RARA Human

Retinoic Acid Receptor Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction and Overview

RARA belongs to the nuclear receptor superfamily (NR1B1) and forms heterodimers with retinoid X receptors (RXRs) to bind retinoic acid response elements (RAREs) in DNA. These complexes regulate transcriptional programs essential for development, cell cycle control, and apoptosis. The RARA gene is located on chromosome 17q21.2 and spans approximately 100 kb, with alternative splicing generating multiple isoforms. Chromosomal translocations involving RARA, such as the PML-RARA fusion, are hallmark genetic lesions in acute promyelocytic leukemia (APL) .

Clinical Relevance and Research Findings

RARA Overexpression in Acute Myeloid Leukemia (AML)
Approximately 30% of non-APL AML patients exhibit RARA mRNA overexpression due to superenhancer (SE) activity at the RARA locus. This biomarker identifies patients responsive to tamibarotene, a selective RARα agonist .

Clinical Trial Outcomes (Tamibarotene + Azacitidine)

CohortCR/CRi RateCR RateTransfusion Independence
RARA-positive (n=18)61%50%72%
RARA-negative (n=28)21%14%38%

CR = Complete Remission; CRi = CR with incomplete hematologic recovery .

Mechanistic Insights

  • RARA overexpression correlates with a monocytic gene expression signature (e.g., CD14, LYZ) and predicts sensitivity to tamibarotene.

  • Tamibarotene induces differentiation of AML blasts and synergizes with azacitidine to enhance therapeutic efficacy .

Functional and Regulatory Mechanisms

Transcriptional Regulation
In the absence of retinoic acid, RARA-RXR heterodimers recruit corepressors (e.g., NCOR1, HDACs) to repress target genes. Ligand binding displaces corepressors and recruits coactivators (e.g., histone acetyltransferases), enabling transcriptional activation .

Ligands and Modulators

TypeExampleFunction
AgonistRetinoic acidActivates RARA-RXR heterodimers
AgonistTamibaroteneSelective RARα activation
AntagonistBMS-189453Inhibits RARA signaling

Assay and Research Tools

Reporter Assay Systems
INDIGO Biosciences offers a panel of human RAR reporter assays to study ligand-receptor interactions:

ComponentDescription
RARα Reporter CellsEngineered mammalian cells expressing RARα
Luciferase Detection ReagentMeasures transcriptional activity
Control AgonistAll-trans retinoic acid (ATRA)

These kits enable high-throughput screening of RARα agonists/antagonists in 96-well plate formats .

Genetic and Disease Associations

Acute Promyelocytic Leukemia (APL)
PML-RARA fusion proteins, resulting from t(15;17) translocations, disrupt RARA’s transcriptional repression, leading to granulocytic differentiation arrest. All-trans retinoic acid (ATRA) therapy induces differentiation of APL blasts .

Myelodysplastic Syndromes (MDS)
RARA overexpression, driven by SEs, is observed in ~50% of MDS cases. Targeting this pathway may offer therapeutic opportunities .

Product Specs

Introduction
Retinoic acid receptor alpha (RAR), a member of the ligand-responsive gene regulatory protein family that also includes steroid and thyroid hormone receptors, plays a crucial role in vertebrate development. Characterized by two highly conserved domains responsible for DNA and ligand binding, RAR proteins exist in three isotypes: alpha, beta, and gamma, each encoded by distinct genetic loci and exhibiting unique transcriptional properties. Notably, RAR-alpha typically represses target gene transcription in the absence of hormones, while RAR-beta and gamma do not. As a receptor for retinoic acid, a morphogen with potent teratogenic effects, RARA directly regulates gene expression to control cell function. Chromosomal abnormalities involving RARA are implicated in acute promyelocytic leukemia (APL).
Description
Recombinant human RARA, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 127 amino acids (68-173 a.a.). With a molecular mass of 14 kDa, it appears larger on SDS-PAGE due to a 21 amino acid histidine tag fused at the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The RARA protein solution 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 (2-4 weeks), the protein should be kept at 4°C. Long-term storage requires freezing at -20°C, preferably with the addition of a carrier protein (0.1% HSA or BSA) to maintain stability. Repeated freezing and thawing should be avoided.
Purity
The purity of the RARA protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Retinoic acid receptor alpha, RAR-alpha, Nuclear receptor subfamily 1 group B member 1,RAR, NR1B1, RARA.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSEEIVPSPP SPPPLPRIYK PCFVCQDKSS GYHYGVSACE GCKGFFRRSI QKNMVYTCHR DKNCIINKVT RNRCQYCRLQ KCFEVGMSKESVRNDRNKKK KEVPKPE.

Q&A

What is the basic function of human RARα in cellular signaling?

Human Retinoic Acid Receptor Alpha (RARα) functions as a nuclear hormone receptor that forms heterodimers with retinoid X receptors (RXRs). These heterodimers act as ligand-dependent transcriptional regulators by binding to specific retinoic acid response elements (RAREs) found in the promoter regions of target genes. In the absence of an RAR agonist, RXR-RAR recruits co-repressor proteins such as NCoR and associated factors including histone deacetylase to maintain a condensed chromatin structure. When an RAR agonist binds, it stimulates the release of co-repressors and recruitment of co-activator complexes, such as histone acetyltransferase, to activate transcription . This mechanism enables RARα to transduce retinoid signals that mediate critical biological processes including proper embryogenesis, cellular differentiation, and growth arrest regulation.

How does human RARα differ from other RAR subtypes?

There are three distinct RAR subtypes: RAR alpha (RARα), RAR beta (RARβ), and RAR gamma (RARγ). The primary distinction is their tissue distribution pattern. RARα is present in most tissue types, exhibiting a more ubiquitous expression pattern, whereas RARβ and RARγ expression is more selective and tissue-specific . Functionally, each RAR subtype appears to have specialized roles despite their similar mechanism of action. For instance, RXRα-RARγ heterodimers are necessary for growth arrest and visceral and primitive endodermal differentiation, while RXRα-RARα is specifically required for cAMP-dependent parietal endodermal differentiation . This functional specialization is important for researchers to consider when designing experiments targeting specific developmental or cellular processes.

What are the most reliable detection methods for studying human RARα activity?

The gold standard for studying human RARα activity is the cell-based luciferase reporter assay. This method utilizes engineered mammalian cells that provide constitutive expression of human RARα along with a luciferase reporter gene that responds to RARα activation. When RARα is activated by an agonist, it induces luciferase expression, which can be quantified to provide a sensitive surrogate measure of RARα activity in treated cells .

Alternative methods include:

  • Chromatin immunoprecipitation (ChIP) assays to detect RARα binding to DNA targets

  • Co-immunoprecipitation to study protein-protein interactions with co-activators or co-repressors

  • RT-qPCR to measure expression changes in RARα target genes

  • Western blotting to detect RARα protein levels and post-translational modifications

When selecting a detection method, researchers should consider factors such as sensitivity requirements, available equipment, and whether they need to measure direct binding, transcriptional activity, or downstream effects.

What controls should be included when studying RARα function?

When designing experiments to study RARα function, several controls are essential to ensure reliability and validity:

  • Positive controls: Include well-characterized RARα agonists such as all-trans retinoic acid (ATRA) at standardized concentrations.

  • Negative controls: Include vehicle controls (solvent used to dissolve test compounds) and potentially RARα antagonists.

  • Specificity controls: Test compounds on cells lacking RARα expression or with mutated RARα to confirm specificity of observed effects.

  • Dose-response controls: Test multiple concentrations of compounds to establish dose-dependent relationships.

  • Time-course controls: Measure responses at different time points to capture both early and late effects of RARα activation.

Proper experimental design requires careful consideration of these controls to minimize bias and ensure reproducibility. Researchers should implement randomization in assigning treatments, blinding during data collection and analysis, and include appropriate technical and biological replicates .

How can I address RARα isoform-specific functions in my research?

Investigating isoform-specific functions of RARα requires sophisticated experimental approaches:

  • CRISPR/Cas9 gene editing: Generate isoform-specific knockouts or introduce mutations in specific domains to assess their functional contributions.

  • Isoform-specific siRNA/shRNA: Design RNA interference tools targeting unique regions of specific RARα isoforms to achieve selective knockdown.

  • Isoform-specific antibodies: Utilize antibodies recognizing unique epitopes for differential detection of isoforms in Western blotting, immunoprecipitation, or ChIP assays.

  • Exogenous expression systems: Express individual isoforms in cells lacking endogenous RARα to study their functions in isolation.

  • Mass spectrometry: Employ proteomic approaches to identify isoform-specific protein interaction partners.

What are the challenges in investigating cross-talk between RARα and other nuclear receptors?

Investigating cross-talk between RARα and other nuclear receptors presents several methodological challenges:

  • Heterodimer formation complexity: RARα forms heterodimers primarily with RXRs, which themselves can partner with multiple nuclear receptors, creating competition for RXR availability. Researchers must account for this competition when designing experiments.

  • Shared co-regulators: Many nuclear receptors utilize overlapping sets of co-activators and co-repressors, making it difficult to attribute specific effects to RARα signaling versus other pathways.

  • Genomic binding site overlap: Response elements for different nuclear receptors may overlap, leading to competitive or cooperative binding that complicates interpretation.

  • Temporal dynamics: Cross-talk may be highly dependent on timing, with sequential activation of different pathways yielding different outcomes than simultaneous activation.

To address these challenges, researchers should consider:

  • Using sequential ChIP (re-ChIP) to identify genomic regions where multiple nuclear receptors co-bind

  • Employing proximity ligation assays to detect protein-protein interactions in situ

  • Utilizing selective agonists/antagonists in combination to dissect pathway interactions

  • Implementing time-course studies with high temporal resolution

  • Developing mathematical models to account for the complex dynamics of nuclear receptor signaling networks

How can I develop valid models for studying RARα in rare disease contexts?

Developing valid models for studying RARα in rare disease contexts requires specialized approaches to overcome the inherent challenges of rare disease research:

  • Patient-derived models: Establish primary cell cultures or induced pluripotent stem cells (iPSCs) from patients with rare RARα-related disorders. These can be differentiated into relevant cell types to study disease mechanisms.

  • Animal models: Generate knockin mouse models harboring specific mutations identified in human patients. Consider using conditional knockout approaches to study tissue-specific effects.

  • Organoid systems: Develop three-dimensional organoid cultures that better recapitulate the complexity of human tissues affected in rare RARα-related disorders.

  • Adaptive trial designs: For clinical studies, employ response-adaptive randomization (RAR) designs that modify treatment allocation based on observed outcomes, which is particularly valuable when studying small patient populations .

When working with rare disease models, researchers should pay special attention to:

  • Statistical power considerations, often requiring alternative analytical approaches appropriate for small sample sizes

  • Patient heterogeneity, which may necessitate stratification or individualized analyses

  • Careful phenotypic characterization to identify relevant outcome measures

  • Collaborative approaches to increase sample size and share resources

The table below outlines key considerations for different model systems in rare RARα-related disease research:

Model SystemAdvantagesLimitationsBest Applications
Patient-derived iPSCsCaptures patient-specific genetic backgroundVariability between cell lines; expensiveMechanistic studies; drug screening
Animal modelsIn vivo systemic effects; behavioral assessmentMay not fully recapitulate human diseaseDevelopmental studies; in vivo drug testing
Organoids3D architecture; multiple cell typesLacks systemic context; variableOrgan-specific effects; tissue interactions
Clinical samplesDirect human relevanceLimited availability; heterogeneityBiomarker validation; genetic studies

Researchers should consider using multiple complementary models to strengthen the validity of their findings .

What are the optimal cell models for studying human RARα function?

Selecting the appropriate cell model is critical for studying human RARα function. Consider these options based on your research questions:

  • Established cell lines expressing endogenous RARα:

    • MCF-7 (breast cancer): High endogenous expression of RARα; good for studying natural regulation

    • HL-60 (promyelocytic leukemia): Responds to retinoic acid with differentiation; ideal for studying RARα in hematopoietic differentiation

    • SH-SY5Y (neuroblastoma): Useful for studying RARα in neuronal contexts

  • Genetically modified reporter cell lines:

    • Engineered cells with stably integrated RARα-responsive luciferase reporter constructs provide sensitive, quantitative readouts of RARα activity

    • These systems can be optimized for high-throughput screening applications

  • Primary cells:

    • Primary human keratinocytes, hepatocytes, or neuronal cells provide physiologically relevant contexts

    • More challenging to work with but offer greater translational relevance

  • iPSC-derived models:

    • Allow study of RARα in development and differentiation

    • Can be generated from patients with RARα-related disorders

When selecting a model, consider:

  • The particular aspects of RARα biology you're investigating

  • Whether endogenous or overexpressed RARα is more appropriate

  • Required sensitivity for your detection methods

  • Availability of model-specific reagents and protocols

  • Relevance to your disease or developmental context of interest

For optimal reproducibility, characterize your chosen model thoroughly, including baseline RARα expression levels, response to known agonists, and expression of RARα target genes .

How should I design experiments to minimize bias in RARα research?

Minimizing bias in RARα research requires rigorous experimental design incorporating several key practices:

  • Randomization: Randomly assign experimental units to treatment groups to prevent selection bias. This includes randomizing:

    • Order of sample processing

    • Well positions in multi-well plates

    • Animal allocation to treatment groups

    • Order of data collection

  • Blinding: Whenever possible, ensure that investigators are unaware of:

    • Treatment group assignments during data collection

    • Sample identities during analysis

    • Expected outcomes when interpreting results

  • Proper controls:

    • Include positive controls (known RARα agonists)

    • Include negative controls (vehicle, inactive compounds)

    • Consider including tool compounds with known mechanisms for comparison

    • Use appropriate genetic controls (e.g., RARα knockout or knockdown)

  • Replication:

    • Include sufficient technical replicates to account for assay variability

    • Conduct biological replication with independent experiments

    • Consider replication across different cell lines or model systems

  • Sample size determination:

    • Conduct power analyses to determine appropriate sample sizes

    • For rare disease research, consider alternative statistical approaches suitable for small sample sizes

  • Standardized protocols:

    • Develop and adhere to detailed standard operating procedures

    • Control for environmental variables (temperature, humidity, time of day)

    • Standardize cell culture conditions (passage number, confluence)

  • Reporting:

    • Follow field-specific reporting guidelines

    • Report all experimental conditions in detail

    • Disclose any deviations from pre-specified protocols

    • Report all results, including negative or inconclusive findings

Implementing these practices will significantly enhance the reliability and reproducibility of RARα research findings, addressing known issues in experimental rigor that have been identified across biomedical research .

What are the best approaches for studying RARα in limited patient samples?

When studying RARα in limited patient samples, researchers must maximize the information obtained while minimizing sample use. Consider these approaches:

  • Multiplexed analyses: Implement techniques that allow simultaneous measurement of multiple parameters from a single sample:

    • Multi-parameter flow cytometry for protein expression

    • Single-cell RNA sequencing for transcriptomic profiles

    • Mass cytometry (CyTOF) for protein expression and modification

    • Multiplex immunoassays for multiple protein analytes

  • Microfluidic technologies: Utilize platforms that require minimal sample input:

    • Droplet-based single-cell analysis

    • Microfluidic qPCR for gene expression

    • Organ-on-a-chip systems for functional studies

  • Ex vivo expansion: When possible, expand limited primary cells:

    • Conditional reprogramming for epithelial cells

    • Ex vivo expansion protocols for immune cells

    • Patient-derived organoids that can be passaged

  • Optimized study design: Employ statistical approaches suited for small sample sizes:

    • N-of-1 trial designs for individual patients

    • Bayesian approaches that can incorporate prior knowledge

    • Response-adaptive randomization when conducting intervention studies

  • Non-invasive alternatives: Consider surrogate measures or samples:

    • Liquid biopsies (blood, urine) for circulating markers

    • Imaging-based endpoints when appropriate

    • Patient-derived iPSCs as renewable surrogates

  • Sample sharing and biobanking: Establish collaborations with:

    • Rare disease registries and biobanks

    • Multi-center research consortia

    • Patient advocacy organizations

When working with limited samples, careful pre-analytical planning is essential. Develop detailed protocols for sample collection, processing, and storage to minimize variability and maximize sample utility. Additionally, consider sequential or staged analysis approaches, where initial results guide subsequent analyses to focus on the most promising directions .

How can I address contradictory findings in RARα research literature?

Addressing contradictory findings in RARα research literature requires a systematic approach to evaluate and reconcile disparate results:

  • Conduct a methodological assessment:

    • Compare experimental designs, including cell types, assay systems, and reagents used

    • Evaluate statistical approaches and power calculations

    • Assess for potential sources of bias or confounding variables

    • Consider differences in RARα isoforms studied

  • Perform independent replication:

    • Reproduce key experiments using multiple methodologies

    • Include appropriate positive and negative controls

    • Consider blinded analysis to minimize confirmation bias

    • Systematically vary experimental conditions to identify context-dependent effects

  • Meta-analytical approaches:

    • Conduct formal meta-analyses when sufficient studies exist

    • Use forest plots to visualize the range of reported effect sizes

    • Assess for publication bias using funnel plots

    • Consider Bayesian approaches to integrate prior knowledge with new data

  • Collaborative resolution:

    • Establish direct collaboration with labs reporting contradictory findings

    • Exchange materials, protocols, and data

    • Conduct parallel experiments with standardized methods

    • Consider multi-laboratory validation studies

  • Consider biological explanations:

    • RARα functions may be context-dependent (cell type, developmental stage)

    • Genetic background differences may explain varying results

    • Environmental factors may influence outcomes

    • Post-translational modifications may alter RARα function in different systems

When reporting your findings in relation to contradictory literature, clearly articulate the methodological differences that may explain discrepancies, avoid selective citation of supporting studies, and present balanced interpretations that acknowledge the complexity of RARα biology .

What statistical approaches are most appropriate for analyzing RARα reporter assay data?

  • Dose-response modeling:

    • Use nonlinear regression to fit dose-response curves (typically sigmoidal)

    • Determine key parameters including EC50/IC50, maximum effect (Emax), and hill slope

    • Compare curves using extra sum-of-squares F-test or AIC/BIC criteria

    • Consider using the four-parameter logistic model: Y = Bottom + (Top-Bottom)/(1+10^((LogEC50-X)*HillSlope))

  • Normalization approaches:

    • Normalize to appropriate controls (vehicle, positive control)

    • Consider percent of maximum response normalization for comparison across experiments

    • Account for background signal and potential signal drift

  • Transformation considerations:

    • Log-transform concentration values to achieve more uniform variance

    • Consider Box-Cox or other transformations if data violate normality assumptions

    • Report both raw and transformed/normalized data for transparency

  • Statistical comparisons:

    • Use ANOVA with appropriate post-hoc tests for comparing multiple groups

    • Consider repeated measures designs when appropriate

    • Apply Bonferroni or other multiple comparison corrections

    • For high-throughput screening, use robust Z-factor and strictly standardized mean difference (SSMD) to assess assay quality

  • Handling outliers:

    • Pre-specify outlier identification criteria

    • Consider robust statistical methods less sensitive to outliers

    • Document and justify any exclusion of data points

  • Sample size and power:

    • Conduct power analyses to determine appropriate replicate numbers

    • Consider sequential design approaches to optimize resource use

For rare disease applications with limited samples, consider:

  • Bayesian methods that can incorporate prior knowledge

  • Permutation tests that make fewer distributional assumptions

  • Response-adaptive designs that can maximize information from small samples

Always report your statistical approach in detail, including software used, model specifications, and handling of unusual data points to ensure reproducibility .

What are the best practices for integrating RARα genomic data across multiple platforms?

Integrating RARα genomic data across multiple platforms requires careful consideration of data compatibility, normalization methods, and integrated analysis approaches:

  • Data harmonization:

    • Ensure consistent genome assemblies and annotation versions across datasets

    • Convert all raw data to comparable formats

    • Standardize gene/protein identifiers across platforms

    • Carefully document batch effects and technical variables

  • Cross-platform normalization:

    • Apply platform-specific normalizations first (e.g., TMM for RNA-seq, quantile normalization for microarrays)

    • Consider ComBat or similar methods for batch effect correction

    • Use reference samples processed on multiple platforms as anchors for cross-platform normalization

    • Validate normalization by examining housekeeping genes or spike-in controls

  • Multi-omics integration methods:

    • Correlation-based approaches: Canonical correlation analysis (CCA), sparse CCA

    • Factor-based methods: Multi-omics factor analysis (MOFA), joint non-negative matrix factorization

    • Network-based integration: Similarity network fusion, multi-layer networks

    • Pathway-based integration: Gene set enrichment analysis across platforms

  • Validation strategies:

    • Cross-validation approaches to assess model stability

    • Independent cohort validation when possible

    • Experimental validation of key findings using orthogonal methods

    • Sensitivity analyses with different integration parameters

  • Visualization for integrated data:

    • Multi-dimensional reduction techniques (t-SNE, UMAP) applied to integrated data

    • Circos plots for genome-wide integration

    • Heatmaps with hierarchical clustering across data types

    • Network visualizations showing cross-platform connections

  • Software and pipelines:

    • MultiAssayExperiment or SummarizedExperiment frameworks in R

    • MixOmics package for integrative analysis

    • Seurat for single-cell multi-omics integration

    • Galaxy workflows for reproducible analysis

When integrating RARα ChIP-seq data with transcriptomics or epigenomics, consider:

  • Time-course analyses to capture dynamics of RARα binding and subsequent effects

  • Motif enrichment analyses to identify cooperative factors

  • Integration with Hi-C or ChIA-PET data to understand 3D chromatin context

  • Comparison with public datasets (e.g., ENCODE, Roadmap Epigenomics) for context

Document all integration steps thoroughly to ensure reproducibility and allow others to build upon your analytical framework .

How can I develop valid biomarkers for RARα pathway activation in patient samples?

Developing valid biomarkers for RARα pathway activation in patient samples requires a systematic approach:

  • Biomarker discovery phase:

    • Conduct transcriptomic profiling to identify genes directly regulated by RARα

    • Perform proteomic analyses to identify proteins whose levels change in response to RARα activation

    • Use ChIP-seq data to identify direct RARα targets with consistent regulation

    • Compare responses across multiple cell types to identify robust markers

  • Assay development and validation:

    • Select candidate biomarkers based on magnitude of response, consistency, and biological relevance

    • Develop quantitative assays (qPCR, ELISA, immunohistochemistry) for selected markers

    • Validate assays for analytical performance (precision, accuracy, sensitivity, specificity)

    • Determine reference ranges in healthy controls

  • Clinical validation:

    • Test biomarkers in samples from patients with known RARα pathway dysregulation

    • Assess correlation with disease severity or treatment response

    • Evaluate biomarker performance across diverse patient populations

    • Conduct longitudinal studies to assess temporal dynamics

  • Consideration for different sample types:

    • Tissue biopsies: Provide direct assessment but invasive

    • Blood: Less invasive but may show diluted signal

    • Urine/other fluids: Non-invasive but further removed from primary site

    • Consider creating biomarker panels that work across multiple sample types

  • Implementation considerations:

    • Develop standardized protocols for sample collection and processing

    • Establish quality control procedures

    • Create clear criteria for interpretation of results

    • Ensure assay transferability across different laboratories

When working with rare diseases, consider:

  • Partnering with patient registries to access larger sample collections

  • Using adaptive trial designs to validate biomarkers in intervention studies

  • Developing composite endpoints that combine multiple biomarkers for increased sensitivity

  • Creating patient-derived cell models to test biomarker performance in controlled settings

What methodological approaches can overcome challenges in RARα-targeted drug discovery?

RARα-targeted drug discovery presents unique challenges requiring specialized methodological approaches:

  • Target engagement assays:

    • Develop cellular thermal shift assays (CETSA) to measure direct binding in cellular contexts

    • Implement time-resolved fluorescence resonance energy transfer (TR-FRET) assays for high-throughput screening

    • Utilize hydrogen-deuterium exchange mass spectrometry to map ligand binding sites

    • Consider bioluminescence resonance energy transfer (BRET) systems for real-time monitoring

  • Selectivity profiling:

    • Screen against all RAR and RXR subtypes to assess selectivity

    • Use structural biology approaches (X-ray crystallography, cryo-EM) to guide selective design

    • Implement transcriptomic profiling to compare activation patterns with subtype-selective reference compounds

    • Develop cellular models expressing individual RAR subtypes for comparative assessment

  • Functional characterization:

    • Use reporter assays to distinguish full agonists, partial agonists, and antagonists

    • Implement ChIP-seq to map genome-wide binding patterns induced by compounds

    • Assess recruitment of co-activators/co-repressors using mammalian two-hybrid systems

    • Evaluate compound effects on RARα protein stability and post-translational modifications

  • Pharmacokinetic/pharmacodynamic (PK/PD) considerations:

    • Develop mechanism-based PK/PD models specific to nuclear receptor biology

    • Implement target engagement biomarkers for in vivo dose setting

    • Consider tissue-specific delivery strategies for targeted effects

    • Evaluate compounds in physiologically-based pharmacokinetic (PBPK) models

  • Disease-relevant models:

    • Utilize patient-derived cell models for compound testing

    • Develop disease-specific assays that capture relevant aspects of RARα biology

    • Consider organoid or microphysiological systems for more complex tissue contexts

    • Implement in vivo models that accurately recapitulate disease phenotypes

For rare disease applications, consider:

  • Repurposing existing RARα modulators with known safety profiles

  • Implementing adaptive trial designs to efficiently evaluate compounds in small populations

  • Developing precision medicine approaches based on patient-specific RARα pathway alterations

  • Creating multi-targeted compounds that modulate RARα along with complementary pathways

How can I design ethical and effective clinical trials for rare diseases involving RARα dysfunction?

Designing ethical and effective clinical trials for rare diseases involving RARα dysfunction requires specialized approaches to address the challenges of small patient populations:

  • Innovative trial designs:

    • Adaptive designs: Modify aspects of the trial based on interim results, including sample size, randomization ratios, or treatment arms

    • N-of-1 trials: Conduct multiple crossovers between treatments in individual patients

    • Platform trials: Test multiple treatments simultaneously with a shared control group

    • Basket trials: Enroll patients based on molecular characteristics rather than traditional disease classifications

  • Endpoint selection and development:

    • Identify sensitive biomarkers that can serve as surrogate endpoints

    • Develop composite endpoints that combine multiple disease aspects

    • Consider patient-reported outcomes specific to the condition

    • Establish the minimal clinically important difference for selected endpoints

  • Statistical considerations:

    • Implement Bayesian methods that can incorporate prior information

    • Consider response-adaptive randomization to maximize patients receiving effective treatment

    • Use historical controls when appropriate

    • Plan for sequential analyses with appropriate alpha spending

  • Patient engagement strategies:

    • Involve patient advocacy organizations in trial design

    • Implement decentralized trial elements to reduce participation burden

    • Provide transparent communication throughout the trial

    • Consider patient preferences in outcome selection

  • Ethical considerations specific to rare diseases:

    • Balance equipoise with the need for treatment access

    • Implement extended access protocols when appropriate

    • Consider crossover designs to ensure all participants eventually receive active treatment

    • Plan for long-term follow-up to assess durability of effects

The following table outlines key trial design options and their applications in rare RARα-related diseases:

Trial DesignKey FeaturesBest Application ScenariosStatistical Considerations
Response-adaptive randomizationAdjusts allocation ratios based on interim resultsWhen early signals of efficacy are expectedRequires careful alpha spending and simulation-based planning
Simon two-stage designStops for futility after first stage if insufficient responseEarly-phase efficacy trialsPre-specified go/no-go criteria based on response rate
Bayesian basket trialGroups patients by molecular characteristics across diagnosesWhen RARα dysfunction spans multiple rare conditionsBorrowing strength across cohorts with hierarchical modeling
Seamless Phase I/IITransitions from dose-finding to preliminary efficacyLimited patient populations where separate trials are impracticalComplex modeling of dose-response relationships

Researchers should document their rationale for trial design choices and ensure transparent reporting of results, including limitations and generalizability considerations .

Product Science Overview

Introduction

Retinoic Acid Receptor Alpha (RARα), also known as NR1B1, is a nuclear receptor encoded by the RARA gene in humans. This receptor plays a crucial role in mediating the effects of retinoic acid, a derivative of vitamin A, which is essential for various biological processes including cell growth, differentiation, and embryonic development .

Structure and Function

RARα is part of the nuclear receptor subfamily 1, group B, member 1. It functions as a transcription factor that regulates gene expression by binding to specific DNA sequences. The receptor forms heterodimers with retinoid X receptors (RXRs), and in the absence of a ligand, these heterodimers repress transcription by recruiting corepressors such as NCOR1, SMRT (NCOR2), and histone deacetylases .

When retinoic acid binds to RARα, it induces a conformational change that allows the recruitment of coactivators, histone acetyltransferases, and the basic transcription machinery. This binding initiates the transcription of target genes, leading to various physiological effects .

Biological Significance

Retinoic acid signaling, mediated by RARα, is vital for several developmental processes:

  • Embryonic Development: RARα is involved in the formation of the embryonic axis, neural differentiation, cardiogenesis, and the development of organs such as the pancreas, kidneys, lungs, and extremities .
  • Cell Growth and Differentiation: Retinoic acid influences cell proliferation and differentiation, making RARα crucial for maintaining normal cellular functions .
Clinical Significance

Mutations or translocations involving the RARA gene are associated with acute promyelocytic leukemia (APL). The most common translocation is t(15;17)(q21;q22), which fuses the RARA gene with the PML gene, leading to the development of APL .

Recombinant RARα

Human recombinant RARα is produced using recombinant DNA technology, which involves inserting the RARA gene into a suitable expression system, such as bacteria or mammalian cells. This allows for the production of large quantities of the receptor for research and therapeutic purposes.

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.