RBM11 exhibits restricted expression in normal tissues, correlating with cellular differentiation:
Expression is induced during differentiation in neuronal (e.g., SH-SY5Y cells) and germ cell lines .
RBM11 regulates alternative splicing (AS) and interacts with key splicing factors:
Antagonism of SRSF1:
Interaction Network:
Key partners include:
These interactions highlight RBM11’s role in coordinating splicing and RNA surveillance .
RBM11 is implicated in oncogenesis, particularly in glioblastoma and ovarian cancer:
Akt/mTOR Activation:
Prognostic Biomarker:
Cancer Type | Expression | Functional Impact | Source |
---|---|---|---|
Glioblastoma | Overexpressed | Promotes cell proliferation and invasion | |
Ovarian Cancer | Overexpressed | Activates Akt/mTOR; poor prognosis |
RNA Binding Specificity: Preferential poly(U) binding is critical for splicing regulation .
Therapeutic Targeting: Inhibition of RBM11 may suppress cancer progression by disrupting Akt/mTOR signaling .
Limited understanding of RBM11’s role in neurodevelopmental disorders (e.g., Down syndrome).
Need for validated inhibitors to test therapeutic potential.
RBM11 (RNA-Binding Motif Protein 11) is an RNA splicing factor containing an RNA Recognition Motif (RRM) at the amino terminus (N-terminal). It functions in posttranscriptional control of RNA metabolism through mechanisms like alternative splicing and RNA modification. RBM11 expression has been observed in multiple normal human tissues, including the brain, testis, and spleen . Functionally, RBM11 participates in RNA processing events that regulate gene expression, though its precise physiological function has not been fully characterized. Recent research has identified RBM11 as a potential oncogenic protein involved in cancer progression through activating signaling pathways such as Akt/mTOR .
Method | Application | Advantages | Limitations |
---|---|---|---|
qRT-PCR | mRNA quantification | High sensitivity, quantitative | Does not assess protein levels |
Western Blot | Protein detection | Protein size confirmation, semi-quantitative | Lower throughput |
IHC | Tissue localization | Spatial context in tissues, clinically applicable | Semi-quantitative |
RNA-seq | Transcriptome-wide expression | Comprehensive, can detect splice variants | Requires bioinformatic expertise |
For reliable RBM11 expression analysis, researchers should employ multiple complementary techniques. In published studies, anti-RBM11 antibodies (such as 17220-1-AP from Proteintech) have been validated for both Western blot and IHC applications . When designing expression studies, include appropriate housekeeping genes or proteins (β-actin for Western blots) as loading controls and validate antibody specificity using positive and negative controls.
Studying RBM11's role in cancer progression requires a multi-faceted experimental approach combining in vitro and in vivo methodologies:
Gene Silencing: Implement RBM11 knockdown using validated shRNAs. Published successful targeting sequences include 5′-GTT CCG AAA GTC TAA GAA GAA-3′ and 5′-CCC AGC TCA TAT AAA TGG ACT-3′ . Always validate knockdown efficiency by both qRT-PCR and Western blot.
Overexpression Studies: Utilize flag-tagged RBM11 plasmids for ectopic expression experiments to confirm phenotypic effects are reproducible with gain-of-function .
Functional Assays:
Cell proliferation: MTT/CCK-8 assays and clonogenic formation assays
Cell invasion: Transwell assays with Matrigel coating
Signaling pathway analysis: Western blots for key pathway components (e.g., phosphorylated and total Akt, mTOR)
In Vivo Models: Xenograft models using cancer cell lines with manipulated RBM11 expression provide critical validation of in vitro findings. Measure tumor growth rates and analyze proliferation markers (e.g., Ki67) in tumor tissues by IHC .
Patient Sample Analysis: Correlate RBM11 expression levels with clinical outcomes using tissue microarrays and survival analysis. High RBM11 expression has been associated with poor survival in ovarian cancer patients .
For robust manipulation of RBM11 expression, researchers should consider the following validated approaches:
For RBM11 Knockdown:
shRNA-mediated silencing using lentiviral vectors for stable integration
CRISPR-Cas9-mediated knockout for complete elimination of expression
siRNA for transient knockdown in initial screening experiments
For RBM11 Overexpression:
Transfection with flag-tagged RBM11 expression vectors using lipofectamine 3000 reagent in ovarian cancer cell lines (A2780, OVCAR-3)
Doxycycline-inducible expression systems for controlled temporal studies
Viral vectors for difficult-to-transfect cell types
Critical Considerations:
Always validate expression changes at both mRNA and protein levels
Include appropriate empty vector and scrambled sequence controls
Optimize transfection conditions for each cell line
Consider rescue experiments with wild-type or mutant RBM11 to confirm specificity
RBM11 positively regulates the Akt/mTOR signaling pathway in ovarian cancer cells through mechanisms that are still being fully characterized. Experimental data shows that:
RBM11 knockdown significantly decreases phosphorylation of Akt (at Ser473) and mTOR (at Ser2448) without affecting total protein levels .
Conversely, overexpression of RBM11 increases phosphorylation of both Akt and mTOR, confirming a positive regulatory relationship .
Potential mechanistic explanations include:
RBM11 may regulate alternative splicing or stability of mRNAs encoding upstream regulators of the Akt pathway
RBM11 could affect translation efficiency of key pathway components
RBM11 might directly interact with pathway components through protein-protein interactions
Research methodologies to further elucidate these mechanisms should include:
RNA immunoprecipitation (RIP) followed by sequencing to identify RBM11-bound transcripts
RNA splicing analysis using RT-PCR or RNA-seq to detect alternative splicing events
Immunoprecipitation followed by mass spectrometry to identify protein interaction partners
Phosphoproteomics to identify changes in the broader signaling network
When facing conflicting data regarding RBM11 function across different studies or cancer types, researchers should implement the following experimental design strategies:
Systematic Replication: Reproduce key experiments using multiple cell lines representing different cancer subtypes or tissues.
Context-Dependent Analysis: Design experiments that directly compare RBM11 function across different contexts:
Cell line panels representing multiple cancer types
Normal vs. cancerous cells from the same tissue origin
Different stages of cancer progression
Comprehensive Pathway Analysis: Examine RBM11's effect on multiple signaling pathways simultaneously:
Use phospho-kinase arrays to screen for differential effects
Implement RNA-seq and proteomic approaches to capture global changes
Validate key findings with targeted assays
Controlled Microenvironment: Test whether RBM11 function varies under different conditions:
Hypoxia vs. normoxia
Different growth factor stimulation
2D vs. 3D culture systems
Genetic Background Consideration: Introduce RBM11 manipulations in isogenic cell lines with defined genetic alterations to identify potential interactions with other cancer-related genes.
When designing RBM11 knockdown experiments, researchers should implement these critical considerations:
Target Selection and Validation:
Design at least two independent shRNAs/siRNAs targeting different regions of RBM11
Validate knockdown efficiency at both mRNA (qRT-PCR) and protein levels (Western blot)
Include appropriate non-targeting controls
Cell Line Selection:
Choose cell lines with confirmed endogenous RBM11 expression
Include multiple cell lines to ensure findings aren't cell line-specific
Consider using paired isogenic cell lines when possible
Phenotypic Assays:
Assess proliferation using multiple time points and methods (MTT/CCK-8 and colony formation)
Measure invasion using standardized transwell assays with appropriate controls
Examine effects on apoptosis and cell cycle progression
Signaling Pathway Analysis:
Monitor phosphorylation status of key Akt/mTOR pathway components (pAkt S473, pmTOR S2448)
Assess total protein levels to rule out degradation effects
Include downstream effectors to confirm pathway inhibition
Rescue Experiments:
Reintroduce shRNA-resistant RBM11 constructs to confirm specificity
Consider using domain-specific mutants to identify functional regions
For robust in vivo evaluation of RBM11 function, researchers should consider these validated models:
Xenograft Models:
Patient-Derived Xenografts (PDXs):
Maintain tumor heterogeneity and microenvironment
Allow for studying RBM11 in different genetic backgrounds
Require RBM11 manipulation through viral delivery or pharmacological approaches
Orthotopic Models:
Better recapitulate the native tumor microenvironment
For ovarian cancer, intraperitoneal injection allows assessment of metastatic potential
Requires specialized imaging techniques for longitudinal monitoring
Genetic Mouse Models:
Consider conditional RBM11 knockout/transgenic models for tissue-specific studies
Can evaluate developmental and tissue-specific functions
May require extensive breeding and characterization
Analysis Parameters:
Tumor growth rate (volume measurements)
Immunohistochemistry for proliferation markers (Ki67)
Assessment of Akt/mTOR pathway activation in tumor tissues
Metastatic burden evaluation when applicable
To comprehensively analyze RBM11's impact on RNA processing, researchers should implement these methodological approaches:
RNA-Binding Profiling:
CLIP-seq (Cross-linking and immunoprecipitation followed by sequencing) to identify direct RNA targets
RIP-seq (RNA immunoprecipitation sequencing) to capture RBM11-associated transcripts
PAR-CLIP for enhanced crosslinking efficiency and precise binding site identification
Splicing Analysis:
RT-PCR with exon-spanning primers to detect alternative splicing events
RNA-seq with specialized computational pipelines for global splicing analysis
Minigene assays to validate specific splicing events in reporter systems
RNA Stability Assessment:
Actinomycin D chase experiments to measure half-life of candidate transcripts
Pulse-chase labeling with modified nucleosides to track newly synthesized RNA
Polysome profiling to assess translation efficiency
Functional Validation:
Rescue experiments using wild-type and mutant versions of identified targets
CRISPR/Cas9 editing of RBM11 binding sites in target RNAs
Structural analysis of RBM11-RNA complexes
Data Analysis Pipeline:
Motif discovery algorithms to identify consensus binding sequences
Integration with proteomic data to correlate RNA changes with protein outcomes
Pathway enrichment analysis of affected transcripts
When facing contradictions between in vitro and in vivo RBM11 studies, researchers should implement this analytical framework:
Systematic Comparison:
Create a detailed comparison table of experimental conditions, cell types, and endpoints
Identify specific variables that differ between systems (growth factors, oxygen levels, etc.)
Determine whether contradictions are complete or context-dependent
Biological Context Considerations:
Tumor microenvironment influence (absent in vitro)
Duration of experiments (acute vs. chronic effects)
Systemic factors present only in vivo (hormones, immune components)
Three-dimensional architecture and cell-cell interactions
Technical Validation:
Verify antibody specificity in both systems
Confirm knockdown/overexpression efficiency is comparable
Assess for compensatory mechanisms that might emerge in vivo
Reconciliation Strategies:
Develop intermediate models (3D organoids, co-culture systems)
Manipulate specific microenvironmental factors in vitro
Conduct time-course studies to capture dynamic effects
Implement more sophisticated in vivo models (orthotopic vs. subcutaneous)
Interpretation Framework:
Consider that contradictions may reveal context-dependent functions
Evaluate whether differences reflect technical limitations or biological reality
Develop integrated models that accommodate conditional functions
For robust statistical analysis of RBM11 expression in patient cohorts, researchers should implement these approaches:
Expression Analysis:
Normalize RBM11 expression against validated housekeeping genes
Use box plots or violin plots to visualize distribution across groups
Apply appropriate parametric (t-test, ANOVA) or non-parametric tests (Mann-Whitney, Kruskal-Wallis) based on data distribution
Survival Analysis:
Kaplan-Meier curves stratified by RBM11 expression levels
Cox proportional hazards models for multivariate analysis
Determine optimal cutoff values using ROC curve analysis or quartile distribution
Correlation Studies:
Spearman or Pearson correlation with clinical parameters
Multiple testing correction (Bonferroni, FDR) for genome-wide analyses
Multivariate regression to account for confounding variables
Cohort Considerations:
Power analysis to determine adequate sample size
Stratification by cancer subtype, stage, and treatment history
Independent validation cohorts to confirm findings
Visualization and Reporting:
Forest plots for hazard ratios across subgroups
Heatmaps for correlation with other molecular markers
Transparent reporting of all statistical parameters (sample sizes, p-values, confidence intervals)
To distinguish direct from indirect effects of RBM11 on cellular phenotypes, implement these methodological approaches:
Temporal Analysis:
Time-course experiments after RBM11 manipulation
Pulse-induction systems (e.g., doxycycline-inducible) to capture immediate effects
Monitor sequential activation of signaling events
Direct Target Identification:
CLIP-seq to identify directly bound RNA targets
Structure-function analysis using RBM11 mutants lacking RNA-binding capacity
In vitro binding assays with purified components
Pathway Dissection:
Selective inhibitors of downstream pathways (e.g., Akt/mTOR inhibitors)
Genetic manipulation of pathway components in combination with RBM11
Phosphoproteomics to map signaling cascades
Rescue Experiments:
Restore expression of specific RBM11 targets to reverse phenotypes
Express constitutively active downstream effectors
Use domain-specific RBM11 mutants to dissect functional regions
System-Level Analysis:
Integrate transcriptomic, proteomic, and phenotypic data
Network analysis to identify direct regulatory relationships
Mathematical modeling of signaling dynamics
Several cutting-edge technologies hold promise for elucidating RBM11 function:
CRISPR Screening Approaches:
Genome-wide CRISPR screens to identify synthetic lethal interactions with RBM11
CRISPRi/CRISPRa for fine-tuned modulation of RBM11 expression
Base editing of endogenous RBM11 regulatory elements
Single-Cell Technologies:
scRNA-seq to capture heterogeneous responses to RBM11 manipulation
Spatial transcriptomics to map RBM11 activity in tissue context
Single-cell proteomics to correlate RNA changes with protein outcomes
Advanced Imaging:
Live-cell imaging of RBM11-RNA interactions using MS2 systems
Super-resolution microscopy to visualize RBM11 in subnuclear structures
FRET-based sensors to monitor RBM11 activity in real-time
Structural Biology:
Cryo-EM of RBM11-RNA complexes
Hydrogen-deuterium exchange mass spectrometry for conformational dynamics
Integrative structural biology combining multiple techniques
Translational Approaches:
Development of small molecule inhibitors targeting RBM11-RNA interactions
RNA therapeutics to modulate RBM11 activity
Biomarker development for patient stratification
Researchers face several methodological challenges when studying RBM11 across cancer types:
Tissue-Specific Functions:
RBM11 may regulate different RNA targets in different tissues
Cell type-specific protein interaction networks may alter function
Baseline expression levels vary across tissues, affecting experimental design
Technical Considerations:
Antibody validation across multiple tissue types
Optimization of transfection/transduction for diverse cell lines
Suitable in vivo models for each cancer type
Context-Dependent Regulation:
Tumor microenvironment varies between cancer types
Genetic background effects on RBM11 function
Treatment history may affect RBM11 activity and dependency
Standardization Challenges:
Consistent measurement methods across studies
Appropriate control selection for each cancer type
Normalization strategies for cross-cancer comparisons
Experimental Design Solutions:
Pan-cancer cell line panels with standardized protocols
Tissue-specific conditional knockout models
Comprehensive multi-omics profiling across cancer types
Meta-analysis frameworks to integrate heterogeneous datasets
The RBM11 gene is located on chromosome 21 and is a protein-coding gene. The protein itself is involved in various cellular processes, primarily related to RNA metabolism. It enables poly (U) RNA binding activity and protein homodimerization activity . The protein is located in nuclear specks, which are subnuclear structures involved in the regulation of gene expression .
RBM11 plays a crucial role in the regulation of alternative mRNA splicing via the spliceosome. It acts upstream of or within the cellular response to oxidative stress . The protein is known to antagonize SRSF1-mediated BCL-X splicing, which may affect the choice of alternative 5’ splice sites by binding to specific sequences in exons . This function is particularly important during neuron and germ cell differentiation .
RBM11 has been associated with various diseases, including papillary cystadenocarcinoma . The dysregulation of RBM proteins, including RBM11, has been linked to the occurrence and development of cancers . Understanding the mechanisms of these proteins in tumorigenesis and development is essential for identifying new therapeutic targets and prognostic markers .