UXT Human

Ubiquitously-Expressed, Prefoldin-Like Chaperone Human Recombinant
Shipped with Ice Packs
In Stock

Description

Transcriptional Regulation

UXT acts as a cofactor for multiple signaling pathways:

Interaction PartnerBiological RoleMechanism
Androgen Receptor (AR)Corepressor in prostate cellsBinds AR N-terminus; suppresses proliferation via differentiation pathways .
NF-κBEssential coactivatorEnhances NF-κB enhanceosome assembly; required for TNF-α-induced transcription .
P53/MDMXModulates apoptosisInhibits P53 by stabilizing MDMX; promotes glycolysis via HIF-1α .

Centrosome Biology

  • Localizes to centrosomes, interacting with γ-tubulin to maintain structure .

  • Overexpression disrupts centrosomal γ-tubulin distribution, causing mitotic defects .

  • siRNA knockdown induces p53-independent cell death, highlighting its essential role .

Role in Cancer

UXT exhibits tissue-specific oncogenic or tumor-suppressive effects:

Tumor-Promoting Contexts

Cancer TypeExpressionMechanism
BreastUpregulatedEnhances glycolysis via HIF-1α; inhibits P53-mediated apoptosis .
OsteosarcomaUpregulatedActivates NF-κB, increasing survival and metabolic adaptation .
Bladder/OvaryUpregulatedCorrelates with centrosome dysfunction and aneuploidy .

Tumor-Suppressive Contexts

Cancer TypeExpressionMechanism
ProstateDownregulatedLoss enhances AR-driven proliferation; promotes dedifferentiation .

Diagnostic Potential

  • Elevated UXT levels in bladder, breast, and ovarian tumors suggest utility as a biomarker .

  • Reduced expression in prostate cancer correlates with aggressive phenotypes .

Therapeutic Targeting

  • UXT-V2 inhibition in breast cancer restores P53 activity and reduces glycolysis .

  • Centrosomal UXT disruption sensitizes cells to apoptosis, offering a strategy for p53-mutant cancers .

Research Gaps

  • Mechanisms underlying UXT’s dual role in tumorigenesis remain unclear.

  • Role in ferroptosis or immunogenic cell death warrants exploration .

Product Specs

Introduction
Ubiquitously-expressed transcript (UXT) belongs to the UXT family and functions as a cofactor, influencing androgen receptor-dependent transcription. It plays a crucial role in tumor necrosis factor-induced apoptosis. UXT gene expression is implicated in tumorigenesis. UXT is expressed ubiquitously, including in prostate epithelial cells, and is overexpressed in various tumor tissues. The highest UXT levels are observed in the heart, skeletal muscle, pancreas, kidney, liver, adrenal gland, peripheral blood leukocytes, lymph nodes, prostate, and thyroid. Conversely, the lowest levels are found in the bladder and uterus.
Description
Recombinant human UXT, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 177 amino acids (residues 1-157) and possessing a molecular weight of 20.4 kDa. It features a 20 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The UXT protein solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms
Protein UXT, Androgen receptor trapped clone 27 protein, ART-27, Ubiquitously expressed transcript protein, UXT, HSPC024, STAP1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MATPPKRRAV EATGEKVLRY ETFISDVLQR DLRKVLDHRD KVYEQLAKYL QLRNVIERLQ EAKHSELYMQ VDLGCNFFVD TVVPDTSRIY VALGYGFFLE LTLAEALKFI DRKSSLLTEL SNSLTKDSMN IKAHIHMLLE GLRELQGLQN FPEKPHH.

Q&A

What is UXT and what are its primary biological functions?

UXT is a protein expressed in virtually all human and mouse tissues, functioning primarily as a transcriptional regulator. As revealed through yeast two-hybrid screening and subsequent validation studies, UXT interacts with several transcription factors including androgen receptor (AR), GATA4, NF-κB, and EVI1 . These interactions enable UXT to regulate the expression of downstream genes involved in various cellular processes. Additionally, UXT participates in cell viability regulation through centrosome interaction and serves as a component of the TNF receptor signaling complex, contributing to antiviral pathways . The protein's widespread expression pattern suggests fundamental roles in cellular homeostasis, while its diverse interaction partners indicate multifaceted functions in gene regulation networks. The methodological approach to studying UXT typically begins with protein interaction identification followed by functional validation through gene expression analysis after UXT modulation.

What established protein-protein interactions define UXT function in human cells?

UXT engages in multiple protein-protein interactions that define its diverse cellular functions. Key validated interactions include:

Interaction PartnerCellular LocationFunctional SignificanceDetection Method
EZH2NucleusEnhances histone methyltransferase activityYeast two-hybrid, co-IP
SUZ12NucleusFacilitates PRC2 complex formationProtein binding assays
AR (Androgen Receptor)NucleusRegulates AR-dependent gene expressionPreviously established
GATA4NucleusTranscriptional regulationPreviously established
NF-κBNucleusInflammatory signaling regulationPreviously established
EVI1NucleusTranscriptional regulationPreviously established
CentrosomeCytoplasmCell viability regulationPreviously established
TNF receptor complexCell membraneAntiviral pathway signalingPreviously established

Notably, UXT does not interact with EED (embryonic ectoderm development), another component of the PRC2 complex, highlighting the specificity of its interactions . When investigating these interactions, researchers should employ multiple complementary approaches, including yeast two-hybrid screening for initial discovery, co-immunoprecipitation for validation in cellular contexts, and functional assays to determine the biological consequences of the interactions. Quantitative analysis of interaction strength under various cellular conditions can provide additional insights into context-dependent functions.

How does UXT mechanistically regulate PRC2 complex function and histone methyltransferase activity?

UXT demonstrates a sophisticated regulatory mechanism for PRC2 complex function through direct protein-protein interactions. Research has established that UXT directly binds to two critical PRC2 complex components: EZH2 (enhancer of zeste homolog 2) and SUZ12 (suppressor of zeste 12 homolog), but notably does not interact with EED (embryonic ectoderm development) . This selective binding pattern suggests a specific architectural role in PRC2 assembly. Mechanistically, UXT activates EZH2 histone methyltransferase activity by facilitating EZH2 binding with SUZ12, essentially promoting optimal complex formation .

Functional studies demonstrate that UXT knockdown significantly inhibits EZH2 histone methyltransferase activity, evidenced by decreased H3K27 methylation and corresponding increased expression of known PRC2 target genes, including HOXA9 and DAB2IP . Quantitative chromatin immunoprecipitation (qChIP) assays further confirm this mechanism, revealing lower levels of H3K27 trimethylation on the HOXA9 promoter in UXT-depleted cells compared to controls . The dependence of this effect on EZH2 was elegantly demonstrated through comparative studies in EZH2-knockout cells, where UXT depletion failed to alter HOXA9 and DAB2IP expression . This mechanistic pathway connects UXT to epigenetic regulation of gene expression through modulation of histone modifications, potentially explaining its diverse effects on cellular phenotypes in different contexts.

What explains the context-dependent roles of UXT in different cancer types?

The paradoxical behavior of UXT across different cancer types represents an intriguing aspect of its biology that warrants mechanistic investigation. Current evidence reveals a stark contrast: UXT functions as a tumor promoter in clear cell renal cell carcinoma (ccRCC), colorectal cancer, sarcoma, and breast tumors, while exhibiting tumor-suppressive properties in prostate cancer . Several hypotheses may explain this context-dependent functionality:

  • Tissue-specific interactome: UXT likely engages with different protein partners across tissue types, potentially leading to activation of distinct downstream pathways. For instance, UXT's interaction with androgen receptor (AR) in prostate tissue may direct different outcomes compared to its PRC2 interaction in renal tissue.

  • Epigenetic landscape variations: The pre-existing epigenetic state of different tissues may determine whether UXT-mediated histone modifications promote or suppress oncogenic programs.

  • Alternative splicing or post-translational modifications: Though not explicitly mentioned in the search results, tissue-specific UXT isoforms or modifications could alter its function.

  • Microenvironmental factors: The search results specifically mention that UXT's role depends on "tumor types and microenvironments" , suggesting that external factors such as inflammation, hypoxia, or stromal interactions may influence UXT function.

  • Concentration-dependent effects: UXT may exhibit dose-dependent effects, with moderate levels promoting certain cellular functions and high levels triggering compensatory mechanisms.

Experimental approaches to investigate these hypotheses should include comparative interactome analysis across tissue types, ChIP-seq to map genome-wide binding patterns, and context-specific knockout models to assess functional dependencies.

What is the significance of UXT upregulation in clear cell renal cell carcinoma (ccRCC) progression?

At the molecular level, UXT promotes ccRCC progression through its interaction with the PRC2 complex. By enhancing EZH2 histone methyltransferase activity, UXT increases H3K27 trimethylation, subsequently repressing tumor suppressor genes . Functional studies provide compelling evidence for UXT's oncogenic role in ccRCC: knockdown of UXT significantly inhibits proliferation, colony formation, migration, and invasion of renal cancer cells . Critically, these effects are EZH2-dependent, as demonstrated through comparative studies in EZH2-knockout cell models .

The UXT-PRC2 axis represents a potential therapeutic vulnerability in ccRCC. Strategies targeting this interaction could include:

  • Small molecule inhibitors disrupting UXT-EZH2 binding

  • Peptide mimetics that compete with natural binding interfaces

  • Degraders specifically targeting UXT in renal tissues

Future research should focus on validating UXT as a biomarker in larger patient cohorts and developing precise methods to target the UXT-PRC2 interaction in ccRCC.

How might UXT's interaction with different transcription factors explain its diverse cellular effects?

UXT's interaction with multiple transcription factors likely underlies its diverse and sometimes contradictory cellular effects. Research has established that UXT regulates several key transcription factors including AR (Androgen Receptor), GATA4, NF-κB, and EVI1 . These interactions potentially create a network of context-dependent regulatory mechanisms:

  • Androgen Receptor (AR) pathway: UXT regulates AR downstream genes as a co-factor by interacting with VHL and URI/RMP . This interaction may explain UXT's tumor-suppressive effects in prostate cancer, where AR signaling plays a central role in cancer progression.

  • NF-κB signaling: UXT's role as a component of the TNF receptor signaling complex suggests involvement in inflammatory pathways . This could influence the tumor microenvironment and immune response, potentially contributing to cancer progression in inflammation-associated malignancies.

  • EVI1 regulation: While UXT has been reported to suppress EVI1-mediated cell transformation , EVI1 overexpression occurs in various myeloid malignancies and solid tumors. This interaction may represent another mechanism through which UXT exhibits tumor-suppressive functions in specific contexts.

  • GATA4 modulation: GATA4 is crucial for cardiac development and function, suggesting UXT may have roles beyond cancer in tissue development and homeostasis.

  • PRC2 complex interaction: Beyond direct transcription factor binding, UXT's enhancement of PRC2 activity likely alters the broader epigenetic landscape, affecting numerous downstream targets .

The relative dominance of these different interaction networks in specific tissue contexts may determine whether UXT ultimately promotes or suppresses cellular proliferation, differentiation, and survival. Investigating the tissue-specific interactome of UXT through techniques like BioID or proximity labeling, combined with transcriptomic analysis following UXT modulation, could help unravel these complex regulatory networks.

What experimental strategies are most effective for studying UXT protein interactions in human cells?

Investigating UXT protein interactions requires a comprehensive experimental approach combining complementary techniques. Based on successful strategies in the literature, researchers should consider the following methodological pipeline:

  • Initial interaction screening:

    • Yeast two-hybrid (Y2H) screening has proven successful in identifying novel UXT binding partners such as EZH2

    • Affinity purification coupled with mass spectrometry (AP-MS) can provide an unbiased view of the UXT interactome

    • Proximity-based labeling approaches (BioID, APEX) offer advantages for detecting transient or weak interactions

  • Interaction validation:

    • Co-immunoprecipitation (Co-IP) confirms interactions in cellular contexts

    • Proximity ligation assay (PLA) visualizes interactions in situ

    • GST pull-down or similar in vitro binding assays determine direct versus indirect interactions

    • FRET or BRET assays can measure real-time interaction dynamics

  • Interaction domain mapping:

    • Truncation mutants help identify specific interaction domains

    • Point mutations in predicted interface residues confirm binding specificity

    • Peptide arrays can map interaction interfaces with high resolution

  • Functional characterization:

    • Knockdown of UXT followed by assessment of partner protein activity (as demonstrated with EZH2)

    • Chromatin immunoprecipitation (ChIP) to analyze effects on chromatin association

    • Transcriptomic analysis to identify genes affected by the interaction

  • Structural studies:

    • X-ray crystallography or cryo-EM to determine the molecular basis of interactions

    • Molecular dynamics simulations to predict interaction stability and drug binding potential

When implementing these approaches, researchers should include appropriate controls such as interaction-deficient mutants and ensure validation across multiple cell types given UXT's context-dependent functions.

What are best practices for analyzing the effects of UXT on histone modifications and gene expression?

Analyzing UXT's effects on histone modifications and gene expression requires rigorous experimental design and integrated multi-omics approaches. Based on successful methodologies in the literature, researchers should consider the following best practices:

  • Chromatin modification analysis:

    • Quantitative chromatin immunoprecipitation (qChIP) assays provide locus-specific information about histone modifications, as demonstrated in studies examining H3K27me3 levels at the HOXA9 promoter following UXT knockdown

    • ChIP-sequencing (ChIP-seq) for genome-wide profiling of histone modifications affected by UXT modulation

    • CUT&RUN or CUT&TAG for higher resolution and lower background in profiling histone modifications

    • Western blotting for global assessment of histone modification levels

  • Gene expression analysis:

    • RT-qPCR for candidate genes known to be regulated by PRC2, such as HOXA9 and DAB2IP

    • RNA-sequencing for unbiased, genome-wide assessment of gene expression changes

    • Single-cell RNA-seq to capture cellular heterogeneity in response to UXT modulation

    • Nascent RNA sequencing (GRO-seq, PRO-seq) to distinguish direct transcriptional effects from secondary changes

  • Integrated analysis approaches:

    • Combined ChIP-seq and RNA-seq analysis to correlate histone modification changes with gene expression

    • ATAC-seq to assess chromatin accessibility changes in conjunction with expression data

    • HiChIP or similar approaches to connect three-dimensional genome organization with gene regulation

  • Experimental design considerations:

    • Include both UXT knockdown and overexpression conditions

    • Perform time-course experiments to distinguish primary from secondary effects

    • Use domain-specific mutants to separate UXT's different functions

    • Include appropriate controls such as EZH2 knockout cells to establish dependency relationships

    • Validate findings across multiple cell lines to account for context-dependent effects

  • Data analysis and validation:

    • Employ robust statistical methods appropriate for each data type

    • Validate key findings using orthogonal techniques

    • Perform pathway enrichment analysis to identify biological processes affected

    • Consider UXT expression levels in interpretation of results

These comprehensive approaches will provide a detailed understanding of how UXT influences the epigenetic landscape and gene expression programs in normal and pathological states.

What are critical considerations for studying UXT expression in patient-derived samples?

Studying UXT expression in patient-derived samples requires careful attention to multiple factors that can influence data quality and interpretation. Based on research methodologies, the following critical considerations should be addressed:

Adhering to these considerations will enhance the reliability and clinical relevance of UXT expression studies in patient-derived samples.

What genetic and pharmacological tools are available for modulating UXT activity in experimental models?

Researchers investigating UXT function can employ a variety of genetic and pharmacological approaches to modulate its activity. Based on research methodologies, the following tools have demonstrated utility:

  • Genetic modulation approaches:

    • RNA interference (RNAi): siRNA and shRNA approaches have been successfully employed to knockdown UXT expression, revealing its role in cellular processes including proliferation, colony formation, migration, and invasion

    • CRISPR/Cas9 genome editing: While not explicitly mentioned for UXT in the search results, this technology has been used for EZH2 knockout and could be applied to generate UXT knockout cell lines or animal models

    • Overexpression systems: Plasmid-based overexpression of wild-type UXT or functional mutants can help elucidate gain-of-function effects

    • Inducible expression systems (e.g., Tet-On/Off): These allow temporal control of UXT expression for studying dynamic processes

    • Domain-specific mutants: Generating mutations in specific functional domains can help dissect the contribution of different UXT interactions

  • Pharmacological approaches:

    • While no direct UXT inhibitors have been reported in the search results, researchers can target UXT-dependent pathways:

      • EZH2 inhibitors (e.g., tazemetostat) can be used to block the downstream effects of UXT on PRC2 activity

      • Protein-protein interaction disruptors designed to interfere with UXT-EZH2 or UXT-SUZ12 binding

      • Proteolysis targeting chimeras (PROTACs) could be developed to induce selective degradation of UXT

  • Cell and animal models:

    • Cell line selection: Given UXT's context-dependent functions, researchers should select models relevant to their specific research question (e.g., renal cell lines for studying oncogenic functions, prostate cancer lines for tumor-suppressive effects)

    • Patient-derived xenografts: These maintain tumor heterogeneity and better recapitulate patient tumors

    • Genetically engineered mouse models (GEMMs): Tissue-specific UXT knockout or overexpression can reveal in vivo functions

    • Organoid systems: These provide a 3D culture environment that better mimics in vivo conditions while maintaining genetic manipulation capabilities

  • Functional readouts:

    • Proliferation and colony formation assays: These have successfully demonstrated UXT's effects on cancer cell growth

    • Migration and invasion assays: These revealed UXT's role in promoting metastatic potential in renal cancer cells

    • Histone methyltransferase activity assays: These can assess the impact of UXT modulation on PRC2 function

    • Transcriptomic analysis: RNA-seq following UXT modulation can identify affected pathways

When designing experiments using these tools, researchers should include appropriate controls and consider potential compensatory mechanisms that may arise in response to long-term UXT modulation.

Product Science Overview

Introduction

The Ubiquitously-Expressed, Prefoldin-Like Chaperone, also known as UXT, is a protein encoded by the UXT gene. This protein plays a crucial role in various cellular processes, including gene transcription regulation and apoptosis. It is involved in the modulation of androgen receptor-dependent transcription and tumor necrosis factor-induced apoptosis .

Gene and Protein Structure

The UXT gene is a protein-coding gene associated with several diseases, including Amyotrophic Lateral Sclerosis 1 and Retinitis Pigmentosa . The protein encoded by this gene has multiple isoforms due to alternatively spliced transcript variants . UXT is known for its chromatin binding and actin filament binding properties .

Functions

UXT functions as a cofactor that modulates androgen receptor-dependent transcription. It plays a critical role in tumor necrosis factor-induced apoptosis, which is essential for maintaining cellular homeostasis and preventing tumorigenesis . Additionally, UXT acts as a nuclear chaperone that facilitates the formation of the NF-kappa-B enhanceosome, thereby positively regulating NF-kappa-B transcription activity .

Mechanisms

UXT works in concert with the corepressor URI1 to regulate androgen receptor-mediated transcription. Together, they associate with chromatin to the NKX3-1 promoter region . UXT also negatively regulates the transcriptional activity of the estrogen receptor ESR1 by inducing its translocation into the cytoplasm . Furthermore, UXT is a potential component of mitochondrial-associated LRPPRC, which integrates mitochondria and the microtubular cytoskeleton with chromosome remodeling .

Recombinant UXT

Recombinant human UXT is available for research purposes. It is typically provided in a solution containing Tris-HCl buffer, urea, and glycerol . This recombinant protein is used in various biochemical assays to study its functions and mechanisms in detail.

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.