RBM17 contains three conserved domains critical for its function:
Structural studies using NMR and GST pull-down assays confirm that the UHM domain binds SF3B1’s ULM5 with higher affinity () compared to SAP30BP’s ULM () . Mutations in these domains (e.g., D319K in UHM) disrupt splicing activity .
RBM17 replaces U2AF in splicing short introns (<150 nt) via a non-canonical pathway:
Cofactor Recruitment: SAP30BP binds RBM17’s UHM domain, forming an intermediate complex that shields RBM17 from inactive SF3B1 .
SF3B1 Phosphorylation: Active, phosphorylated SF3B1 (Thr313) displaces SAP30BP, enabling RBM17 to anchor the U2 snRNP to the branch site .
Spliceosome Assembly: RBM17’s G-patch motif stabilizes interactions with U2 snRNP, enabling excision of short introns with weak PPT signals .
This mechanism is validated by siRNA knockdown experiments, where SAP30BP or RBM17 depletion specifically inhibits splicing of RBM17-dependent introns (e.g., HNRNPH1) .
Leukemic Stem Cell (LSC) Maintenance: RBM17 is overexpressed in AML LSCs, promoting survival and self-renewal. Knockdown reduces colony formation by 60–80% and impairs engraftment in murine models .
Drug Resistance: Overexpression in solid tumors confers resistance to doxorubicin and cisplatin by unclear mechanisms .
NSCLC Immunotherapy: RBM17 expression correlates with improved response to immune checkpoint inhibitors (ICIs) in PD-L1-low patients (ORR: 45% vs. 18%, ) .
AML Targeting: siRNA-mediated RBM17 knockdown induces myeloid differentiation and reduces leukemic burden in vivo .
Splice-Modulating Drugs: Inhibitors of RBM17-SAP30BP or RBM17-SF3B1 interactions could block splicing in RBM17-dependent cancers .
Immunotherapy Optimization: RBM17 expression may stratify NSCLC patients for ICI monotherapy vs. chemo-immunotherapy .
Splicing Specificity: RBM17 mediates splicing in ~8% of human short introns, typically lacking canonical PPT signals .
Evolutionary Conservation: The RBM17-SAP30BP interaction is conserved in Drosophila, suggesting a fundamental role in metazoan splicing .
Phospho-Dependent Recruitment: RBM17 preferentially binds SF3B1 phosphorylated at Thr313, a marker of active spliceosomes .
RBM17 (also known as SPF45) is a splicing factor that plays a critical role in pre-mRNA processing, particularly in a subset of human short introns. Recent research has revealed that the well-established pre-mRNA splicing mechanism involving U2AF heterodimer cannot function efficiently in certain short introns. Instead, a novel mechanism has been discovered where RBM17 forms a complex with SAP30BP to mediate splicing of short introns with truncated polypyrimidine tracts (PPTs) .
The mechanism involves SAP30BP guiding RBM17 to active early spliceosomes, which is particularly important because RBM17 cannot directly bind to truncated PPTs in vitro. This discovery challenges the conventional understanding that U2AF heterodimer (U2AF2–U2AF1) mediates early splicing reactions in all introns regardless of length .
Methodologically, researchers investigating RBM17's fundamental role should employ both knockdown experiments and rescue studies, combined with comprehensive RNA-seq analysis to identify global changes in splicing patterns.
RBM17 expression patterns across human tissues can be analyzed using resources like the Human Protein Atlas. In brain tissue specifically, RBM17 expression data is organized across 13 main brain structures, representing the maximum values found in constituent brain areas .
To properly investigate tissue-specific expression, researchers should:
Utilize RNA-seq data from resources like GTEx and FANTOM5 to compare expression levels across tissues
Employ tissue microarrays with validated antibodies for protein-level confirmation
Consider single-cell RNA-seq to understand cell type-specific expression within tissues
Correlate expression patterns with tissue-specific splicing events to establish functional relevance
The tissue distribution of RBM17 may help explain tissue-specific splicing regulation and potentially illuminate why certain diseases associated with RBM17 dysfunction affect specific tissues more than others.
RBM17 has been identified as a critical factor in leukemic stem cell (LSC) maintenance in acute myeloid leukemia (AML). Research demonstrates that RBM17 upregulation preferentially marks and sustains LSCs and directly correlates with shortened patient survival .
Experimental evidence shows that RBM17 knockdown in primary AML cells leads to:
Increased myeloid differentiation
Impaired colony formation capacity
Reduced in vivo engraftment potential
Through integrative multi-omics analyses, researchers have discovered that RBM17 repression causes inclusion of poison exons and production of nonsense-mediated decay (NMD)-sensitive transcripts for pro-leukemic factors and the translation initiation factor EIF4A2 . This mechanistic understanding provides a rationale for targeting RBM17 or its downstream NMD-sensitive splicing substrates as a potential AML treatment strategy.
For researchers investigating this area, recommended methodologies include:
Patient-derived xenograft models to assess effects on LSC function in vivo
Colony-forming assays to measure self-renewal capacity
RNA-seq combined with rMATS analysis to identify relevant splicing changes
Proteomic analysis to confirm downstream effects on translation and ribosome biogenesis
RBM17 has been found to be overexpressed in glioma patients, and this overexpression correlates with poor prognosis. Research indicates that downregulation of RBM17 suppresses proliferation and induces apoptosis in glioma cells .
The underlying mechanisms appear to involve RBM17's control over apoptosis and proliferation pathways, potentially through its splicing regulatory function. One pathway potentially involved is the Fas receptor signaling pathway, which regulates apoptosis and immune responses .
For researchers investigating RBM17's role in glioma:
Patient tissue analysis should compare RBM17 expression levels between different grades of glioma and correlate with survival data
Functional studies should employ both in vitro and in vivo models with RBM17 knockdown or overexpression
Mechanistic studies should focus on identifying glioma-specific splicing targets of RBM17 using RNA-seq
Therapeutic potential could be explored by testing RBM17 inhibition in combination with standard glioma treatments
Based on published research, the following comprehensive RNA-seq methodology is recommended for investigating RBM17-dependent splicing:
Sample preparation:
Perform siRNA-mediated knockdown using two independent RBM17-targeted siRNAs
Include appropriate control siRNAs (e.g., universal negative control siRNA)
Extract total RNA using high-quality isolation kits (e.g., NucleoSpin RNA kit)
Enrich for poly(A) mRNA using magnetic isolation modules
Library preparation and sequencing:
Prepare RNA libraries using directional RNA library prep kits
Sequence using high-throughput platforms (e.g., NovaSeq6000) with 150bp paired-end strategy
Aim for sufficient depth to detect alternative splicing events (≥30M reads per sample)
Bioinformatic analysis:
This methodology has successfully identified RBM17-dependent splicing events in previous studies and provides a robust framework for further investigations.
To study the RBM17-SAP30BP complex and its functional significance, researchers should employ an integrated approach:
Interaction characterization:
Co-immunoprecipitation to confirm the interaction in different cell types
Proximity ligation assays to visualize the interaction in situ
Structural studies (X-ray crystallography or cryo-EM) to determine interaction interfaces
Functional validation:
Compare knockdown effects of RBM17 alone versus SAP30BP alone versus double knockdown
RNA-seq analysis to identify splicing events dependent on either or both factors
Rescue experiments with wild-type and mutant proteins to identify critical interaction domains
Mechanistic investigation:
RNA-protein interaction studies (CLIP-seq) to map binding sites on target RNAs
In vitro splicing assays with purified components to reconstitute the activity
Live-cell imaging to track the dynamics of complex formation during splicing
This comprehensive approach would provide insights into how the RBM17-SAP30BP complex functions in splicing short introns with truncated polypyrimidine tracts, a process that cannot be effectively carried out by the canonical U2AF heterodimer .
The discovery of RBM17-SAP30BP-dependent splicing represents a paradigm shift in understanding pre-mRNA processing. A comprehensive comparison between these mechanisms reveals:
Feature | RBM17-SAP30BP Splicing | U2AF-dependent Splicing |
---|---|---|
Intron characteristics | Short introns with truncated polypyrimidine tracts | Various introns with sufficient polypyrimidine tracts |
Recognition factors | RBM17-SAP30BP complex | U2AF heterodimer (U2AF2-U2AF1) |
Binding mechanism | SAP30BP guides RBM17 to active early spliceosomes | Direct binding of U2AF2 to polypyrimidine tract |
Sequence requirements | Functions with truncated PPTs | Requires sufficient PPT length |
Evolutionary consideration | Potentially evolved to handle constraints in short introns | Canonical mechanism for most introns |
Researchers investigating these differences should:
Perform comparative binding studies with model pre-mRNAs
Use mutational analysis to define minimal sequence requirements
Employ in vitro splicing assays with purified components to directly compare efficiencies
Analyze the evolutionary conservation of both pathways across species
To effectively identify and characterize RBM17-dependent alternative splicing events, researchers should implement a multi-step computational workflow:
Data generation:
Perform RNA-seq following RBM17 knockdown/knockout
Include both control and experimental conditions with sufficient replicates
Consider time-course experiments to capture primary versus secondary effects
Primary analysis pipeline:
Advanced characterization:
Integration with other data types:
Correlation with RBM17 binding sites from CLIP-seq data
Functional annotation of affected genes and pathways
Prediction of NMD sensitivity for alternatively spliced transcripts
Translation efficiency analysis for affected transcripts
This comprehensive approach enables not only identification of RBM17-dependent events but also mechanistic insights into the rules governing RBM17's splicing activity.
Integrative multi-omics analyses have revealed a critical connection between RBM17 and nonsense-mediated decay (NMD). RBM17 repression leads to inclusion of poison exons and production of NMD-sensitive transcripts for pro-leukemic factors and the translation initiation factor EIF4A2 .
This connection presents several research directions:
Mechanistic investigation:
Identify the sequence features of RBM17-regulated poison exons
Determine whether RBM17 directly suppresses poison exon inclusion or acts through intermediaries
Assess whether RBM17 affects other aspects of NMD beyond regulating poison exon inclusion
Functional significance:
Compare the transcriptome-wide effects of RBM17 knockdown with and without NMD inhibition
Identify the subset of RBM17 splicing targets that are most sensitive to NMD
Determine whether RBM17-regulated NMD contributes to cancer progression
Therapeutic implications:
Evaluate whether combined inhibition of RBM17 and NMD would have synergistic effects
Identify cancer types most susceptible to disruption of this regulatory axis
Develop strategies to selectively target RBM17-regulated NMD-sensitive transcripts
For researchers pursuing this question, a combination of transcriptome profiling, functional validation, and mechanistic dissection would be essential to fully understand how RBM17 leverages NMD to maintain cancer cell survival.
The discovery that SAP30BP guides RBM17 to active early spliceosomes represents a novel regulatory mechanism in RNA processing . To fully understand the impact of this interaction on gene expression programs:
Context-dependent interaction analysis:
Compare the SAP30BP-RBM17 interaction strength across different cell types
Investigate how the interaction is regulated during cell differentiation or stress
Determine whether specific signaling pathways modulate the interaction
Genome-wide splicing impact:
Perform comparative RNA-seq analysis after knockdown of either RBM17, SAP30BP, or both
Identify splicing events that specifically depend on the interaction versus independent roles
Characterize the features of introns most affected by disruption of the interaction
Downstream effects on gene expression programs:
Analyze how disruption of the interaction affects specific cellular pathways
Determine tissue-specific consequences of interaction inhibition
Identify whether certain disease states show altered SAP30BP-RBM17 interaction
This research would provide valuable insights into how the SAP30BP-RBM17 complex functions as a regulatory hub in RNA processing and potentially identify new therapeutic opportunities.
Given RBM17's critical role in cancer biology, particularly in maintaining leukemic stem cells and promoting glioma progression , several therapeutic strategies could be considered:
Direct inhibition approaches:
Small molecule inhibitors targeting RBM17's RNA binding domain
Peptide-based inhibitors disrupting RBM17-SAP30BP interaction
Antisense oligonucleotides to downregulate RBM17 expression
PROTAC-based degradation of RBM17 protein
Indirect targeting strategies:
Inhibition of downstream effectors like EIF4A2, which is enriched in LSCs
Targeting specific RBM17-regulated splicing events critical for cancer maintenance
Combination with NMD inhibitors to enhance the effect of poison exon inclusion
Synthetic lethality approaches based on cancer-specific RBM17 dependencies
Considerations for clinical development:
Biomarker development to identify patients most likely to respond
Evaluation of potential on-target toxicities in normal tissues
Assessment of resistance mechanisms that might emerge
Rational combination strategies with standard-of-care treatments
The research provides "a rationale to target RBM17 and/or its downstream NMD-sensitive splicing substrates for AML treatment" , and similar approaches could be explored for other cancers like glioma where RBM17 plays a pro-tumorigenic role .
To develop clinically relevant biomarkers based on RBM17 activity:
Expression-based biomarkers:
Quantify RBM17 mRNA and protein levels in patient samples
Correlate expression with clinical outcomes across cancer types
Develop standardized assays for clinical implementation
Establish threshold values for stratifying patients
Splicing signature biomarkers:
Identify a panel of RBM17-dependent splicing events detectable in patient samples
Develop RT-PCR or NanoString-based assays to quantify these events
Validate the signature in retrospective patient cohorts
Assess prognostic and predictive value in prospective studies
Functional biomarkers:
Develop assays to measure RBM17 activity in patient-derived cells
Correlate functional measurements with treatment response
Explore liquid biopsy approaches to monitor RBM17 activity during treatment
Integrate with other prognostic factors for comprehensive risk assessment
Research has already demonstrated that RBM17 upregulation correlates with shortened patient survival in AML and poor prognosis in glioma , providing a foundation for biomarker development across multiple cancer types.
Several high-priority research questions emerge from the current understanding of RBM17:
Developmental biology:
How does RBM17 expression and function change during embryonic development?
What are the consequences of RBM17 knockout in specific lineages during development?
Does RBM17-SAP30BP-mediated splicing play tissue-specific roles during organogenesis?
Disease mechanisms:
Beyond cancer, does RBM17 dysregulation contribute to neurodegenerative or cardiovascular diseases?
How does RBM17 respond to cellular stress conditions like hypoxia or DNA damage?
Are there genetic variations in RBM17 or its binding sites associated with disease susceptibility?
Regulatory mechanisms:
How is RBM17 expression and activity regulated at transcriptional, post-transcriptional, and post-translational levels?
What signaling pathways modulate the RBM17-SAP30BP interaction?
Does RBM17 have functions beyond splicing regulation, such as roles in transcription or translation?
Evolutionary perspectives:
How conserved is the RBM17-SAP30BP-dependent splicing mechanism across species?
Did this mechanism evolve specifically to handle constraints in short introns with truncated polypyrimidine tracts?
Are there species-specific adaptations in RBM17 function related to genome complexity?
Addressing these questions will provide a more comprehensive understanding of RBM17's place in cellular physiology and pathology.
Emerging technologies that could significantly advance RBM17 research include:
Single-cell technologies:
Single-cell RNA-seq to reveal cell-type-specific RBM17-dependent splicing events
Single-cell proteomics to correlate RBM17 protein levels with cellular phenotypes
Spatial transcriptomics to map RBM17 activity in tissue contexts
Genome engineering:
CRISPR-based screening to identify synthetic lethal interactions with RBM17
Base editing to introduce specific mutations in RBM17 or its binding sites
CRISPRi/CRISPRa for temporal control of RBM17 expression
Structural biology:
Cryo-EM to visualize the RBM17-SAP30BP complex bound to RNA substrates
Hydrogen-deuterium exchange mass spectrometry to map protein-protein interaction surfaces
Integrative structural modeling to understand complex assembly dynamics
High-throughput functional assays:
Massively parallel reporter assays to characterize RBM17 binding preferences
CRISPR tiling screens to identify functional domains within RBM17
Synthetic splicing constructs to systematically test sequence requirements
These technological advances would enable researchers to address current knowledge gaps and develop a more comprehensive understanding of RBM17 biology in health and disease.
RBM17 is a crucial component of the spliceosome complex, which is involved in the second catalytic step of mRNA splicing . The protein binds to the single-stranded 3’AG at the exon/intron border and promotes its utilization in the splicing process . This function is essential for the regulation of alternative splicing and the utilization of cryptic splice sites .
The primary function of RBM17 is to facilitate the splicing of pre-mRNA by promoting the utilization of specific splice sites . This role is critical for the proper processing of mRNA and the regulation of gene expression. Additionally, RBM17 is involved in the regulation of alternative splicing, which allows for the generation of multiple protein isoforms from a single gene .
The expression and activity of RBM17 are regulated at multiple levels, including transcriptional, post-transcriptional, and post-translational modifications . Alternative splicing of the RBM17 gene itself results in multiple transcript variants, which may have distinct functions and regulatory mechanisms .