RBM3 contains a single RNA recognition motif (RRM) domain and glycine-rich regions, enabling RNA/DNA interactions. The N-terminal RRM domain adopts a βαββαβ topology, forming an RNA-binding interface via hydrogen bonds, π-π stacking, and π-cation interactions . The C-terminal glycine-rich regions are disordered and mediate interactions with ribosomal subunits and microRNAs (miRNAs) .
Domain | Function | Key Interactions |
---|---|---|
RRM (N-terminal) | RNA/DNA binding | mRNA stability, translation |
Glycine-rich (C-terminal) | Ribosome/miRNA interactions | 60S ribosomal subunits, miRNAs |
Cold Stress Adaptation: Induced by hypothermia, RBM3 stabilizes mRNAs and maintains translation efficiency during cold shock .
mRNA Regulation: Enhances global protein synthesis by binding to 60S ribosomal subunits and suppressing miRNA activity .
Anti-Apoptotic Effects: Inhibits p38 MAPK signaling and caspase-3 activation, protecting cells from stress-induced apoptosis .
RBM3 exhibits tissue-specific roles in cancer progression and prognosis:
Breast Cancer: RBM3 binds the 3'UTR of ARPC2, enhancing actin remodeling and metastasis .
Ovarian Cancer: High RBM3 correlates with shorter survival and cisplatin resistance, linked to DNA integrity checkpoint genes (e.g., MCM3) .
Neuroprotection: Overexpression in Alzheimer’s and prion disease models restores synaptic plasticity and delays neurodegeneration .
RBM3 modulates RhoA-ROCK signaling, influencing cell spreading and migration:
Spreading Initiation: Localizes to filopodia and blebs, regulating RhoA expression .
Migration Mode: Enhances mesenchymal migration via long protrusions; knockdown induces amoeboid migration .
p38 Inhibition: Blocks NO-induced apoptosis in neuroblastoma cells by suppressing p38 phosphorylation .
miRNA Regulation: Reduces miR-143 levels, mitigating cytotoxicity in hypoxic conditions .
RBM3 is widely expressed in human tissues, with nuclear localization dominating in most cell types :
Tissue | Expression Level | Localization | Source |
---|---|---|---|
Brain (Hippocampus) | High | Nucleoplasm | |
Breast | Moderate | Nuclear/Cytoplasmic | |
Liver | Low | N/A | |
Skeletal Muscle | High | Cytoplasm |
Cancer Therapy: Targeting RBM3 in breast/ovarian cancers may reduce metastasis, while enhancing its expression in colorectal cancer could improve prognosis .
Neurodegeneration: Mimicking RBM3’s cold-shock response (e.g., hypothermia) or overexpressing RBM3 may combat synaptic loss in Alzheimer’s disease .
Stress Responses: RBM3’s role in ER stress and hypoxia suggests potential applications in ischemia-reperfusion injury .
Tissue-Specific Functions: Clarifying RBM3’s oncogenic vs. tumor-suppressive roles in distinct cancers.
Structural Dynamics: Elucidating how RBM3’s disordered regions mediate ribosome/miRNA interactions .
Translational Potential: Developing RBM3-based therapies for neurodegeneration and metastatic cancers.
RBM3 is a cold-shock protein that is ubiquitously expressed throughout the human body in a temperature-dependent manner. It belongs to the family of RNA-binding proteins and contains an RNA recognition motif. In the human brain, RBM3 is particularly expressed in areas with high translational rates, similar to what has been observed in cerebral tissue of adult rats .
When studying RBM3 expression patterns, researchers should consider:
Temperature-dependent variation (highest expression at mild hypothermia, around 32-33.5°C)
Age-related expression differences (higher in developing brains)
Regional expression variations across different brain areas
Methodologically, immunohistochemistry combined with quantitative RT-PCR provides the most reliable assessment of RBM3 distribution across human tissues. Western blot analysis should be used to confirm protein-level expression.
RBM3 expression is regulated through multiple mechanisms:
Temperature-controlled alternative splicing coupled to nonsense-mediated decay (NMD):
TrkB signaling pathway:
Splicing factor regulation:
RBM3 influences multiple cellular functions in human neurons:
Protein synthesis regulation:
Cell survival:
Synaptic plasticity:
RNA processing:
Affects transcriptome-wide pre-mRNA splicing
Deficiency leads to widespread splicing alterations that can be reversed through RBM3 cDNA co-expression
When designing studies to investigate these functions, researchers should employ multiple complementary approaches, including gain-of-function and loss-of-function experiments in relevant neuronal models.
The temperature-sensitive alternative splicing of RBM3 is a sophisticated regulatory mechanism:
Poison exon structure and evolution:
Temperature response gradient:
E3a inclusion responds gradually to temperature changes within the physiologically relevant temperature range (33-39°C)
At warmer temperatures (38-39°C), E3a inclusion increases, leading to NMD-mediated degradation
At cooler temperatures (33-34°C), E3a is predominantly skipped, resulting in productive RBM3 expression
Experimental validation:
Using splicing-sensitive RT-PCR to detect temperature-dependent isoforms
Employing cycloheximide to stabilize NMD-targeted transcripts for detection
Developing minigene constructs to study cis-regulatory elements involved in temperature-sensitive splicing
RBM3's neuroprotective effects in neurodegenerative disorders are multifaceted:
Synapse preservation and regeneration:
Neuronal survival promotion:
Neurogenesis stimulation:
Disease-modifying effects:
Combining in vitro models (neuronal cultures) with in vivo disease models
Assessing multiple endpoints (cellular, synaptic, behavioral)
Investigating both acute and chronic effects of RBM3 modulation
The relationship between TrkB signaling and RBM3 expression reveals a regulatory circuit:
Activation pathway:
Feedback regulation:
Therapeutic implications:
Pharmacological manipulation of TrkB signaling combined with RBM3 expression analysis
Assessment of downstream signaling components (PLCγ1, pCREB, DUSP6)
Testing TrkB agonists for RBM3 induction in absence of cooling
RBM3's impact on RNA processing and protein synthesis is complex:
Global splicing regulation:
Ribosomal interaction:
Cold-stress response coordination:
RNA-seq to detect global splicing changes
Ribosome profiling to assess translation efficiency
CLIP-seq to identify direct RNA binding targets of RBM3
Polysome profiling to evaluate effects on translation
Several approaches can be employed to modulate RBM3 expression independent of temperature:
Antisense oligonucleotide (ASO) therapy:
ASOs targeting the poison exon (E3a) can increase RBM3 expression at normal temperatures
Target site optimization:
In vivo application:
TrkB pathway modulation:
Genetic manipulation:
Dose-response relationships for ASOs or TrkB modulators
Pharmacokinetics and tissue distribution of compounds
Cell-type specific responses to RBM3 modulation
Optimal experimental models for studying RBM3 in neuroprotection include:
In vitro models:
Human SK-N-SH neurons:
Primary mouse hippocampal neurons:
Neurodegenerative disease models:
Prion disease model (tg37⁺/⁻ mice):
Alzheimer's disease models:
Molecular tools:
RBM3 minigenes:
CRISPR/Cas9-modified cell lines:
To comprehensively assess RBM3-mediated effects on synaptic plasticity, researchers should employ:
Morphological analysis:
Functional assessment:
Electrophysiological recordings (patch-clamp, field potentials)
Calcium imaging to measure synaptic activity
Analysis of synaptic protein expression and localization
Behavioral testing:
Molecular profiling:
Transcriptomic analysis of synaptic components
Proteomic analysis of synaptic fractions
Assessment of local translation at synapses
The integration of these techniques provides a comprehensive understanding of how RBM3 influences synaptic plasticity across multiple scales.
Optimization of ASOs for targeting RBM3 poison exon splicing requires systematic approach:
Target site identification:
Chemistry optimization:
Delivery optimization:
Validation approaches:
RT-PCR to confirm reduced E3a inclusion
Western blot to verify increased RBM3 protein levels
Functional assays to confirm neuroprotective effects
This structured approach to ASO development has demonstrated remarkable efficacy in preclinical models, with potential applications in diverse neurological conditions.
RBM3 modulation holds substantial therapeutic potential across multiple neurological disorders:
Acute neurological conditions:
Neurodegenerative disorders:
Advantages over hypothermia:
Current evidence:
Safety and efficacy studies in large animal models
Biomarker development to monitor RBM3 induction
Combination therapies targeting multiple neuroprotective mechanisms
Despite promising preclinical results, several challenges must be addressed:
Delivery challenges:
Safety considerations:
Patient selection:
Identifying appropriate patient populations for clinical trials
Developing biomarkers predictive of response
Determining optimal timing of intervention
Regulatory pathway:
Comparative studies of different RBM3-inducing approaches
Development of companion diagnostics
Careful design of early-phase clinical trials
RBM3 is known to be induced by cold shock and low oxygen tension . This induction is part of the cellular response to stress, allowing cells to adapt to harsh conditions. The expression of RBM3 is regulated at the transcriptional level, and multiple alternatively spliced transcript variants have been identified .
RBM3 enhances global protein synthesis at both physiological and mild hypothermic temperatures . It reduces the relative abundance of microRNAs when overexpressed and enhances the phosphorylation of translation initiation factors, leading to active polysome formation . These functions are crucial for maintaining cellular homeostasis and promoting cell survival under stress conditions.
Human recombinant RBM3 is used in various research applications to study its role in cellular processes and its potential as a therapeutic target. The recombinant protein is produced using advanced biotechnological methods to ensure high purity and activity.