KEGG: bta:369019
UniGene: Bt.27277
Bovine XRRA1 is a highly conserved protein belonging to the X-ray radiation resistance-associated protein family. The significance of XRRA1 in radiation biology stems from its involvement in cellular response to X-radiation (XR). Research indicates that XRRA1 plays a crucial role in DNA damage response pathways and influences cellular resistance or sensitivity to radiation . The gene was initially identified as being down-regulated in an XR-resistant cell clone (HCT116 Clone2_XRR) .
To study Bovine XRRA1 effectively, researchers should consider its sequence homology with human XRRA1 and its conserved functional domains. The bovine version likely possesses similar mechanisms of action as its human counterpart given the high degree of conservation across mammalian species . Experimental approaches to study its significance include knockdown or overexpression studies followed by radiation exposure and assessment of cell survival, DNA damage repair kinetics, and activation of DNA damage response pathways.
Comparative structural analysis reveals significant homology between Bovine XRRA1 and human XRRA1, reflecting their evolutionary conservation. The Bovine Xrra1 gene likely contains 11 exons similar to the human version, with noted truncations in exons 4 and 8 . This conservation extends to other mammals including mouse, rat, and pig, with all exon-intron borders being well preserved .
For methodical structure comparison, researchers should employ sequence alignment tools and structural prediction software. The Bovine Xrra1 (BtXrra1) was assembled from 5 ESTs and maintains high similarity to the human gene structure . When designing expression constructs for recombinant protein production, researchers must account for these structural characteristics. For structural validation, techniques such as circular dichroism spectroscopy, X-ray crystallography, or NMR spectroscopy are recommended after producing the protein in sufficient quantities and purity.
Expression and purification of recombinant Bovine XRRA1 presents several technical challenges due to its structural properties. For bacterial expression systems using E. coli, optimization of induction conditions is crucial for improving protein solubility. Since the native protein localizes to both nucleus and cytoplasm in mammalian cells , expression systems should be selected based on the experimental requirements.
The purification strategy typically involves multi-step chromatography:
Initial capture via affinity chromatography (His-tag or GST-tag)
Secondary purification using size exclusion or ion exchange chromatography
Quality assessment via SDS-PAGE, Western blot, and functional assays
For structural studies, researchers should consider adding stabilizing agents to prevent aggregation during concentration and storage at -80°C in small aliquots to avoid freeze-thaw cycles. When expressing partial XRRA1 fragments, domain boundaries should be carefully selected based on the known functional regions to ensure proper folding and activity.
XRRA1 plays a critical role in regulating the ATM/CHK1/2 DNA repair pathway in response to radiation damage. Experimental evidence indicates that low expression of XRRA1 increases the phosphorylation of key DNA repair pathway factors including CHK1, CHK2, and ATM . This increased phosphorylation correlates with reduced expression of γ-H2AX, which participates in DNA repair in the nucleus .
To study this relationship, researchers should design experiments that:
Manipulate XRRA1 expression levels through knockdown or overexpression
Expose cells to controlled radiation doses
Monitor phosphorylation status of pathway components at multiple time points
Assess DNA repair efficiency through γ-H2AX foci formation and resolution
The novel link between XRRA1 and the ATM/CHK1/2 pathway suggests that XRRA1 is involved in DNA damage response mechanisms that drive radio- and chemoresistance . This relationship provides a promising target for overcoming therapy resistance in cancer treatment.
XRRA1 significantly influences cell cycle progression, which directly impacts cellular radiation sensitivity. Experiments with CRC cell lines demonstrate that XRRA1 expression levels affect cell cycle distribution in a specific manner. Blocked XRRA1 expression leads to cell cycle G2/M arrest through the regulation of cyclin A, cyclin E, and p21 proteins . Conversely, overexpression of XRRA1 reduces cell cycle arrest and increases cell proliferation .
The molecular mechanism underlying this effect involves several key cell cycle regulators:
Overexpression of XRRA1 activates cyclin A and cyclin E but decreases p21 expression
Downregulation of XRRA1 decreases expression of cyclin A, cyclin E, and p21 proteins
These findings suggest that XRRA1 regulates the cell cycle by targeting these specific proteins. Since cell cycle arrest is known to influence radioresistance, this regulatory function of XRRA1 may be a critical factor in determining cellular response to radiation therapy. Researchers studying radiation sensitivity should therefore consider XRRA1's cell cycle effects when designing experiments and interpreting results.
Tissue-specific expression patterns of XRRA1 have important implications for radiation therapy strategies. In humans and macaques, XRRA1 is expressed predominantly in the testis, with cDNA microarray analysis showing three-fold higher expression in macaque testis relative to other tissues . This differential expression pattern suggests that radiation sensitivity may vary across tissues based on XRRA1 levels.
To leverage this knowledge for therapeutic strategies, researchers should:
Map comprehensive expression profiles across normal and cancerous tissues
Correlate expression levels with radiation sensitivity in various cell types
Investigate whether XRRA1 can serve as a biomarker for predicting treatment response
Develop tissue-specific approaches for modulating XRRA1 activity
The finding that XRRA1 is expressed in most tumor cell types, including mouse embryonic stem cells , suggests it may play a role in cancer development and early gene expression. Understanding tissue-specific expression patterns can guide targeted therapeutic interventions that exploit differences in XRRA1 levels between normal and cancerous tissues.
Designing robust experiments to study XRRA1's role in radiation response requires careful consideration of multiple parameters. Based on current research, the following experimental conditions are recommended:
Cell Models:
Radiation Parameters:
Test multiple radiation doses (2-10 Gy) to establish dose-response curves
Include both single-dose and fractionated radiation protocols
Allow sufficient time (24-72 hours) post-radiation for assessing repair capacity
Assessment Methods:
These conditions enable comprehensive evaluation of XRRA1's function in radiation response while accounting for variables that might influence experimental outcomes.
Accurate detection and quantification of Bovine XRRA1 expression requires a multi-faceted approach using complementary techniques:
Transcriptional Analysis:
RT-PCR and qRT-PCR with primers specific to conserved regions
RNA-Seq for comprehensive transcript variant identification
Northern blotting for transcript size verification
Protein Analysis:
Western blotting using antibodies against conserved epitopes
Immunofluorescence for subcellular localization studies
ELISA for quantitative measurement in tissue samples
Expression Systems:
When analyzing Bovine XRRA1 variants, researchers should be aware that the gene likely has 11 exons with truncated exons 4 and 8 . Splice variants may exist, similar to the human XRRA1 splice variant identified in HCT116 Clone2_XRR that lacked exon three and ended on exon four .
Effective XRRA1 knockout or knockdown studies require strategic planning and rigorous validation:
Knockdown Approach:
Knockout Strategy:
Target conserved functional domains using CRISPR-Cas9
Design sgRNAs with minimal off-target effects
Screen clones using genomic PCR, sequencing, and protein expression analysis
Control Considerations:
Include appropriate empty vector controls
Generate rescue cell lines expressing shRNA-resistant XRRA1 variants
Use domain-specific knockouts to dissect protein function
Functional Validation:
These approaches enable comprehensive analysis of XRRA1 function while minimizing experimental artifacts and misinterpretation of results.
Contradictory findings regarding XRRA1's role in radiation resistance require careful interpretation within the context of experimental conditions. Several factors may contribute to these apparent contradictions:
Cell Type Specificity:
Expression Level Dependencies:
Methodological Differences:
Radiation protocols (dose, fractionation, radiation quality)
Timing of assessments post-radiation
Endpoint measurements (proliferation versus survival)
Pathway Context:
To resolve contradictions, researchers should standardize protocols, use multiple cell models, examine dose-response relationships, and consider temporal dynamics of XRRA1's effects on radiation response.
Data Normalization:
Use multiple reference genes for qRT-PCR normalization
Apply appropriate normalization for Western blot densitometry
Consider batch effects in multi-experiment comparisons
Statistical Tests:
For comparing two groups: Student's t-test with appropriate corrections
For multiple groups: ANOVA followed by post-hoc tests (Tukey, Bonferroni)
For non-normally distributed data: Non-parametric alternatives (Mann-Whitney, Kruskal-Wallis)
Experimental Design Considerations:
Determine sample size through power analysis
Include biological replicates (minimum n=3) and technical replicates
Control for confounding variables (passage number, cell density)
Advanced Analytical Approaches:
For time-course experiments: repeated measures ANOVA or mixed models
For dose-response relationships: regression analysis or EC50 determination
For complex datasets: multivariate analysis or principal component analysis
These statistical approaches enhance data reliability and facilitate meaningful interpretation of XRRA1 expression patterns across experimental conditions.
Translating in vitro findings about XRRA1 to clinical applications requires methodical approaches to establish correlations with patient outcomes:
This systematic approach enables researchers to establish clinically relevant correlations that may ultimately inform personalized radiation therapy strategies.
Several promising approaches for targeting XRRA1 to enhance radiation sensitivity warrant further investigation:
Direct XRRA1 Modulation:
siRNA or antisense oligonucleotides for transient knockdown
PROTAC-based approaches for protein degradation
Small molecule inhibitors targeting functional domains
Pathway-Based Approaches:
Radiation Protocol Optimization:
Fractionation schedules based on XRRA1 expression kinetics
Combination with radiosensitizing agents
Temporal targeting of XRRA1 at critical phases of radiation response
Advanced Delivery Systems:
Nanoparticle-based delivery of XRRA1 inhibitors
Tumor-specific targeting strategies
Radiation-activated drug release systems
These approaches offer diverse strategies for leveraging XRRA1 biology to improve radiation therapy outcomes and overcome resistance mechanisms.
Despite progress in understanding XRRA1, several critical aspects remain unexplored:
Structural Biology:
High-resolution crystal structure of full-length and partial Bovine XRRA1
Structural basis of interactions with ATM/CHK1/2 pathway components
Conformational changes upon phosphorylation or radiation exposure
Regulatory Mechanisms:
Transcriptional and post-translational regulation of Bovine XRRA1
MicroRNA and epigenetic control of expression
Protein stability and turnover rates after radiation
Evolutionary Aspects:
Functional differences between human and bovine XRRA1
Evolutionary pressures driving conservation across mammals
Species-specific regulatory mechanisms
Tissue-Specific Functions:
Role in normal tissue homeostasis versus cancer cells
Function in highly radiosensitive tissues
Developmental expression patterns and requirements
Investigating these unexplored aspects will provide deeper insights into XRRA1 biology and potentially reveal novel therapeutic opportunities.