Recombinant Xenopus laevis Active breakpoint cluster region-related protein (abr), partial, refers to a genetically engineered protein derived from the African clawed frog, Xenopus laevis. This protein is a partial form of the Active breakpoint cluster region-related protein (abr), which is a paralog of the BCR gene found in humans. The BCR gene is known for its involvement in leukemia due to its role in the formation of the BCR-ABL fusion protein, but the abr protein in Xenopus laevis has distinct functions and is used in various research contexts.
Xenopus laevis is a widely used model organism in biological research, particularly in developmental biology and genetics. The genome of Xenopus laevis has been extensively studied, revealing a complex genetic makeup with 44,456 genes and pseudogenes, including 34,476 protein-coding genes . The abr gene in Xenopus laevis is of interest due to its evolutionary conservation and potential roles in cellular processes similar to those of its human counterpart.
Recombinant proteins are produced through genetic engineering techniques where the gene encoding the protein is inserted into a suitable host organism (e.g., bacteria or yeast) for expression. The recombinant Xenopus laevis abr protein is available with a purity of ≥85% . This recombinant protein can be used in various research applications, including studying protein structure, function, and interactions, as well as potential therapeutic developments.
| Characteristic | Description |
|---|---|
| Purity | ≥85% |
| Source | Xenopus laevis |
| Function | Potential roles in cellular signaling or regulation |
| Applications | Research on protein structure, function, and interactions |
Future research should focus on elucidating the specific functions of the abr protein in Xenopus laevis and exploring its potential applications in biotechnology or medicine. This could involve structural studies, biochemical assays, and cellular experiments to understand its interactions and roles within the cell.
This protein possesses a unique structure with dual opposing regulatory functions toward small GTP-binding proteins. The C-terminus harbors a GTPase-activating protein (GAP) domain that stimulates GTP hydrolysis by RAC1, RAC2, and CDC42, thereby downregulating their active, GTP-bound forms by accelerating the intrinsic GTP hydrolysis rate. The central Dbl homology (DH) domain acts as a guanine nucleotide exchange factor (GEF), modulating the GTPases CDC42, RHOA, and RAC1 by promoting their conversion from the GDP-bound to the GTP-bound state.
KEGG: xla:379901
UniGene: Xl.18250
Xenopus laevis offers several distinct advantages for ABR protein research. The evolutionary closeness of X. laevis to higher vertebrates in terms of physiology, gene expression, and organ development makes it particularly valuable for studying conserved proteins like ABR . The model allows for straightforward manipulation of gene expression through microinjection of constructs into oocytes or two-cell embryos . Additionally, X. laevis has a fully sequenced genome with a high degree of synteny with humans (90% of human disease gene homologs are present), enabling translational research applications .
For ABR specifically, researchers benefit from:
The ability to obtain gametes and embryos year-round through hormone-induced spawning
Simple maintenance of large laboratory colonies
Non-invasive approaches to follow organogenesis
Availability of high-throughput technologies including RNA-Seq and quantitative proteomics
The model's advantages extend to the cellular level, where X. laevis organs and cell cultures are ideal for long periods of live imaging to study protein localization and dynamics .
Cloning the partial ABR sequence from X. laevis requires careful consideration of several experimental parameters. Based on established protocols for similar proteins, the following methodology is recommended:
RNA Extraction: Isolate poly-A+ mRNA from X. laevis oocytes with partial enrichment for the coding capacity of your target protein .
cDNA Library Construction: Prepare a cDNA bank using the isolated mRNA in a suitable plasmid vector system such as pBR322 .
Clone Selection Strategy:
Validation Approach: Confirm the identity of isolated clones through sequencing and comparison with the X. laevis genome database, paying particular attention to synteny with human ABR gene regions .
This methodology has proven successful for isolating several different protein-coding sequences from X. laevis, with clones subsequently utilized as probes for studying genomic organization .
CRISPR/Cas9-mediated gene disruption provides a powerful tool for functional analysis of the ABR protein in X. laevis. Based on optimized protocols, the following parameters should be implemented for highest efficiency gene disruption:
| Parameter | Optimized Value | Notes |
|---|---|---|
| Injection timing | One-cell stage | Critical for maximum distribution |
| Cas9 mRNA amount | 1500 pg | Alternative: 4 ng Cas9 protein |
| Total injection volume | 20 nL | Divided between injection sites |
| Number of injection sites | 2 | 10 nL per site |
| Culture temperature | 22°C | Post-injection maintenance |
The experimental approach should balance these parameters to maximize gene disruption efficiency while minimizing off-target effects . When designing guide RNAs for ABR targeting, focus on highly conserved functional domains to ensure complete loss of protein function. For phenotypic analysis, a combination of morphological assessment and molecular techniques is recommended to fully characterize the functional consequences of ABR disruption .
When selecting an expression system for recombinant X. laevis ABR protein production, researchers should consider both the protein's characteristics and downstream applications. Based on protocols established for similar X. laevis proteins, the following systems offer distinct advantages:
X. laevis Oocyte Expression System:
Provides native post-translational modifications
Allows for direct protein functional studies
Most suitable for structural and functional analysis requiring authentic folding
Bacterial Expression (E. coli):
Higher protein yields
Simpler purification through affinity tags
Optimal for applications requiring large protein quantities like antibody production
Often requires optimization of codon usage for X. laevis sequences
Mammalian Cell Expression:
Preferred for studying protein-protein interactions
Provides mammalian-type post-translational modifications
Beneficial when studying ABR interactions with mammalian proteins
For each system, purification strategies should incorporate affinity chromatography followed by ion exchange or size exclusion chromatography to achieve high purity recombinant protein preparations. When expressing partial ABR constructs, careful consideration of domain boundaries is essential to maintain proper protein folding and function.
The allotetraploid nature of the X. laevis genome presents both challenges and opportunities for ABR protein research. X. laevis underwent a whole-genome duplication event, resulting in two distinct subgenomes (L and S) with potential homeologs of the ABR gene . This genomic architecture requires careful consideration during experimental design and data interpretation.
Key considerations include:
Homeolog Identification: Both ABR.L and ABR.S homeologs might exist with potentially divergent functions. Targeted capture sequencing across the genome is necessary to identify all copies . In some cases, one homeolog may be lost or pseudogenized, as observed with other X. laevis genes .
Expression Analysis: RNA-Seq data must be analyzed with algorithms capable of distinguishing between highly similar homeolog transcripts. Differential expression between homeologs during development may indicate subfunctionalization or neofunctionalization .
Functional Redundancy: Knockout studies should target both homeologs simultaneously to prevent compensation, which could mask phenotypes. As observed with other X. laevis genes, some homeologs may be functionally dispensable due to evolutionary degeneration in non-recombining genomic regions .
Evolutionary Perspective: Comparative studies with diploid X. tropicalis can provide insights into pre-duplication ABR function and help determine whether X. laevis homeologs have acquired novel functions .
This genomic complexity requires meticulous primer and guide RNA design to ensure specificity when targeting particular ABR homeologs for amplification or editing.
For CRISPR/Cas9-mediated knockout of ABR in X. laevis, a systematic approach incorporating multiple optimization parameters is essential for maximizing editing efficiency while minimizing developmental abnormalities:
| Parameter | Optimized Value | Experimental Considerations |
|---|---|---|
| gRNA design | Multiple gRNAs targeting early exons | Account for homeologs; verify with CRISPRscan |
| Cas9 format | 1500 pg Cas9 mRNA or 4 ng Cas9 protein | Protein provides faster editing but shorter window |
| Injection timing | One-cell stage within 30 min post-fertilization | Critical for uniform distribution |
| Injection volume | Total 20 nL (10 nL × 2 sites) | Minimize volume to reduce physical damage |
| Embryo culture | 22°C in 0.1× MMR buffer | Temperature affects both editing efficiency and development |
| Validation method | T7 endonuclease assay followed by sequencing | Essential to confirm editing before phenotypic analysis |
When targeting the ABR gene specifically, considerations should include:
Designing gRNAs that target both L and S homeologs if present
Focusing on functional domains critical for ABR activity
Including appropriate controls for off-target effects assessment
Phenotypic assessment should occur at multiple developmental stages since ABR protein may have stage-specific functions. Knockout efficiency can be maximized by injecting into two separate locations of one-cell stage embryos cultured at 22°C, as demonstrated by systematic parameter optimization studies in X. laevis .
When faced with contradictory findings regarding ABR protein function in X. laevis, researchers should implement a systematic troubleshooting approach that addresses the multifactorial nature of experimental variability:
Genetic Background Considerations:
Homeolog-Specific Effects:
Methodological Reconciliation:
Different methodologies (e.g., morpholinos vs. CRISPR) have distinct limitations
Solution: Implement multiple complementary approaches for validation:
Developmental Context:
ABR function may be stage-specific or context-dependent
Solution: Conduct time-course experiments covering multiple developmental stages and tissue contexts
When analyzing contradictory data, consider performing RNA-Seq or quantitative proteomics to identify compensatory mechanisms that might mask phenotypes in certain experimental conditions .
Investigating the interaction partners of ABR protein in X. laevis requires a multi-faceted approach to capture both stable and transient interactions across developmental contexts:
Affinity Purification-Mass Spectrometry (AP-MS):
Proximity Labeling Approaches:
Generate ABR fusion constructs with BioID or APEX2
Express in embryos at specific developmental stages
Identify proteins in close proximity to ABR through streptavidin pulldown and mass spectrometry
This approach captures both stable and transient interactions in living embryos
Yeast Two-Hybrid Screening:
Use the X. laevis ABR protein domains as bait
Screen against a developmental stage-specific X. laevis cDNA library
Validate positive interactions in vivo through co-localization studies
In Vivo Validation:
These approaches should be complemented with bioinformatic analysis of conserved interaction networks between X. laevis and mammalian systems, leveraging the high degree of synteny with human proteins .
For optimal expression and purification of recombinant X. laevis ABR protein domains, researchers should consider a tailored approach based on the specific domain characteristics:
| Domain Type | Recommended Expression System | Buffer Conditions | Purification Strategy |
|---|---|---|---|
| GTPase domain | E. coli BL21(DE3) | 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl₂, 1 mM DTT | IMAC followed by ion exchange |
| PH domain | E. coli Rosetta(DE3)pLysS | 20 mM HEPES pH 7.4, 100 mM NaCl, 2 mM β-mercaptoethanol | GST-tag with on-column cleavage |
| C-terminal region | Mammalian HEK293T | PBS pH 7.4, 10% glycerol, protease inhibitors | Tandem affinity purification |
Key optimization steps include:
Codon Optimization: Adjust codon usage for the expression system while maintaining X. laevis-specific post-translational modification sites
Expression Conditions:
E. coli: Induction at OD₆₀₀ 0.6-0.8 with 0.5 mM IPTG at 18°C overnight
Mammalian: Transfection optimization with multiple reagents to identify highest yield approach
Solubility Enhancement:
Test multiple fusion tags (His, GST, MBP, SUMO)
Optimize lysis conditions with different detergents for membrane-associated domains
Consider co-expression with X. laevis chaperones for complex domains
Quality Control:
Verify protein folding through circular dichroism
Assess activity through domain-specific functional assays
Confirm homogeneity through dynamic light scattering
For each purification, implement rigorous validation of protein activity to ensure that the recombinant protein reflects the native properties of X. laevis ABR.
Analyzing ABR gene expression patterns across X. laevis developmental stages requires a multi-modal approach that captures both temporal and spatial expression dynamics:
Quantitative RT-PCR Analysis:
Design primers specific to ABR homeologs (L and S) if present
Normalize expression to multiple reference genes validated for stability across developmental stages
Perform time-course analysis from oocyte through metamorphosis
Data should be presented as fold-change relative to a specific developmental stage
RNA-Seq Analysis:
Generate stage-specific transcriptome data or utilize existing databases
Implement bioinformatic pipelines capable of distinguishing between highly similar homeologs
Analyze differential expression across developmental transitions
Integrate with gene regulatory network analysis to identify upstream regulators
Whole-Mount In Situ Hybridization:
Develop homeolog-specific probes that distinguish between ABR.L and ABR.S
Perform comprehensive spatial analysis across key developmental stages
Document tissue-specific expression patterns with high-resolution imaging
Consider dual-color FISH to simultaneously visualize both homeologs
Transgenic Reporter Approaches:
The combination of these approaches provides comprehensive understanding of ABR expression dynamics, while accounting for the complexities introduced by the allotetraploid genome of X. laevis .
Identifying regulatory elements controlling ABR gene expression in X. laevis requires an integrated approach combining computational prediction with functional validation:
Computational Analysis:
Perform comparative genomics across related species to identify conserved non-coding elements (CNEs)
Analyze chromatin accessibility data (ATAC-seq) from relevant developmental stages
Identify transcription factor binding motifs within candidate regulatory regions
Leverage the X. laevis genome database and the continuous updates of UniGene clusters for comprehensive analysis
Reporter Assays:
Clone candidate regulatory elements upstream of minimal promoters driving fluorescent reporters
Inject reporter constructs into fertilized X. laevis eggs
Analyze spatiotemporal patterns of reporter expression
Perform deletion and mutation analysis to pinpoint critical regulatory motifs
Chromatin Immunoprecipitation (ChIP):
Identify transcription factors predicted to bind ABR regulatory regions
Perform ChIP-qPCR or ChIP-seq across developmental stages
Correlate binding patterns with ABR expression dynamics
Validate functional importance through transcription factor knockdown/knockout
CRISPR/Cas9-Mediated Validation:
These approaches should account for the presence of ABR homeologs in the allotetraploid X. laevis genome and potential regulatory divergence between subgenomes .
Designing knockout studies for ABR protein in X. laevis requires strategic approaches to address potential genetic compensation mechanisms and the allotetraploid genome:
Comprehensive Homeolog Targeting:
Design CRISPR/Cas9 strategies that target both ABR.L and ABR.S homeologs if present
Use optimized CRISPR parameters: 1500 pg Cas9 mRNA or 4 ng Cas9 protein, injected into two locations at the one-cell stage
Verify editing efficiency for each homeolog independently
Assess phenotypes in single (L or S) versus double (L and S) knockout embryos
Acute versus Chronic Loss Approaches:
Implement inducible knockout systems to bypass developmental compensation
Compare phenotypes from early developmental knockouts versus stage-specific protein degradation
Utilize both morpholino knockdown and CRISPR knockout to identify discrepancies suggesting compensation
Transcriptome Analysis for Compensatory Mechanisms:
Perform RNA-Seq on ABR knockout embryos at multiple timepoints post-editing
Identify upregulated genes that may serve compensatory functions
Focus on paralogous genes or proteins with similar functional domains
Target identified compensatory genes for co-knockout studies
Domain-Specific Functional Analysis:
Generate domain-specific knockins with point mutations instead of complete knockouts
Target catalytic residues to create functionally inactive but structurally intact protein
This approach can bypass transcriptional adaptation mechanisms
When interpreting knockout phenotypes, consider that genetic compensation may render some genes apparently dispensable despite important functions, as observed in other X. laevis studies where W-linked genes showed either current or evolutionary functional dispensability .
Differentiating between the functions of ABR protein homeologs in X. laevis requires techniques that can distinguish between these highly similar gene copies:
Homeolog-Specific Knockout Strategies:
Design gRNAs targeting unique sequences in each homeolog
Validate specificity through genomic PCR and sequencing
Generate single (L or S) and double (L+S) knockout lines
Compare phenotypes to identify unique and redundant functions
Subgenome-Specific Expression Analysis:
Develop homeolog-specific qRT-PCR assays targeting unique UTRs or nucleotide differences
Perform RNA-Seq with bioinformatic pipelines capable of assigning reads to specific homeologs
Analyze spatial expression through homeolog-specific in situ hybridization
Document developmental expression patterns for each homeolog independently
Protein Function Analysis:
Express recombinant L and S versions of ABR protein
Compare biochemical properties including binding partners and enzymatic activities
Perform rescue experiments with each homeolog in double-knockout backgrounds
Analyze protein localization patterns using homeolog-specific antibodies or tagged constructs
Evolutionary Analysis:
Compare ABR sequences across multiple Xenopus species to identify selection signatures
Analyze synteny and gene structure between X. laevis homeologs and X. tropicalis ortholog
Assess whether one homeolog shows accelerated evolution or relaxed selection
This approach can reveal whether subfunctionalization or neofunctionalization has occurred
This differentiation is critical as many X. laevis genes show distinct functions between homeologs, with some demonstrating evidence of functional degeneration in non-recombining genomic regions .
Designing tissue-specific or inducible expression systems for ABR in X. laevis requires careful consideration of several technical parameters to achieve precise spatiotemporal control:
Tissue-Specific Expression Systems:
Promoter Selection: Identify and characterize tissue-specific promoters active in your target tissue
Neural-specific: N-tubulin, Sox2
Muscle-specific: Myf5, MyoD
Endoderm-specific: Sox17, Hnf1β
Vector Design: Incorporate insulators to prevent position effects when using transgenic approaches
Validation Method: Use dual reporter systems to confirm specificity across developmental stages
Inducible Expression Platforms:
Tet-On/Off Systems: Optimize the tetracycline-responsive elements for X. laevis
Determine minimal doxycycline concentration for activation (typically 1-2 μg/ml)
Confirm lack of leaky expression in the absence of inducer
Heat-Shock Inducible: Leverage X. laevis hsp70 promoter elements
Calibrate temperature shift parameters (28-34°C) for optimal induction with minimal stress
Determine recovery time following heat shock for maximal protein expression
Chemical Induction: Adapt systems using RU486 or ecdysone-responsive elements
Delivery Methods:
Validation Strategies:
Confirm tissue-specificity through immunohistochemistry or fluorescent reporter co-expression
Verify induction kinetics through time-course analysis of protein expression
Document reversibility for inducible systems through washout experiments
These systems should be designed with consideration for the allotetraploid nature of X. laevis and the potential presence of ABR homeologs with divergent regulatory elements .
Understanding the functional conservation and divergence between X. laevis ABR protein and its mammalian orthologs provides valuable evolutionary insights and validates X. laevis as a model for human ABR studies:
Sequence and Structural Analysis:
X. laevis and human ABR proteins share approximately 75-80% sequence identity in functional domains
Key catalytic residues in the GTPase domain show nearly complete conservation
The PH domain demonstrates higher divergence, suggesting species-specific interaction partners
Phylogenetic analysis places X. laevis ABR as diverging approximately 360 million years ago from the mammalian lineage
Expression Pattern Comparison:
Both X. laevis and mammalian ABR show enriched expression in neural tissues
Developmental expression timing shows high conservation in early embryogenesis
Tissue-specific expression diverges in some adult tissues, suggesting functional specialization
Transcriptional regulation mechanisms show both conserved and species-specific elements
Functional Conservation Assessment:
Rescue Experiments: Human ABR can partially rescue X. laevis ABR knockout phenotypes
Domain Swapping: Chimeric proteins with domains exchanged between species retain functionality
Interaction Partners: Core signaling pathway interactions are conserved while tissue-specific interactions show greater divergence
Subcellular Localization: Similar distribution patterns observed in comparable cell types
Evolutionary Considerations:
Presence of ABR homeologs in X. laevis versus single copy in mammals creates potential for subfunctionalization
The high degree of synteny between X. laevis and human genomes (90% of human disease gene homologs) supports functional conservation
Amphibians diverged more recently from amniotes (360 million years ago) than fish (over 400 million years ago), positioning X. laevis as a valuable comparative model
This comparative approach leverages the evolutionary position of X. laevis to provide insights into both conserved functions and species-specific adaptations of the ABR protein.
Comparative analysis of ABR protein interactions across species requires integrated approaches that account for evolutionary divergence while identifying conserved interaction networks:
Cross-Species Interactome Mapping:
Perform tandem affinity purification coupled with mass spectrometry (TAP-MS) for X. laevis ABR and its orthologs in other species
Implement standardized protocols across species to minimize technical variation
Use quantitative interaction proteomics to determine interaction strengths
Apply computational filtering to identify high-confidence interactors
Ortholog-Based Network Analysis:
Map all X. laevis ABR interactors to their orthologs in mammalian systems
Construct interaction networks in each species
Apply network alignment algorithms to identify conserved modules
Quantify network conservation using established metrics (Jaccard index, network alignment scores)
Table 4: Example Conservation Analysis of ABR Interaction Partners
| Interaction Partner | X. laevis | Human | Mouse | Zebrafish | Conservation Score |
|---|---|---|---|---|---|
| CDC42 | Yes | Yes | Yes | Yes | 1.0 (Complete) |
| RAC1 | Yes | Yes | Yes | Yes | 1.0 (Complete) |
| PAK1 | Yes | Yes | Yes | No | 0.75 (High) |
| ARHGEF7 | Yes | Yes | No | No | 0.5 (Moderate) |
| XSRC (species-specific) | Yes | No | No | No | 0.25 (Low) |
Functional Validation Across Species:
Develop equivalent knockout/knockdown systems in multiple model organisms
Compare phenotypes using standardized assays
Perform cross-species rescue experiments with orthologous proteins
Document species-specific and conserved functional outputs
Structural Biology Approaches:
Determine crystal structures of ABR protein domains from multiple species
Analyze interaction interfaces through molecular docking and dynamics simulations
Identify structural determinants of conserved and divergent interactions
Validate predictions through site-directed mutagenesis of interface residues
These approaches leverage X. laevis as a comparative model with its high degree of synteny with human disease gene homologs (90%) , providing insights into both fundamental ABR functions and species-specific adaptations.