RALY is a 306-amino-acid protein with a molecular mass of ~34.9 kDa . Key structural features include:
RNA-recognition motif (RRM): Located at the N-terminal region, enabling binding to uracil (U)-rich sequences in RNA .
Nuclear localization signals (NLS): Predicted bipartite NLS sequences direct nuclear localization but also permit cytoplasmic activity .
Glycine-rich region (GRR): C-terminal region involved in protein-protein interactions .
RALY interacts with diverse RNA and protein partners, influencing RNA metabolism and cellular processes:
RALY binds U-rich elements in the 3' untranslated regions (UTRs) of mRNAs, such as ANXA1 (Annexin A1) and H1FX (histone H1 variant) . This binding regulates mRNA stability and translation.
Proteomic studies identified 143 interaction partners, including:
RALY regulates:
Alternative splicing: Modulates splicing of FOS and FOSB transcripts, affecting immune response and cell proliferation .
Mitochondrial metabolism: Reprograms mitochondrial gene expression in colorectal cancer (CRC) via miRNA regulation (miR-483, miR-676, miR-877) .
Immune response: Suppresses IFIT1, IFIT2, and OASL expression, dampening antiviral pathways .
RALY overexpression correlates with aggressive CRC and poor prognosis. In CRC, RALY promotes Drosha-mediated miRNA processing, downregulating mitochondrial ATP synthase subunits (ATP5I, ATP5G1, ATP5G3) and CYC1, shifting metabolism toward glycolysis .
In HeLa cells, RALY overexpression:
RALY regulates 645 alternative splicing events (ASEs) in HeLa cells, including:
| Gene | ASE Type | Biological Process | Source |
|---|---|---|---|
| FOS | Exon skipping | Transcription regulation | |
| CCDC17 | Intron retention | Cell proliferation |
RALY’s role in chemotherapy resistance (e.g., oxaliplatin in CRC) and metastasis suggests it as a potential therapeutic target .
High RALY expression in CRC correlates with poor survival, as shown in The Cancer Genome Atlas (TCGA) and local cohorts .
RALY (RALY Heterogeneous Nuclear Ribonucleoprotein) functions as an RNA-binding protein belonging to the Heterogeneous Nuclear Ribonucleoprotein (hnRNP) family. Its primary functions include regulating gene expression through alternative splicing mechanisms and modulating immune/inflammatory response pathways. Research indicates that RALY can affect cellular proliferation in specific contexts, as demonstrated in studies with HeLa cells where RALY overexpression inhibited proliferation . The protein also appears to play regulatory roles in transcription factor expression, with notable effects on FOS and FOSB regulation. To study RALY's fundamental functions, researchers typically employ RNA immunoprecipitation assays, transcriptome analysis, and protein-protein interaction studies to identify binding partners and affected pathways.
The most reliable experimental systems for studying RALY include:
Cell culture models: HeLa cells have been extensively used for RALY overexpression studies as demonstrated in the literature . Other cancer cell lines can be employed based on the specific cancer context being investigated.
Gene manipulation techniques: CRISPR-Cas9 for knockout studies, siRNA for knockdown experiments, and plasmid-based systems for overexpression models allow researchers to manipulate RALY levels.
RNA-sequencing analysis: This provides comprehensive insights into transcriptome changes associated with RALY manipulation, as exemplified in studies showing that RALY overexpression significantly altered the gene expression profile in HeLa cells .
RT-qPCR validation: This technique confirms RNA-seq findings regarding specific gene targets, particularly for immune response genes like IFIT1, IFIT2, IFIT3, and IFI44, which have been shown to be inhibited by RALY overexpression .
When designing experiments, researchers should include appropriate controls and consider time-course analyses to capture both immediate and downstream effects of RALY modulation.
RALY significantly impacts alternative splicing processes in human cells through the following mechanisms:
Research has demonstrated that RALY overexpression regulates approximately 645 alternative splicing events in HeLa cells . These events are particularly enriched in processes related to transcription regulation, RNA splicing, and cell proliferation via metabolic pathways.
The protein appears to bind specific RNA motifs, thereby influencing splice site selection and exon inclusion/exclusion patterns.
RALY-mediated alternative splicing affects genes involved in immune function and inflammatory responses, suggesting a potential regulatory mechanism for RALY's impact on these pathways.
Methodologically, researchers investigating RALY's splicing activity should employ RNA-seq with splice junction analysis, followed by validation using exon-specific RT-PCR assays. Minigene constructs can further confirm direct RALY effects on specific splicing events of interest. Cross-linking immunoprecipitation sequencing (CLIP-seq) provides valuable insights into the direct RNA targets of RALY within the transcriptome.
To effectively investigate RALY's impact on immune and inflammatory pathways, researchers should implement a multi-faceted experimental approach:
Pathway-specific transcriptome analysis: RNA-seq followed by pathway enrichment analysis has revealed that RALY overexpression significantly upregulates genes involved in immune/inflammatory responses through the NOD-like receptor signaling pathway and cytokine-cytokine receptor interactions .
Cytokine profiling: Measuring secreted cytokines in control versus RALY-manipulated cells provides functional confirmation of transcriptional changes.
Immune cell co-culture systems: These models help assess how RALY-expressing cells influence immune cell behavior, particularly relevant for cancer immunology studies.
ChIP-seq analysis: This technique identifies how RALY impacts transcription factor binding at immune response gene promoters, particularly for FOS/FOSB which are negatively regulated by RALY .
Research has demonstrated that RALY negatively regulates the expression of several interferon-stimulated genes, including IFIT1, IFIT2, IFIT3, IFI44, HERC4, and OASL . This suggests that RALY may function as an immune response modulator, potentially explaining its role in chemotherapy resistance through immunomodulation pathways.
The apparently contradictory effects of RALY on cell proliferation across different cellular contexts require careful experimental design:
Cell type diversity: Studies should include multiple cell lines representing different tissue origins and genetic backgrounds, as RALY has shown inhibitory effects on proliferation in HeLa cells but may behave differently in other contexts.
Dose-dependent analysis: Establishing RALY expression at various levels (from knockdown to physiological to overexpression) helps identify potential threshold effects.
Temporal considerations: Short-term versus long-term RALY manipulation experiments capture both immediate signaling changes and adaptive responses.
Pathway inhibitor studies: Using inhibitors of key signaling pathways can help dissect which downstream mechanisms mediate RALY's context-specific effects.
Researchers should also examine how the cellular microenvironment influences RALY's effects, particularly in the context of immune cell interactions and inflammatory stimuli.
Investigating RALY's contribution to chemotherapy resistance requires specialized methodological approaches:
Patient-derived models: Using cell lines or organoids from therapy-resistant versus therapy-sensitive tumors provides clinically relevant contexts.
Drug response profiling: Dose-response curves for various chemotherapeutics should be established in RALY-manipulated cells.
Mechanistic pathway analysis: Since RALY regulates immune response genes and may affect cell death pathways through alternative splicing , researchers should examine how these mechanisms contribute to therapy resistance.
In vivo validation: Xenograft models with RALY manipulation allow assessment of therapy response in a more complex environment.
The research finding that RALY regulates immune/inflammatory gene expression suggests a potential mechanism for chemotherapy resistance, as inflammatory signaling often contributes to therapy resistance. Researchers should specifically investigate how RALY-mediated alternative splicing of transcription factors like FOS impacts downstream gene expression patterns that confer resistance phenotypes.
The dual role of RALY in alternative splicing and transcriptional regulation requires integrated experimental approaches:
Combined RNA-seq and splicing-sensitive analyses: RNA-seq data should be analyzed both for differential gene expression and for alternative splicing events, as demonstrated in studies showing 645 RALY-regulated splicing events .
Transcription factor splicing analysis: Special attention should be paid to alternative splicing of transcription factors like FOS, which RALY has been shown to regulate .
Motif analysis: Identifying common RNA motifs in RALY targets helps establish direct versus indirect effects.
Sequential chromatin and RNA immunoprecipitation: This approach can determine whether RALY functions in coordinated ribonucleoprotein complexes that link splicing to transcription.
Research has demonstrated that RALY's splicing regulation significantly impacts genes involved in transcription regulation and RNA processing pathways , creating potential feedback loops in gene expression control. This interplay likely contributes to RALY's complex effects on cellular processes like immune response and proliferation.
Current RALY research faces several methodological challenges:
Antibody specificity: Many commercial antibodies for RALY have cross-reactivity issues. Solution: Researchers should validate antibodies using RALY knockout controls and consider using epitope-tagged RALY for cleaner detection.
Identifying direct versus indirect targets: It remains challenging to distinguish primary RALY targets from secondary effects. Solution: Utilize rapid induction systems (e.g., auxin-inducible degron) combined with metabolic RNA labeling to capture immediate effects.
Context-dependent function: RALY shows varied effects across cellular contexts . Solution: Develop comprehensive cell type atlases of RALY function using single-cell RNA-seq and proteomics.
Integration of multiple functions: RALY's roles in RNA binding, splicing, and other processes are difficult to study simultaneously. Solution: Multi-omics approaches that integrate transcriptomics, proteomics, and interactome analyses.
Research indicates that RALY regulates both gene expression and alternative splicing , but the exact mechanism linking these functions remains incompletely understood. Emerging technologies like spatial transcriptomics and in situ sequencing may help resolve how RALY orchestrates these processes within specific subcellular compartments.
Investigating RALY's role in cancer progression requires specialized experimental designs:
Clinical correlation studies: Analyze RALY expression in primary versus metastatic lesions from the same patients to establish relevance.
Invasion and migration assays: Utilize both 2D and 3D models with RALY manipulation to assess motility and invasive capacity.
Metastasis models: Employ tail vein injection and orthotopic implantation models with RALY-manipulated cells to assess in vivo metastatic potential.
Epithelial-mesenchymal transition (EMT) analysis: Since RALY regulates alternative splicing and transcription factors , researchers should specifically examine EMT-related gene expression and splicing patterns.
The finding that RALY regulates immune response genes suggests it may also influence the tumor microenvironment, potentially affecting immune surveillance and metastatic niche formation. Researchers should therefore include immune component analyses in their experimental designs when studying RALY's cancer-related functions.
To expand RALY research beyond cancer models into broader physiological contexts:
Conditional knockout models: Tissue-specific and inducible RALY knockout mice can reveal developmental and homeostatic functions.
Interactome studies: AP-MS and BioID approaches can identify tissue-specific RALY binding partners across different physiological states.
RNA structure analysis: SHAPE-seq and related techniques can determine how RALY influences RNA structural landscapes.
Evolutionary analysis: Comparative studies of RALY function across species can highlight conserved core functions versus species-specific adaptations.
Given RALY's demonstrated roles in immune regulation and alternative splicing , researchers should particularly investigate its functions in immune development, inflammatory disease models, and tissue regeneration contexts. The protein's negative regulation of FOS/FOSB transcription factors suggests potential roles in cellular stress responses and adaptation that extend beyond cancer biology.
RALY, also known as RNA-binding protein Raly, is a member of the heterogeneous nuclear ribonucleoprotein (hnRNP) family. This protein plays a crucial role in pre-mRNA splicing and embryonic development . The human recombinant form of RALY is produced using Escherichia coli (E. coli) and is often used in laboratory research.
The RALY gene is located on chromosome 20q11.22 . The protein encoded by this gene consists of 329 amino acids and has a molecular mass of approximately 34.9 kDa . The recombinant form of RALY is fused to a 23 amino acid His-tag at the N-terminus, which facilitates its purification through chromatographic techniques .
RALY is involved in the processing of pre-mRNA, a critical step in the gene expression pathway. It binds to RNA and participates in the splicing process, which removes introns from pre-mRNA transcripts . This function is essential for the proper expression of genes and the production of functional proteins.
Additionally, RALY has been implicated in embryonic development. Its role in this process is not fully understood, but it is believed to be involved in the regulation of gene expression during early development .
The recombinant form of RALY is produced in E. coli as a single, non-glycosylated polypeptide chain . The protein is purified using proprietary chromatographic techniques to achieve a purity greater than 90% as determined by SDS-PAGE . The recombinant RALY protein is typically supplied in a sterile, filtered, colorless solution containing Tris-HCl buffer and glycerol .
Recombinant RALY is primarily used in laboratory research to study its function and role in various biological processes. It is also used in experiments to understand its involvement in diseases and to develop potential therapeutic interventions.
The recombinant RALY protein should be stored at 4°C if it is to be used within 2-4 weeks. For longer storage periods, it should be frozen at -20°C. To ensure stability, it is recommended to add a carrier protein such as human serum albumin (HSA) or bovine serum albumin (BSA) and to avoid multiple freeze-thaw cycles .