TBL1XR1-B is implicated in:
Transcriptional Regulation: Acts as an adaptor for ubiquitin-proteasome-mediated exchange of NCoR/SMRT corepressors for coactivators in nuclear receptor signaling .
Wnt/β-Catenin Signaling: Facilitates β-catenin recruitment to TCF/LEF target promoters, enhancing oncogenic transcription in cancers .
NF-κB Activation: Cooperates with TBL1X to mediate SMRT phosphorylation and coactivator recruitment during immune responses .
In Xenopus, TBL1XR1 orthologs are essential for developmental gene regulation, though specific studies on the B isoform remain limited compared to human TBL1XR1 .
The partial recombinant TBL1XR1-B is typically used for:
Mechanistic Studies: Investigating WD40 domain interactions via co-immunoprecipitation .
Signaling Pathway Analysis: Reconstituting β-catenin-dependent transcription in vitro .
Antibody Production: Serving as an antigen for generating isoform-specific probes .
A commercially available Xenopus TBL1XR1-A partial recombinant protein (MBS1453909) shares 94% sequence identity with the human N-terminus, suggesting similar utility for TBL1XR1-B studies .
While direct links to Xenopus disease models are unreported, human TBL1XR1 dysregulation is associated with:
Cancer: Amplifications in 15% of prostate cancers drive androgen receptor hyperactivity .
Lymphoma: Mutations (e.g., D370Y) impair plasma cell differentiation, promoting memory B-cell reentry into germinal centers .
Neurodevelopmental Disorders: Germline mutations cause Pierpont syndrome (facial dysmorphism, intellectual disability) .
Isoform-specific functions of TBL1XR1-B versus TBL1XR1-A in Xenopus.
Structural characterization of the partial recombinant protein’s active regions.
In vivo validation of its role in embryogenesis or metamorphosis.
F-box-like protein functioning as an integral component of the N-CoR transcriptional corepressor complex. It likely regulates transcription activation mediated by nuclear receptors. It may mediate the recruitment of the 19S proteasome complex, leading to subsequent proteasomal degradation of the N-CoR complex, thus enabling cofactor exchange and transcription activation.
KEGG: xla:779369
UniGene: Xl.57610
The Xenopus laevis TBL1XR1-B is a 522 amino acid protein with a molecular mass of approximately 56.3 kDa. It contains a distinctive domain architecture featuring:
An N-terminal LisH (Lis1 homology) domain
An F-box-like domain adjacent to the LisH domain
Seven WD40 repeats at the C-terminus that form a β-propeller structure
This structure is evolutionarily conserved across species, with the WD40 repeats being particularly important for mediating protein-protein interactions. The WD40 domain in TBL1XR1 forms a barrel-like structure with aromatic residues exposed on the surface that are critical for its function .
TBL1XR1 belongs to the larger WD repeat EBI family of proteins. The high degree of conservation between Xenopus and human TBL1XR1 suggests fundamental functional importance across vertebrate species .
TBL1XR1-B functions as an integral component of the N-CoR (Nuclear receptor Corepressor) transcriptional repressor complex in Xenopus laevis. Its primary functions include:
Regulation of transcriptional repression mediated by unliganded nuclear receptors
Facilitation of the exchange between corepressors and coactivators during transcriptional regulation
Recruitment of the 19S proteasome complex that mediates the degradation of the N-CoR complex
Participation in multiple signaling pathways essential for amphibian development
In Xenopus specifically, TBL1XR1 plays a role in thyroid hormone-regulated processes during metamorphosis, acting as a transcriptional bridge between hormone signals and developmental responses .
To evaluate the transcriptional regulatory functions of recombinant TBL1XR1-B, researchers should implement these methodological approaches:
Co-repressor Complex Assembly Assays:
Co-immunoprecipitation experiments to assess interactions with N-CoR/SMRT complex components
Size exclusion chromatography to verify complex formation
FRET or BiFC assays to visualize protein-protein interactions in live cells
Transcriptional Repression Assays:
Luciferase reporter assays using promoters known to be regulated by nuclear receptors
ChIP assays to detect TBL1XR1-B recruitment to specific genomic loci
RNA-seq to identify genes differentially regulated following TBL1XR1-B overexpression or depletion
Proteasome Recruitment Analysis:
Ubiquitination assays to assess the ability of TBL1XR1-B to promote substrate ubiquitination
Proteasome activity assays in the presence/absence of TBL1XR1-B
Co-localization studies with proteasome components using fluorescence microscopy
Nuclear Receptor Interaction Studies:
GST pull-down assays with various nuclear receptors
Mammalian two-hybrid assays to study TBL1XR1-nuclear receptor interactions
Competitive binding assays to assess coregulator exchange dynamics
These assays should be calibrated against known TBL1XR1 activities in other species as benchmarks for functional conservation .
Mutations in mammalian TBL1XR1 have significant effects on germinal center (GC) development with potential implications for comparative amphibian studies:
Observed Mammalian Phenotypes:
TBL1XR1 mutations impair rather than enhance GC reactions
Mutations lead to decreased GC B-cell abundance and premature GC resolution
Reduced proliferation of GC B-cells without affecting apoptosis rates
TBL1XR1 mutations skew humoral immune response toward generating abnormal immature memory B-cells while impairing plasma cell differentiation
Molecular Mechanisms:
TBL1XR1 mutants co-opt SMRT/HDAC3 repressor complexes toward binding the memory B-cell transcription factor BACH2
This occurs at the expense of interactions with germinal center transcription factor BCL6
Results in pre-memory transcriptional reprogramming and cell-fate bias
Mutant memory B-cells fail to differentiate into plasma cells upon antigen recall
Mutation Characteristics:
TBL1XR1 mutations in lymphoma occur as heterozygous missense alleles (5-10% of DLBCL cases)
Mutations primarily affect the WD40 domain, specifically aromatic residues exposed on the barrel structure
Evidence suggests mutations function as dominant-negative loss-of-function alleles
Implications for Amphibian Research:
Amphibian TBL1XR1 likely plays conserved roles in immune cell development
Xenopus models could be valuable for studying evolutionary conservation of TBL1XR1 functions in lymphocyte development
Comparing amphibian vs. mammalian TBL1XR1 could reveal fundamental versus specialized immune regulatory functions
This comparison provides a framework for investigating whether TBL1XR1 functions are conserved in amphibian immune system development and how evolutionary divergence might have shaped specialized roles .
TBL1XR1 plays a crucial role in thyroid hormone signaling during Xenopus metamorphosis, with interesting parallels and differences compared to mammals:
Xenopus-Specific Regulation:
In Xenopus laevis, TBL1XR1 is duplicated due to the tetraploid genome
Both copies are regulated by thyroid hormone during metamorphosis
A consensus thyroid hormone response element (TRE) lies far upstream of the transcriptional start site of both genes
This TRE is nested within a 200-bp region of high sequence conservation between the duplicated genes
The TRE acts as a strong response element in transfection assays in both heterologous promoters and native contexts
Functional Role During Metamorphosis:
TBL1XR1 appears to be one of the earliest thyroid hormone-induced genes in tadpoles
It functions as a transcription factor predicted to play an important role in downstream gene regulation
This regulation leads to growth and remodeling of tissues during metamorphosis
The regulation mechanism involves xBTEB1, a Xenopus homolog of basic transcription element-binding protein 1
Comparative Aspects with Mammals:
Both amphibian and mammalian TBL1XR1 are components of nuclear receptor co-repressor complexes
In both systems, TBL1XR1 modulates the repression/activation switch of nuclear receptor signaling
The evolutionary conservation of TRE elements suggests fundamental importance in hormone responsiveness
Mammalian systems lack the metamorphosis-specific developmental transitions seen in amphibians
This comparative analysis highlights the adaptation of a conserved transcriptional regulatory protein to species-specific developmental programs while maintaining core molecular functions .
TBL1XR1 mutations contribute to lymphomagenesis through specific molecular mechanisms that could potentially be modeled using Xenopus TBL1XR1-B:
Mutation Patterns in Lymphoma:
TBL1XR1 mutations occur in 5-10% of diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma cases
Mutations are more frequent in ABC-DLBCL and are highly enriched in the MCD subtype
Mutations often co-occur with MYD88 mutations
Mutations primarily affect the WD40 domain with a predilection for aromatic residues that mediate protein-protein interactions
Functional Consequences Leading to Lymphomagenesis:
TBL1XR1 mutations drive extranodal lymphoma by disrupting normal B-cell differentiation
Mutant TBL1XR1 skews the humoral immune response toward generating abnormal immature memory B-cells
Upon antigen recall, TBL1XR1 mutant memory B-cells fail to differentiate into plasma cells
Instead, these cells preferentially reenter new germinal center reactions, creating a cyclic reentry lymphomagenesis mechanism
Molecular Mechanisms:
TBL1XR1 mutants redirect SMRT/HDAC3 repressor complexes toward binding the memory B-cell transcription factor BACH2
This occurs at the expense of interactions with the germinal center transcription factor BCL6
The resulting pre-memory transcriptional reprogramming leads to cell-fate bias and impaired plasma cell differentiation
TBL1XR1 mutations appear to function as dominant-negative loss-of-function alleles
Xenopus Model System Potential:
Recombinant Xenopus TBL1XR1-B could be engineered with equivalent mutations found in human lymphomas
The evolutionary conservation of TBL1XR1 structure suggests functional mutations would have similar effects
Xenopus B-cell development could serve as a simplified model system to study the fundamental aspects of TBL1XR1 function
Transgenic Xenopus models expressing mutant TBL1XR1-B could provide insights into early developmental effects of these mutations
A comparative approach using both mammalian and amphibian systems could provide complementary insights into the pathological mechanisms of TBL1XR1 mutations in lymphomagenesis .
Advanced experimental strategies to map the Xenopus TBL1XR1-B interactome include:
Unbiased Interactome Analysis:
Affinity purification coupled with mass spectrometry (AP-MS) using tagged recombinant TBL1XR1-B
BioID or APEX proximity labeling in Xenopus cell systems to capture transient interactions
Cross-species yeast two-hybrid screening using Xenopus TBL1XR1-B as bait against cDNA libraries
Domain-Specific Interaction Mapping:
Generation of domain deletion/mutation constructs (ΔLisH, ΔF-box, ΔWD40) to identify domain-specific interactors
Peptide array analysis to map specific binding interfaces within TBL1XR1-B
Competitive binding assays to identify differential binding partners between domains
Developmental Stage-Specific Analysis:
Temporal interactome analysis across key Xenopus developmental stages, particularly during metamorphosis
Comparison with equivalent mammalian developmental transitions
Correlation of interactome shifts with changes in gene expression
Comparative Interactome Analysis:
Direct comparison between Xenopus TBL1XR1-B and human TBL1XR1 interactomes under equivalent conditions
Identification of conserved versus species-specific interactors
Functional validation of key interactions using knockdown/rescue experiments
Data Analysis Framework:
Network analysis to identify interaction clusters and functional modules
Enrichment analysis for biological processes and signaling pathways
Evolutionary conservation scoring of identified interactions
This comprehensive approach would provide insights into both conserved functions of TBL1XR1 and adaptations specific to amphibian biology, particularly in developmental contexts and nuclear receptor signaling pathways .
Structural studies of TBL1XR1-B WD40 repeats can significantly advance therapeutic design through:
High-Resolution Structural Analysis:
X-ray crystallography or cryo-EM structures of Xenopus TBL1XR1-B WD40 domain
Comparison with mammalian structures to identify conserved binding pockets
Molecular dynamics simulations to understand conformational flexibility
Identification of "hotspot" residues that mediate critical protein-protein interactions
Structure-Function Relationship Assessment:
Analysis of disease-associated mutations mapped onto the WD40 structure
Characterization of how mutations in aromatic residues on the WD40 barrel surface affect protein binding
Comparison between wild-type and mutant structures to identify conformational changes
Therapeutic Target Identification:
Mapping of druggable pockets within the WD40 domain
Virtual screening against identified pockets to discover potential binding molecules
Fragment-based drug discovery approaches focused on the protein-protein interaction surfaces
Rational Drug Design Strategy:
Development of small molecules that can:
Disrupt pathological interactions of mutant TBL1XR1
Restore normal binding profiles in disease states
Stabilize productive conformations of the WD40 domain
Peptide-based inhibitors designed to mimic natural binding partners
Translation to Human Applications:
Comparative analysis of binding affinities between amphibian and human proteins
Assessment of species-specific differences in drug binding and efficacy
Development of broad-spectrum versus selective therapeutic agents
This structure-based approach leverages the evolutionary conservation of TBL1XR1 to develop potential therapeutics for TBL1XR1-associated pathologies including lymphomas, intellectual disability syndromes, and potentially other cancers where TBL1XR1 is dysregulated .
Researchers frequently encounter several technical challenges when working with recombinant Xenopus TBL1XR1-B. Here are the most common issues and evidence-based solutions:
Protein Solubility Issues:
Challenge: TBL1XR1-B with its multiple WD40 repeats often forms inclusion bodies in bacterial expression systems.
Solution:
Use lower induction temperatures (16-18°C) and reduced IPTG concentrations (0.1-0.5 mM)
Express as fusion proteins with solubility tags (MBP, SUMO, or TrxA)
Consider insect cell or mammalian expression systems for improved folding
Include low concentrations (1-5%) of non-ionic detergents during purification
Protein Stability Concerns:
Challenge: The multi-domain structure of TBL1XR1-B can lead to degradation during purification.
Solution:
Include protease inhibitor cocktails throughout purification
Maintain samples at 4°C and minimize freeze-thaw cycles
Consider addition of stabilizing agents (5-10% glycerol, 1-5 mM DTT)
Screen buffer conditions using thermal shift assays to identify optimal stability conditions
Complex Formation Difficulties:
Challenge: Reconstituting functional complexes with N-CoR/SMRT components.
Solution:
Co-expression of interaction partners in the same system
Stepwise assembly of complexes with controlled stoichiometry
Validation of complex formation using analytical size exclusion chromatography
Implementation of gradient purification techniques to isolate intact complexes
Activity Assessment Problems:
Challenge: Verifying that recombinant TBL1XR1-B retains native functionality.
Solution:
Compare activity to known mammalian orthologs as benchmarks
Develop robust in vitro assays for specific functions (co-repressor binding, proteasome recruitment)
Use rescue experiments in TBL1XR1-depleted systems to verify functionality
Check for proper post-translational modifications that might affect function
These methodological solutions are based on general principles for working with complex multi-domain proteins containing WD40 repeats and should be optimized for specific experimental conditions and research objectives .
Distinguishing between wild-type and mutant TBL1XR1-B effects requires rigorous experimental design:
Molecular and Biochemical Differentiation:
Binding Affinity Analysis:
Quantitative interaction studies (SPR, ITC, MST) to measure differences in binding affinities to key partners
Competition assays between wild-type and mutant proteins for limiting interaction partners
Detailed kinetic analyses to identify differences in association/dissociation rates
Structural Analysis:
Comparative circular dichroism to detect secondary structure differences
Thermal shift assays to identify stability differences
Limited proteolysis to detect conformational changes between wild-type and mutant forms
Cellular and Functional Differentiation:
Transcriptional Readouts:
RNA-seq analysis comparing global transcriptional effects of wild-type vs. mutant TBL1XR1-B
ChIP-seq to identify differential genomic binding sites
Reporter gene assays using known TBL1XR1-responsive promoters
Protein Complex Analysis:
IP-MS to identify differential interactomes between wild-type and mutant proteins
Size exclusion chromatography to detect differences in complex formation
Live-cell imaging with fluorescently tagged proteins to observe differences in localization or dynamics
Experimental Controls and Validation:
Rescue Experiments:
Depletion of endogenous TBL1XR1 followed by complementation with either wild-type or mutant forms
Quantitative assessment of rescue efficiency across multiple phenotypic readouts
Dose-response studies to detect dominant-negative effects of mutants
Domain Swap/Chimeric Proteins:
Creation of chimeric proteins to map functionally important regions
Point-mutation series to identify critical residues
Structure-guided mutagenesis to confirm mechanism-based hypotheses
Based on lymphoma studies, researchers should particularly focus on:
Germinal center development phenotypes
B-cell differentiation markers
Interactions with SMRT/HDAC3 complexes
Binding to transcription factors BCL6 and BACH2
Cell proliferation without effects on apoptosis
These approaches provide a framework for distinguishing the subtle but important differences between wild-type and mutant TBL1XR1-B in experimental systems .
Several cutting-edge technologies show promise for advancing TBL1XR1-B research:
CRISPR-Based Technologies:
Base and Prime Editing: For precise introduction of disease-relevant mutations in Xenopus models
CRISPRi/CRISPRa: For temporal and spatial control of TBL1XR1-B expression
CRISPR Screens: To identify synthetic lethal interactions with TBL1XR1-B mutations
Single-Cell Omics Approaches:
scRNA-seq: To resolve cell-type specific effects of TBL1XR1-B modulation during development
scATAC-seq: To map chromatin accessibility changes governed by TBL1XR1-B activity
Spatial Transcriptomics: To map TBL1XR1-B activity in tissue contexts during amphibian development
Multi-omics Integration: To correlate TBL1XR1-B activity with epigenetic and transcriptional outcomes
Advanced Structural Biology Methods:
Cryo-EM: For visualization of complete TBL1XR1-containing complexes
Hydrogen-Deuterium Exchange MS: To map dynamic interactions and conformational changes
Integrative Structural Biology: Combining multiple techniques to build comprehensive structural models
Live Imaging Technologies:
Optogenetic Control: For temporal manipulation of TBL1XR1-B activity
Biosensors: To visualize TBL1XR1-B interactions and activity in living cells and tissues
Super-Resolution Microscopy: To visualize TBL1XR1-B-containing complexes at near-molecular resolution
Computational and AI-Based Approaches:
Deep Learning: For prediction of TBL1XR1-B interaction networks and functional outcomes
Molecular Dynamics Simulations: To model the effects of mutations on protein behavior
Network Analysis: To position TBL1XR1-B within developmental and disease-relevant pathways
These technologies, particularly when applied to comparative studies between amphibian and mammalian systems, could provide unprecedented insights into the fundamental biology of TBL1XR1 and its role in disease processes like lymphomagenesis and developmental disorders .
Comparative studies between amphibian and mammalian TBL1XR1 offer unique insights into the evolution of nuclear receptor signaling:
Evolutionary Conservation Analysis:
Sequence Conservation Patterns:
Identification of invariant residues across species suggesting fundamental functional importance
Analysis of lineage-specific adaptations in TBL1XR1 structure
Mapping of evolutionary rates across different protein domains to identify selection pressures
Functional Domain Evolution:
Comparative analysis of WD40 repeat structures between species
Assessment of LisH and F-box domain conservation and specialization
Identification of species-specific insertions/deletions that might confer specialized functions
Nuclear Receptor Co-regulator Network Evolution:
Interactome Comparison:
Systematic comparison of TBL1XR1 binding partners across species
Identification of conserved versus species-specific interactions
Analysis of co-evolution between TBL1XR1 and its binding partners
Regulatory Mechanism Conservation:
Comparison of thyroid hormone response elements across species
Analysis of the evolution of SMRT/NCoR complexes and their regulation
Assessment of proteasome recruitment mechanisms across vertebrates
Developmental Context Analysis:
Metamorphosis vs. Mammalian Development:
Comparison of TBL1XR1 functions in amphibian metamorphosis versus mammalian development
Analysis of how TBL1XR1 regulation has adapted to different developmental programs
Investigation of tissue-specific roles across species
Immune System Evolution:
Comparative analysis of TBL1XR1 functions in amphibian versus mammalian immune development
Study of how TBL1XR1 mechanisms in germinal center regulation evolved
Investigation of lineage-specific adaptations in immune function
Hormone Response Evolution:
Response Element Architecture:
Analysis of the 200-bp conserved region containing the thyroid hormone response element
Comparison of response element positioning and strength across species
Investigation of the evolution of hormone responsiveness in TBL1XR1 regulation