RXRA Human

Retinoid X Receptor Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RXRA Human

Retinoid X Receptor Alpha (RXRA), encoded by the RXRA gene, is a nuclear receptor that mediates retinoid signaling. As a member of the steroid-thyroid hormone receptor superfamily, RXRA regulates gene expression by forming heterodimers with other nuclear receptors (e.g., RARs, PPARs) and binding to retinoic acid response elements (RAREs) in DNA . Its primary ligand is 9-cis-retinoic acid (9-cRA), which triggers conformational changes in the receptor, enabling transcriptional activation .

Molecular Structure and Ligand Specificity

The ligand-binding domain (LBD) of RXRA is critical for its function. The crystal structure of human RXRA LBD bound to 9-cRA reveals a unique ligand-binding pocket (LBP) that accommodates the 9-cis isomer due to a pronounced kink (~70°) and a rotated β-ionone ring orientation . This structural specificity excludes all-trans-retinoic acid (a-tRA), which binds preferentially to RARs .

FeatureRXRARARγ
Ligand9-cRA (high affinity)9-cRA and a-tRA
LBP GeometrySharper kink (~70°)Gentler kink (~60°)
β-Ionone OrientationPoints downward (away from H12)Points toward H12
Hydrophobic Contacts67 (with 9-cRA)83 (with 9-cRA), 94 (with a-tRA)

Comparative structural features of RXRA and RARγ LBDs .

Biological Functions and Pathways

RXRA regulates diverse physiological processes through heterodimerization with partner receptors:

Metabolic Regulation

  • Lipid Metabolism: Forms heterodimers with PPARA to activate fatty acid oxidation genes (e.g., ACOX1) .

  • Glucose Homeostasis: Modulates PPARγ activity in adipose tissue, influencing insulin sensitivity .

Development and Disease

  • Cardiovascular Health: Endothelial RXRA activation repairs anthracycline-induced cardiomyopathy by restoring tight-junction proteins (e.g., ZO-1) .

  • Cancer Resistance: Downregulation in chronic myeloid leukemia (CML) cells correlates with imatinib resistance; RXRA ligands (e.g., bexarotene) enhance drug sensitivity .

PathwayFunctionClinical Relevance
PPARA/RXRAFatty acid oxidationMetabolic disorders
RAR/RXRARetinoid signalingAPL, cancer differentiation
Endothelial RXRABarrier repairAnthracycline-induced cardiomyopathy

Therapeutic Applications

RXRA agonists are explored for their therapeutic potential:

Cancer Therapy

  • Bexarotene: A pan-RXR agonist used in cutaneous T-cell lymphoma (CTCL) and breast cancer. Enhances RXRA/PPARA activity, promoting differentiation and apoptosis .

  • Imatinib Synergy: Pre-treatment with RXRA ligands (e.g., bexarotene) sensitizes CML cells to imatinib by upregulating RXRA and restoring transcriptional activity .

Neurological and Cardiovascular Disorders

  • Multiple Sclerosis: Bexarotene promotes remyelination by enhancing oligodendrocyte differentiation .

  • Anthracycline Toxicity: RXRA activation in endothelial cells mitigates cardiomyopathy by preserving vascular integrity .

Congenital Heart Defects

  • Tetralogy of Fallot (TOF): Elevated promoter methylation of RXRA in right ventricular outflow tract (RVOT) myocardium correlates with reduced mRNA expression, impairing cardiac development .

Cancer

  • Bladder/Pancreatic Cancers: RXRA downregulation is linked to tumor progression, though mechanisms remain under investigation .

Transcriptional Control

  • Corepressor Recruitment: In the absence of ligand, RXRA-RAR heterodimers recruit HDAC-containing complexes, suppressing transcription .

  • Coactivator Binding: 9-cRA binding induces helical rearrangements (H3, H11, H12), enabling interaction with coactivators (e.g., CBP/p300) .

Epigenetic Modifications

  • Promoter Methylation: Hypermethylation of RXRA regulatory regions (e.g., −1453 to −1000) silences transcription in TOF and colon cancer .

Key Research Findings

  1. Structural Insights: The RXRA LBD adopts an agonist conformation without direct ligand-H12 contact, distinguishing it from RARs .

  2. Cancer Therapy: RXRA overexpression or ligand activation enhances imatinib efficacy in CML by targeting CD34+ stem cells .

  3. Cardioprotection: Endothelial-specific rxraa activation in zebrafish models rescues anthracycline-induced cardiomyopathy .

  4. Epigenetics: RXRA promoter methylation is a biomarker for TOF, highlighting epigenetic therapy potential .

Product Specs

Introduction
The retinoid X receptor (RXR) is a nuclear receptor transcription factor with diverse functions. It interacts with various nuclear receptor dimeric partners. RXR binds to specific DNA sequences called response elements. It does this as a homodimer when its ligand, 9-cis retinoic acid, is present. RXR can also bind as a heterodimer with other nuclear hormone receptor superfamily members. These include retinoic acid receptors (RAR), thyroid hormone receptors (TR), vitamin D receptors, and peroxisome proliferator-activated receptors (PPAR). The RXR family consists of three isoforms: RXRα, RXRβ, and RXRγ. The human RXRα gene is located on chromosome 9q34.9. It encodes two main isoforms: RXRα1 and RXRα2. A purified portion of the RXR DNA binding domain (amino acids 111-228) was obtained using standard column chromatography techniques.
Description
Recombinant human RXRA, produced in E. coli, is a single polypeptide chain without any glycosylation modifications. It contains 119 amino acids (residues 111-228) and has a molecular weight of 13.6 kDa.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The protein is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 7.5), 0.1 M NaCl, and 5 mM β-mercaptoethanol.
Stability
For short-term storage (up to 2-4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is greater than 95.0% as assessed by SDS-PAGE analysis.
Synonyms
Retinoic acid receptor RXR-alpha, Retinoid X receptor alpha, Nuclear receptor subfamily 2 group B member 1, RXRA, NR2B1, FLJ00280, FLJ00318, FLJ16020, FLJ16733, MGC102720, RXR Alpha, RXR-a.
Source
Escherichia Coli.
Amino Acid Sequence

MLGLNGVLKV PAHPSGNMAS FTKHICAICG DRSSGKHYGV YSCEGCKGFF KRTVRKDLTY TCRDNKDCLI DKRQRNRCQY CRYQKCLAMG MKREAVQEER QRGKDRNENE VESTSSANE.

Q&A

What is RXRA and what is its primary function in human cells?

RXRA is a nuclear receptor that regulates transcription either as a homodimer or as an obligate heterodimerization partner for 14 other nuclear receptors, including the three peroxisome proliferator-activated receptors (PPARA, PPARD, and PPARG). It functions as a ligand-activated transcription factor that controls gene expression by binding to specific DNA sequences called response elements .

The primary mechanism of action involves:

  • Formation of homo- or heterodimers

  • Binding to direct repeat (DR) sequences in DNA

  • Recruitment of co-activators or co-repressors

  • Regulation of target gene transcription

RXRA and its partners show preference for direct repeats with specific spacer lengths, with RXR homodimers and RXR/PPAR heterodimers preferentially binding to direct repeats with a single nucleotide spacer (DR1) .

How does RXRA interact with other nuclear receptors?

RXRA serves as an obligate heterodimerization partner for 14 different nuclear receptors. The specificity and function of these heterodimers depend on:

  • The partner receptor (e.g., PPARs, RAR, etc.)

  • The DNA response element configuration

  • The presence of specific ligands

For PPAR partnerships specifically, experimental evidence shows:

  • RXRA/PPAR heterodimers preferentially bind to DR1 motifs

  • Both PPARD and PPARG can form functional heterodimers with RXRA

  • These heterodimers show distinct but sometimes overlapping transcriptional activities

When forming heterodimers with retinoic acid receptors (RARs), RXRA shows different binding preferences, favoring DR5 elements (direct repeats with 5 nucleotide spacers) .

What experimental methods are commonly used to study RXRA binding to DNA?

The primary methods for studying RXRA-DNA interactions include:

MethodApplicationAdvantagesLimitations
ChIP-seqGenome-wide binding site identificationIdentifies in vivo binding sitesRequires high-quality antibodies
Reporter assays (e.g., luciferase)Functional validation of binding sitesQuantifiable transcriptional outputArtificial context
EMSADirect binding assessmentCan detect complex formationIn vitro conditions only
DNase footprintingPrecise binding site determinationBase-pair resolutionTechnical complexity

As demonstrated in the bladder cancer studies, ChIP-seq for RXRA combined with H3K27ac marks can identify "RXR-bound Active Enhancer/promoters" (RAEs) to determine where RXRA is actively regulating transcription .

How are RXRA mutations implicated in bladder cancer development?

RXRA mutations, particularly at position S427 (S427F/Y), are found in 5-8% of bladder cancer cases across multiple independent cohorts. These mutations represent a gain-of-function mechanism that drives cancer progression through:

  • Hyperactivation of PPAR signaling pathways

  • Upregulation of PPAR target genes (e.g., PLIN2, FABP3)

  • Enhanced promoter/enhancer activity at PPAR response elements

  • Growth factor-independent proliferation of bladder cells

Experimental validation has shown that these specific mutations always lead to substitution with an aromatic amino acid (phenylalanine ~5% or tyrosine ~1%), which appears critical for the observed hyperactivation .

The mechanism has been demonstrated through both transcriptomic analysis and functional studies in bladder cancer cell lines, where RXRA S427F/Y mutations cause expression changes similar to those induced by PPAR agonists .

What methodologies are recommended for studying RXRA mutation effects on transcriptional regulation?

To comprehensively study RXRA mutation effects, a multi-modal approach is recommended:

  • Transcriptomic analysis:

    • RNA-seq of cells expressing wild-type vs. mutant RXRA

    • Pathway analysis to identify enriched pathways (e.g., KEGG pathway analysis)

    • Comparison with agonist-induced expression patterns

  • Enhancer/promoter activity assessment:

    • ChIP-seq for RXRA and active enhancer marks (H3K27ac)

    • Differential analysis of enhancer/promoter activation

    • Motif enrichment analysis using tools like HOMER

  • Functional validation:

    • Luciferase reporter assays with DR1 elements

    • siRNA knockdown of potential heterodimer partners

    • Rescue experiments

This approach successfully identified that mutant RXRA (S427F/Y) specifically hyperactivates enhancers/promoters with RXR/PPAR (DR1) motifs, establishing mechanistic understanding of mutation effects .

How do RXRA mutations affect PPAR signaling and what experimental approaches can demonstrate this relationship?

RXRA S427F/Y mutations selectively enhance PPAR signaling through molecular mechanisms that can be experimentally demonstrated through:

  • Transcriptome profiling:

    • RNA-seq analysis showing upregulation of PPAR target genes

    • Comparison to PPAR agonist treatment effects (correlation analysis)

  • Partner dependency experiments:

    • siRNA knockdown of individual PPARs (PPARD, PPARG)

    • Combined knockdown to assess redundancy

    • Measurement of target gene expression (RT-qPCR)

  • Mechanistic validation in reconstituted systems:

    • Reporter assays in cells with low endogenous PPAR expression

    • Co-transfection with specific PPAR subtypes

    • Comparison with other RXRA partners (e.g., RARA)

These approaches revealed that RXRA mutations activate both PPARD and PPARG with functional redundancy, as individual knockdown had limited effects while combined knockdown strongly inhibited mutation-driven gene expression .

How can organoid models be used to study RXRA function in urothelial biology?

Organoid models provide powerful systems for studying RXRA function in a physiologically relevant context. A methodological approach includes:

  • Organoid generation:

    • Isolation of urothelial cells from appropriate mouse models

    • Culture in 3D matrices with defined media components

    • Generation of genetically defined backgrounds (e.g., tumor suppressor knockout)

  • Experimental manipulation:

    • Retroviral transduction for RXRA variant expression

    • Growth factor dependency assessment

    • PPAR agonist/antagonist treatment

  • Phenotypic assessment:

    • Growth rate measurement

    • Cell counting across multiple passages

    • Calculation of population doublings

This approach demonstrated that RXRA S427F promotes growth factor-independent growth in organoids with tumor suppressor loss (Trp53/Kdm6a null), providing a model system that recapitulates aspects of bladder cancer biology .

What are appropriate control conditions when studying RXRA mutations in experimental systems?

Proper experimental design for RXRA mutation studies requires multiple controls:

Control TypePurposeImplementation
Wild-type RXRADistinguish mutation-specific effectsExpress at equivalent levels to mutant
Empty vectorControl for overexpression artifactsUse identical vector backbone
Expression level matchingPrevent artifacts from varying expressionConfirm by qPCR and Western blot
Partner specificityDetermine heterodimer dependencyTest multiple heterodimer partners (PPARG, PPARD, RARA)
Biological replicatesEnsure reproducibilityTest in multiple cell lines or organoid preparations

Additionally, pharmacological controls comparing mutation effects to agonist treatment can provide mechanistic insights. This approach showed that RXRA S427F/Y effects mimicked PPAR agonist treatment, supporting the hypothesis of PPAR pathway hyperactivation .

How can ChIP-seq data be integrated with transcriptomic data to understand RXRA-mediated gene regulation?

Integration of ChIP-seq and transcriptomic data provides powerful insights into RXRA-mediated gene regulation:

  • Data generation:

    • ChIP-seq for RXRA (wild-type and mutant)

    • ChIP-seq for histone modifications (e.g., H3K27ac)

    • RNA-seq under matching conditions

  • Analytical approach:

    • Identify RXRA-bound regions

    • Assess differential enhancer/promoter activation (H3K27ac)

    • Correlate with differential gene expression

    • Perform motif enrichment analysis at hyperactivated sites

  • Validation experiments:

    • Reporter assays for identified regulatory elements

    • Functional studies of target genes

This integrated approach successfully identified that RXRA S427F/Y mutations selectively hyperactivate enhancers/promoters containing DR1 motifs, explaining the observed transcriptional changes in PPAR target genes .

What are the structural mechanisms by which S427F/Y mutations alter RXRA function?

The molecular mechanism of S427F/Y mutation effects involves allosteric regulation of heterodimer partners:

  • Structural features:

    • The mutation introduces an aromatic amino acid (F or Y) at position 427

    • This creates specific aromatic interactions with PPAR partners

    • Specifically, it interacts with the terminal tyrosine found in PPARs

    • This interaction allosterically regulates the PPAR AF2 domain

  • Experimental approaches for structural studies:

    • Computational simulations of RXRA-PPAR interactions

    • Structure-function analyses with targeted mutations

    • Biochemical assays measuring conformational changes

    • X-ray crystallography or cryo-EM of complexes

The specificity of the effect to PPAR partners explains why RXRA S427F/Y mutations do not enhance activation with other heterodimer partners like RARA .

How can redundancy between PPARD and PPARG be experimentally addressed in RXRA mutation studies?

Functional redundancy between PPARD and PPARG presents a research challenge that can be addressed through:

  • Expression analysis in relevant tissues:

    • Analysis of TCGA data shows that both PPARD and PPARG are expressed in bladder cancer specimens

    • qPCR validation in cell lines and primary tissues

  • Combinatorial knockdown/knockout approaches:

    • Individual vs. combined siRNA knockdown

    • CRISPR-based knockout of individual or both receptors

    • Inducible systems for temporal control

  • Isoform-specific pharmacological tools:

    • Selective agonists for PPARD vs. PPARG

    • Selective antagonists to block specific pathways

    • Combination treatments

These approaches revealed that in bladder cancer cells, both PPARD and PPARG contribute to mutant RXRA-mediated transcriptional hyperactivity, with combined knockdown having stronger effects than individual knockdown .

What considerations should inform the development of therapeutic strategies targeting RXRA-PPAR signaling in cancer?

Development of therapeutic strategies targeting RXRA-PPAR signaling requires careful consideration of several factors:

  • Target selection:

    • Approximately 20-25% of bladder cancers show hyperactive PPAR signaling

    • Multiple mechanisms: PPARG amplification (17%) or RXRA mutations (5-8%)

    • Both PPARD and PPARG may need targeting due to functional redundancy

  • Preclinical model selection:

    • Cell lines with defined RXRA/PPAR status

    • Organoid models with relevant genetic backgrounds

    • PDX models from patients with specific mutations

  • Pharmacological approach:

    • PPAR antagonists showed efficacy in reversing mutant RXRA-driven growth

    • Combined PPARD/PPARG inhibition may be necessary

    • Potential for resistance through homologue exploitation

  • Clinical translation considerations:

    • Patient selection based on RXRA mutation or PPARG amplification status

    • Biomarkers for pathway activation (e.g., PLIN2 expression)

    • Monitoring for on-target and off-target effects

The observation that mutant RXRA-driven growth of bladder organoids is reversible by PPAR inhibition provides preclinical support for PPAR targeting in RXRA-mutant bladder cancer .

How should researchers analyze RNA-seq data to identify RXRA mutation-specific gene expression signatures?

Analysis of RNA-seq data to identify RXRA mutation-specific signatures requires a structured approach:

  • Experimental design:

    • Multiple cell lines expressing wild-type vs. mutant RXRA

    • Matched expression levels (confirmed by qPCR/Western)

    • Appropriate replicates

  • Analysis pipeline:

    • Quality control and normalization

    • Differential expression analysis (e.g., DESeq2, edgeR)

    • Selection criteria (fold change ≥2, FDR <0.05)

    • Pathway enrichment analysis (ORA, GSEA)

  • Validation strategies:

    • Comparison across multiple cell lines

    • Intersection analysis for robust signatures

    • qPCR validation of selected targets

    • Comparison to agonist-induced changes

This approach successfully identified the PPAR signaling pathway (KEGG-hsa03320) as the top enriched pathway in genes upregulated by RXRA S427F mutation across multiple cell lines .

What approaches are recommended for motif analysis in RXRA ChIP-seq data?

Effective motif analysis in RXRA ChIP-seq data involves:

  • Data processing:

    • Peak calling (e.g., MACS2)

    • Differential binding analysis between conditions

    • Integration with enhancer/promoter marks (H3K27ac)

  • Motif discovery:

    • De novo motif finding (e.g., HOMER, MEME)

    • Known motif enrichment analysis

    • Comparison to established motif databases

  • Validation strategies:

    • Reporter assays with identified motifs

    • Mutation of motif sequences

    • Correlation with gene expression changes

In RXRA S427F/Y studies, motif analysis of hyperactivated regulatory elements identified the canonical RXR/PPAR (DR1) motif as significantly enriched, confirming the selective effect on PPAR-responsive elements .

How can researchers integrate multiple data types to build comprehensive models of RXRA function in disease?

Integration of multiple data types requires computational and experimental approaches:

  • Multi-omics integration:

    • ChIP-seq (protein-DNA interactions)

    • RNA-seq (transcriptional output)

    • ATAC-seq (chromatin accessibility)

    • Proteomics (protein levels and interactions)

  • Computational modeling:

    • Network analysis of regulatory relationships

    • Machine learning for pattern identification

    • Pathway modeling and simulation

  • Functional validation:

    • Targeted perturbation experiments

    • CRISPR screens for genetic dependencies

    • Pharmacological interventions

  • Clinical correlation:

    • Patient sample analysis

    • Correlation with disease phenotypes

    • Biomarker identification

This integrated approach has successfully established connections between RXRA mutations, PPAR signaling hyperactivation, and bladder cancer growth, providing a model that spans from molecular mechanism to potential therapeutic implications .

Product Science Overview

Introduction

Retinoid X Receptor Alpha (RXRα), also known as NR2B1 (nuclear receptor subfamily 2, group B, member 1), is a nuclear receptor encoded by the RXRA gene in humans . This receptor plays a crucial role in mediating the biological effects of retinoids by participating in retinoic acid-mediated gene activation .

Structure and Function

RXRα is a member of the steroid and thyroid hormone receptor superfamily of transcription factors . It functions by forming homodimers or heterodimers with other nuclear receptors, such as retinoic acid receptors (RARs), and binding to specific sequences in the promoters of target genes to regulate their transcription . The protein encoded by the RXRA gene is involved in various biological processes, including fatty acid oxidation and the cytochrome P450 system .

In the absence of a ligand, RXR-RAR heterodimers associate with a multiprotein complex containing transcription corepressors that induce histone deacetylation, chromatin condensation, and transcriptional suppression . Upon ligand binding, the corepressors dissociate from the receptors, and coactivators are recruited, leading to transcriptional activation .

Recombinant Human RXRα

Recombinant human RXRα is a fragment protein expressed in Escherichia coli, with a purity greater than 95% . This recombinant protein is used in various research applications, including SDS-PAGE, to study the receptor’s function and interactions . The high-affinity ligand for RXRs is 9-cis retinoic acid, which binds to the receptor and regulates gene expression .

Interactions

RXRα interacts with several proteins and nuclear receptors, including BCL3, BRD8, CLOCK, FXR, IGFBP3, ITGB3BP, LXR-β, MyoD, NCOA6, NFKBIB, NPAS2, NRIP1, NR4A1, NCOA2, NCOA3, POU2F1, PPARGC1A, PPAR-γ, RNF8, RAR-α, SHP, TADA3L, TBP, TRIM24, TR-β, and VDR . These interactions are essential for the receptor’s role in regulating gene expression and various biological processes.

Clinical Significance

Mutations or dysregulation of the RXRA gene have been associated with several diseases, including pancreatic adenocarcinoma and bladder urothelial carcinoma . Understanding the function and interactions of RXRα is crucial for developing targeted therapies for these conditions.

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.