ALYREF (THOC4) is a 27 kDa protein that binds to m5C-modified RNAs, regulating RNA metabolism, nuclear export, and gene expression . The ALYREF antibody targets this protein to study its localization, expression levels, and functional interactions in cellular and tumor models.
ALYREF antibodies have been instrumental in elucidating the protein’s role in oncogenesis across multiple cancers. Key findings include:
Knockdown Effects: ALYREF depletion suppresses CRC cell proliferation, migration, and tumor growth in vivo .
Mechanism: ALYREF recruits ELAVL1 to promote tumorigenesis via m5C-dependent RNA regulation .
Tumor Growth: ALYREF overexpression enhances cellular growth, colony formation, and mammosphere formation .
In Vivo Impact: Inducible knockdown reduces tumor volume and ki-67 staining in xenograft models .
Cell Cycle Arrest: ALYREF depletion induces G1-phase arrest and apoptosis in gastric cancer cells .
Prognostic Biomarker: Elevated ALYREF expression associates with poor prognosis in gastric adenocarcinoma .
ALYREF antibodies have revealed its binding preferences and interactions:
ALYREF is an export adapter protein crucial for the nuclear export of both spliced and unspliced mRNA. It binds to mRNA, facilitating its transfer to the NXF1-NXT1 heterodimer for export via the TAP/NXF1 pathway. ALYREF is a component of the TREX complex, which couples mRNA transcription, processing, and nuclear export. Notably, ALYREF preferentially associates with spliced mRNA. TREX recruitment to spliced mRNAs is independent of transcription and occurs upstream of the exon-junction complex (EJC). This recruitment, dependent on splicing and the mRNA cap, positions TREX near the 5' end of the mRNA for its export function. This recruitment involves an interaction between ALYREF/THOC4 and the cap-binding protein NCBP1. The TREX complex is essential for the export of Kaposi's sarcoma-associated herpesvirus (KSHV) intronless mRNAs and subsequent infectious virus production; ALYREF/THOC4 mediates TREX complex recruitment to these viral mRNAs. ALYREF is vital for TREX complex assembly and links DDX39B to the cap-binding complex (CBC). In collaboration with THOC5, it participates in NXF1-NXT1 mediated nuclear export of HSP70 mRNA, enhancing NXF1's RNA binding and nuclear rim localization. ALYREF is involved in the nuclear export of intronless mRNA, potentially recruited by ATP-bound DDX39B. Its roles extend to transcription elongation, genome stability, and the export of mRNAs containing 5-methylcytosine (m5C), where it recognizes, binds, and facilitates nucleo-cytoplasmic shuttling of these mRNAs. Furthermore, ALYREF acts as a chaperone, promoting dimerization of basic leucine zipper (bZIP) domain-containing transcription factors, thereby activating transcription.
ALYREF is an RNA-binding protein involved in transcriptional regulation and nuclear mRNA export. It has emerged as a significant factor in cancer biology, particularly in breast carcinogenesis. Research has identified ALYREF gene amplification in human cancers, with high expression levels correlating with poor clinical outcomes in human breast cancer patients . ALYREF has been demonstrated to significantly influence cellular growth in triple-negative breast cancer (TNBC) cells and tumor formation in vivo, suggesting its potential as both a prognostic biomarker and therapeutic target . Additionally, ALYREF has been shown to drive cancer cell proliferation in glioblastoma through an ALYREF-MYC positive feedback loop mechanism .
ALYREF antibodies are versatile tools suitable for multiple experimental applications:
Application | Recommended Dilution | Verified Cell/Tissue Samples |
---|---|---|
Western Blotting (WB) | 1:500-1:2000 | HeLa, A549, B-cells, SKOV3, HL-60, Mouse spleen, Rat brain |
Immunohistochemistry (IHC) | 1:50-1:200 | Rat ovary, Human vermiform appendix, Mouse testis |
Immunofluorescence (IF) | 1:50-1:200 | C6, NIH/3T3, U-2OS, U2OS |
These applications allow researchers to detect ALYREF protein expression, localization, and interactions in various experimental contexts .
For optimal Western blotting results with ALYREF antibodies:
Be aware that while the calculated molecular weight of ALYREF is 26 kDa, the observed band typically appears at approximately 30 kDa . This discrepancy is normal and likely due to post-translational modifications.
Use appropriate blocking buffers (typically 5% non-fat milk or BSA in TBST) to minimize background signal.
Include positive control samples such as HeLa or A549 cell lysates, which have been verified to express detectable levels of ALYREF .
For challenging samples, consider longer exposure times or signal enhancement systems, as ALYREF expression levels may vary across tissue types.
When interpreting results, remember that ALYREF has multiple modified forms which may result in additional bands on the membrane .
ALYREF exhibits a complex subcellular distribution pattern that should be considered when designing immunofluorescence experiments. It is primarily localized in the nucleus and nuclear speckles, but can also be found in the cytoplasm . ALYREF colocalizes with the exon junction complex (EJC), THOC4, NXF1, and DDX39B in the nucleus and nuclear speckles, and travels to the cytoplasm as part of the EJC bound to mRNA .
For successful immunofluorescence experiments:
Use appropriate nuclear counterstains (such as DAPI) to verify nuclear localization.
Consider co-staining experiments with markers of nuclear speckles or EJC components to confirm functional localization.
Optimize fixation and permeabilization methods to preserve both nuclear and potential cytoplasmic signals.
Use confocal microscopy when possible to better resolve the nuclear speckle pattern characteristic of ALYREF.
ALYREF plays a crucial role in both polyadenylated and non-polyadenylated mRNA metabolism pathways, coordinating processing and nuclear export . To investigate these functions:
RNA Immunoprecipitation (RIP) assay: Use ALYREF antibodies to immunoprecipitate ALYREF-bound RNAs followed by RT-qPCR analysis. This approach has successfully demonstrated ALYREF binding to histone mRNAs . Protocol considerations include:
RNase inhibitor addition is critical throughout the procedure
Crosslinking optimization (UV crosslinking at 254 nm is often used)
Stringent washing conditions to reduce background
iCLIP-seq (individual-nucleotide-resolution UV crosslinking and immunoprecipitation and sequencing): This technique has revealed that ALYREF universally binds to regions next to the stem-loop on replication-dependent histone mRNAs . Implementation requires:
Specialized crosslinking equipment
Library preparation optimization
Bioinformatic pipeline for data analysis
Co-immunoprecipitation: To study protein-protein interactions, as demonstrated in investigating ALYREF association with SLBP (stem-loop binding protein). Research has shown that ALYREF associates with SLBP through protein-protein interaction .
To investigate ALYREF's functional role in cancer:
Knockdown/knockout experiments: siRNA-mediated knockdown or CRISPR-based knockout of ALYREF has been shown to significantly reduce cellular growth, colony formation, mammosphere formation, and anchorage-independent growth in triple-negative breast cancer cell lines . Similar approaches in glioblastoma cells demonstrated that ALYREF inhibition dramatically downregulated cell proliferation and abolished tumorigenicity in vivo .
Overexpression studies: Stable overexpression of ALYREF in SUM159 cells led to enhanced cellular growth, increased colony formation, more colonies in soft agar assays, and increased mammosphere formation . This complementary approach confirms the oncogenic potential of ALYREF.
In vivo tumor formation assays: Inducible knockdown of ALYREF expression in orthotopic breast tumors significantly decreased tumor formation, indicating therapeutic potential . For glioblastoma, xenograft experiments using CRISPR to target ALYREF showed dramatically abolished tumorigenicity .
Molecular mechanism studies: In glioblastoma, CLIP-qPCR analysis demonstrated that ALYREF binds to MYC mRNA, specifically in the 3′-UTR region, controlling its stability . 3′-UTR reporter assays and mRNA stability assays confirmed this mechanism.
Researchers frequently encounter challenges when working with ALYREF antibodies in tissue samples:
Variable expression levels: ALYREF expression varies significantly between cancer subtypes and even within the same cancer type. For example, triple-negative breast cancers typically show higher expression than other subtypes . Consider:
Including multiple positive and negative control tissues
Using amplification steps for low-expressing samples
Optimizing antigen retrieval methods for specific tissue types
Background signal: Nuclear proteins can exhibit high background staining. To minimize this:
Test several blocking buffers (BSA, normal serum, commercial blockers)
Optimize antibody concentration through titration experiments
Increase washing steps or duration
Consider using monoclonal antibodies if polyclonal antibodies show high background
Specificity verification: Always validate antibody specificity through:
Peptide competition assays
Comparison with RNA expression data
Use of ALYREF-knockout tissues or cells as negative controls
Western blot verification of a single band at approximately 30 kDa
When investigating ALYREF across cancer types, consider:
Cancer-specific expression patterns: Analysis of large patient cohorts (METABRIC, TCGA-PanCancer Atlas, INSERM, MBC project) has demonstrated variable ALYREF amplification and expression patterns across cancer types . Design experiments to account for:
Tissue-specific expression levels
Cancer subtype variations
Correlation with other biomarkers
Context-dependent molecular partners: ALYREF interacts with different molecular partners depending on cancer type. In breast cancer, its interactions with transcriptional machinery are critical , while in glioblastoma, its binding to MYC mRNA is crucial for oncogenesis . Consider:
Co-immunoprecipitation experiments targeting tissue-specific interacting partners
Proximity ligation assays to verify protein-protein interactions in situ
RNA-IP followed by sequencing to identify cancer-specific RNA targets
Functional readouts: Different cancer types may require different functional assays:
Discrepancies between protein and mRNA expression levels are common and may reflect important biological mechanisms:
Post-transcriptional regulation: ALYREF itself regulates mRNA processing and export, creating potential feedback loops. Research has shown that ALYREF can bind to MYC mRNA and enhance its stability in GBM cells , suggesting that similar mechanisms may apply to other mRNAs.
Post-translational modifications: ALYREF undergoes modifications that affect its stability and function. The observed molecular weight (30 kDa) differs from the calculated weight (26 kDa) , indicating modifications that may vary between tissues or disease states.
Localization changes: As ALYREF shuttles between the nucleus and cytoplasm , subcellular fractionation experiments may be necessary to determine whether apparent expression changes reflect redistribution rather than absolute quantity changes.
Technical considerations: When encountering discrepancies:
Verify antibody specificity using knockdown/knockout controls
Test multiple antibodies targeting different epitopes
Use absolute quantification methods like quantitative Western blotting with recombinant protein standards
When performing co-immunoprecipitation to study ALYREF interactions:
RNase treatment control: Since ALYREF is an RNA-binding protein, determine whether interactions are RNA-dependent by comparing results with and without RNase A treatment, as demonstrated in studies of ALYREF-SLBP interaction .
Antibody specificity controls:
IgG control immunoprecipitation must be performed in parallel
Input samples (pre-IP lysate) should be analyzed to confirm target protein expression
Reverse co-IP (immunoprecipitating the suspected interacting partner and blotting for ALYREF) should confirm results
Competition controls: For validation, consider peptide competition or expression of truncated protein domains to map interaction interfaces.
Crosslinking considerations: For transient or weak interactions, chemical crosslinking may be necessary but requires careful optimization to avoid artifacts.
Research has revealed that ALYREF may have context-dependent functions:
Cancer type specificity: While ALYREF is upregulated and promotes proliferation in breast cancer and glioblastoma , it has been reported as downregulated in skin and testicular cancers . When analyzing contradictory results:
Consider tissue-specific transcriptional programs
Evaluate experimental models (cell lines vs. primary cells vs. tissues)
Examine genetic background differences between experimental systems
Functional redundancy: ALYREF belongs to the TREX complex, and other components may compensate for its loss in certain contexts. Studies show that UAP56 and THO components can also be detected on histone mRNAs , suggesting potential functional overlap.
Direct vs. indirect effects: Distinguish between direct ALYREF functions and secondary effects:
Use acute vs. chronic depletion systems
Employ rescue experiments with wild-type and mutant ALYREF
Conduct time-course experiments to establish causality
ALYREF's role in promoting cancer cell proliferation makes it an intriguing therapeutic target:
Target validation approaches:
Inducible knockdown systems in established tumors have demonstrated that reducing ALYREF expression can decrease tumor formation in vivo
CRISPR-mediated knockout of ALYREF dramatically abolished tumorigenicity in xenograft models
These findings indicate strong potential for therapeutic interventions targeting ALYREF
Biomarker development:
Immunohistochemistry with validated ALYREF antibodies could stratify patients according to expression levels
Survival analyses from two independent breast cancer cohorts suggest ALYREF as a novel prognostic biomarker
Correlation studies between ALYREF and MYC expression could identify patients who might benefit from targeted therapies
Therapeutic resistance monitoring:
Monitor ALYREF expression changes during treatment using validated antibodies
Investigate whether ALYREF-mediated mRNA export contributes to therapeutic resistance by regulating stress response genes
To investigate ALYREF's selective regulation of mRNAs:
Integrated RNA-binding and functional analyses:
mRNA stability assays:
Reporter assays with mutated binding sites:
Integrating ALYREF studies with multi-omics approaches offers powerful insights:
Proteogenomic integration:
Single-cell analyses:
Apply ALYREF antibodies in single-cell imaging mass cytometry or multiplexed immunofluorescence
This would reveal intratumoral heterogeneity of ALYREF expression and localization
Correlation with other cancer markers at single-cell resolution could identify previously unrecognized cellular states
Spatial transcriptomics with protein validation:
Combine spatial transcriptomics with ALYREF immunohistochemistry in sequential sections
This would map relationships between ALYREF protein expression and spatially resolved transcriptomes
Such approaches could reveal tumor microenvironment influences on ALYREF function
When selecting ALYREF antibodies:
Epitope location: Consider where the antibody binds within the ALYREF protein structure:
Cross-reactivity: Verify species reactivity for your experimental system:
Application-specific validation:
Monoclonal vs. polyclonal considerations:
Polyclonal antibodies provide signal amplification but may have batch variation
Monoclonal antibodies offer consistency but may be more sensitive to epitope masking
To capture dynamic ALYREF interactions:
Live-cell imaging approaches:
Fluorescent protein tagging of ALYREF combined with potential interaction partners
FRET/BRET systems to detect proximity in living cells
Photobleaching techniques to measure binding kinetics
Sequential immunoprecipitation strategies:
Proximity labeling technologies:
ALYREF coordinates both processing and nuclear export of mRNAs . To dissect these functions:
Function-specific mutants:
Design ALYREF mutants that selectively disrupt specific protein interactions
Test their impact on discrete steps of mRNA processing and export
Use these mutants in rescue experiments following endogenous ALYREF depletion
Sequential process analysis:
Nuclear/cytoplasmic fractionation to track mRNA movement
Nascent RNA capture to study ALYREF's role during transcription
RNA processing assays to evaluate 3′-end formation efficiency
Structured experimental approaches: