specific phosphorylation patterns and kinase interactions

  • Evaluating differences in cellular responses to BEX4 overexpression or knockdown

  • Investigating the relationship between BEX4 expression and aneuploidy adaptation across species

  • Examining evolutionary conservation of the PLK1-BEX4 regulatory axis

These comparative studies could reveal evolutionary adaptations in cell cycle regulation and provide insights into species-specific susceptibilities to certain diseases, particularly cancer .

What are the molecular mechanisms by which BEX4 inhibits apoptosis in response to mitotic stress?

The anti-apoptotic role of BEX4 during mitotic stress involves several molecular mechanisms:

  • BEX4 expression significantly decreases activation of apoptotic markers including:

    • Cleaved-PARP

    • Active-caspase 9

    • Active-caspase 7

    • Active-caspase 3

  • BEX4 augments expression of cIAP-1 (cellular inhibitor of apoptosis protein-1)

  • The PLK1–BEX4 interaction allows abnormal mitotic cells to adapt to aneuploidy rather than undergo apoptotic cell death

  • BEX4 may contribute to mitotic adaptation or "mitotic slippage" in response to spindle damage

These mechanisms suggest BEX4 acts as a molecular switch determining whether cells with mitotic abnormalities undergo apoptosis or adapt to aneuploidy. This function is particularly relevant in the context of cancer development and resistance to anti-mitotic therapies .

How would you design experiments to investigate potential differences between human and Pongo abelii BEX4 functions?

A comprehensive experimental approach would include:

  • Comparative sequence analysis and structural modeling of human vs. Pongo abelii BEX4

  • Reciprocal expression studies:

    • Express Pongo abelii BEX4 in human cell lines

    • Express human BEX4 in orangutan-derived cells

    • Compare phenotypic outcomes (aneuploidy, apoptosis resistance, etc.)

  • Molecular interaction studies:

    • Co-immunoprecipitation of BEX4 with potential partners (PLK1, CDK1) in both species

    • Yeast two-hybrid or proximity labeling approaches to identify species-specific interactors

  • Functional assays:

    • Response to microtubule inhibitors (e.g., nocodazole)

    • Mitotic progression and checkpoint activation

    • Apoptosis resistance

    • Aneuploidy development

  • CRISPR-based gene editing to introduce species-specific mutations and assess functional consequences

This multi-faceted approach would identify conserved and divergent aspects of BEX4 function between humans and orangutans .

How can Pongo abelii BEX4 studies contribute to cancer research?

Research on Pongo abelii BEX4 can provide valuable comparative insights for cancer research:

  • Evolutionary perspective on oncogenic mechanisms:

    • Identifying conserved vs. species-specific oncogenic pathways

    • Understanding fundamental vs. derived aspects of cell cycle regulation

  • Natural resistance mechanisms:

    • Some primate species may have evolved different regulatory controls for potentially oncogenic proteins

    • Comparative studies could reveal protective mechanisms against BEX4-mediated oncogenesis

  • Translational applications:

    • Conservation-based approaches to identify critical functional domains that could be targeted therapeutically

    • Development of model systems for testing cancer interventions

  • Biomarker potential:

    • Understanding if BEX4 overexpression is a universal oncogenic mechanism across primates

    • Development of cross-species validated biomarkers

The observation that BEX4 expression is highly elevated in human lung cancer cells and contributes to mTOR-induced lung carcinogenesis suggests comparative studies could reveal important insights about cancer susceptibility across primate species .

What contradictions exist in the current understanding of BEX4 function, and how might they be resolved?

Several notable contradictions or knowledge gaps exist in BEX4 research:

Contradiction/Knowledge GapPossible Resolution Approach
BEX4 promotes aneuploidy but is not lethal to cellsInvestigate compensatory mechanisms that allow survival despite chromosomal instability
Limited understanding of BEX4 tissue specificityConduct comprehensive expression profiling across tissues in multiple primate species
Unclear evolutionary conservation of BEX4 functionPerform phylogenetic analysis and functional testing across diverse primate species
Contradictory roles in cell death vs. survivalIdentify context-dependent factors that determine whether BEX4 promotes survival or death
Unknown regulation of BEX4 beyond PLK1Screen for additional kinases, phosphatases, and other regulatory proteins that modify BEX4

Resolving these contradictions requires cross-disciplinary approaches combining evolutionary biology, molecular cell biology, and cancer research. Comparative studies between human and Pongo abelii BEX4 could provide particularly valuable insights .

How might iPSC technology be leveraged to study BEX4 function in Pongo abelii?

The development of iPSCs from orangutans provides a powerful platform for studying BEX4:

  • Expression analysis during pluripotency and differentiation:

    • Monitor BEX4 levels during the reprogramming process

    • Analyze BEX4 expression across differentiated cell lineages

  • Genetic manipulation in a controlled system:

    • CRISPR/Cas9-mediated knockout or mutation of BEX4

    • Overexpression studies using viral vectors

    • Introduction of reporter constructs for live-cell imaging

  • Disease modeling:

    • Differentiate iPSCs into cell types relevant to BEX4-associated diseases (e.g., lung epithelial cells)

    • Induce stress conditions to assess BEX4-dependent responses

  • Comparative studies with human iPSCs:

    • Parallel differentiation experiments to identify species-specific differences

    • Cross-species complementation studies with BEX4 variants

The successful derivation of iPSCs from Bornean orangutans using Sendai virus-mediated Yamanaka factor-based reprogramming provides a methodological foundation for similar work with Pongo abelii, enabling sophisticated comparative studies of BEX4 function .

What emerging technologies could advance the study of BEX4 in Pongo abelii?

Several cutting-edge technologies show promise for advancing BEX4 research:

  • Spatial transcriptomics and proteomics:

    • Mapping BEX4 expression and interactions at subcellular resolution

    • Identifying cell type-specific expression patterns in tissues

  • Single-cell analysis:

    • Characterizing cell-to-cell variability in BEX4 expression and function

    • Identifying rare cell populations with unique BEX4-dependent phenotypes

  • Cryo-electron microscopy:

    • Determining high-resolution structures of BEX4 and its complexes

    • Comparing structural features between human and orangutan variants

  • Organoid technologies:

    • Developing 3D culture systems from Pongo abelii iPSCs

    • Modeling complex tissue environments for BEX4 functional studies

  • Multi-omics integration:

    • Combining genomic, transcriptomic, proteomic, and metabolomic data

    • Building comprehensive models of BEX4 regulation and function

These technologies would enable more sophisticated comparative analyses between human and Pongo abelii BEX4, potentially revealing species-specific adaptations relevant to health and disease .

How can understanding BEX4 function contribute to conservation efforts for endangered orangutans?

BEX4 research in the context of conservation biology offers several benefits:

  • Biobanking and genetic resource preservation:

    • Understanding the function of key regulatory proteins facilitates better preservation methods

    • Development of functional assays to verify viability of preserved genetic material

  • Reproductive technology advancement:

    • Knowledge of cell cycle regulators like BEX4 can improve assisted reproductive technologies

    • iPSC-derived gametes might serve as a future conservation tool

  • Health monitoring in wild populations:

    • Development of molecular markers for health assessment

    • Identification of potential susceptibilities to environmental toxins

  • Disease resistance understanding:

    • Comparative oncology may reveal why certain cancers affect humans differently than orangutans

    • Insights into species-specific immune and cellular defense mechanisms

The successful derivation of iPSCs from orangutans represents an important step toward preserving the genetic diversity of these endangered primates while offering insights into primate stem cell biology that may have conservation applications .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.