RPL35 is a component of the large 60S ribosomal subunit that participates in protein synthesis. It belongs to the L29P family of ribosomal proteins and is located in the cytoplasm. As part of the ribosome, RPL35 contributes to the translation machinery that converts mRNA into proteins. The ribosome consists of a small 40S subunit and a large 60S subunit, together composed of 4 RNA species and approximately 80 structurally distinct proteins . Gene Ontology annotations indicate that RPL35 possesses RNA binding and mRNA binding capabilities, which are essential for its function in translation .
RPL35 expression follows tissue-specific patterns that can be visualized using dimensionality reduction techniques such as t-distributed stochastic neighbor embedding (t-SNE). Analysis of RNA-seq data from The Cancer Genome Atlas (TCGA) has shown that RPL35, along with other ribosomal proteins, displays expression patterns that strongly correlate with tissue type .
While Principal Component Analysis (PCA) can distinguish tissues based on RPL35 expression to some degree, t-SNE analysis more clearly identifies tissue-specific clusters due to its ability to detect non-linear relationships between ribosomal proteins . This tissue-specific regulation suggests that RPL35 may have specialized functions beyond its canonical role in protein synthesis that contribute to tissue identity and function.
Several experimental models have proven valuable for RPL35 research:
Human cancer cell lines: Neuroblastoma cell lines such as BE(2)-C and Kelly cells are frequently used to study RPL35's role in cancer progression .
E. coli expression systems: These are employed for producing recombinant RPL35 protein for structural and functional studies .
RNA interference models: siRNA-mediated knockdown of RPL35 in cell culture is effective for examining functional consequences on protein synthesis, cell proliferation, and signaling pathways .
Mouse models: NIH3T3 mouse fibroblasts have been used to study the transformative potential of ribosomal proteins in cancer contexts .
Tumor xenograft models: These are valuable for evaluating the in vivo effects of RPL35 modulation on tumor growth and progression.
Each model offers distinct advantages for investigating different aspects of RPL35 biology, from basic molecular interactions to complex disease-related functions.
RPL35 promotes cancer progression in neuroblastoma through several mechanisms:
Regulation of E2F1 translation: RPL35 binds to both long non-coding RNA lncNB1 and E2F1 mRNA, promoting E2F1 protein translation without affecting its mRNA levels . E2F1 is a transcription factor crucial for cell cycle progression.
Activation of oncogenic signaling: RPL35 increases DEPDC1B protein expression, which enhances ERK protein phosphorylation and N-Myc protein phosphorylation at S62 . This signaling cascade promotes neuroblastoma cell proliferation and survival.
Metabolic reprogramming: RPL35 functions as a positive regulator of aerobic glycolysis through the RPL35/ERK/HIF1α axis, supporting the high energy demands of rapidly proliferating cancer cells .
When RPL35 is knocked down in neuroblastoma cell lines, significant reductions are observed in DEPDC1B, N-Myc, and E2F1 protein expression, ERK phosphorylation, and N-Myc phosphorylation at S62 . These findings demonstrate that RPL35 acquires non-canonical functions in cancer cells that extend beyond its role in ribosome biogenesis and protein synthesis.
The interaction between RPL35 and long non-coding RNAs (lncRNAs) represents an emerging paradigm in translational regulation:
LncNB1 interaction: Mass spectrometry analysis has revealed that lncNB1 specifically binds to RPL35 in neuroblastoma cells . This interaction has significant functional consequences for cancer progression.
Mechanism of action: RNA immunoprecipitation assays have demonstrated that:
Functional significance: The lncNB1-RPL35-E2F1 axis promotes neuroblastoma tumorigenesis by enhancing E2F1 protein synthesis, which drives cell proliferation .
This relationship illustrates how RPL35 can function beyond its conventional role in ribosome structure, participating in specialized translation regulation through interactions with lncRNAs. This mechanism may represent a broader paradigm of how ribosomal proteins acquire additional functions in disease contexts.
RPL35 has been identified as a key regulator of multiple signaling pathways involved in cancer progression:
ERK pathway activation:
HIF1α pathway regulation:
N-Myc stabilization:
These interconnected signaling effects demonstrate how RPL35 can exert wide-ranging influences on cellular behavior beyond its primary role in ribosome function. The integration of RPL35 into these signaling networks makes it a potential therapeutic target, particularly in cancers where these pathways are dysregulated.
RPL35 expression patterns have significant diagnostic and prognostic implications in cancer:
Cancer classification models:
Tumor subtyping:
Prognostic stratification:
Tissue-of-origin identification:
These findings suggest that analyzing RPL35 expression as part of broader ribosomal protein expression patterns could become an important tool in cancer diagnostics, prognostication, and therapeutic decision-making.
Several complementary approaches can be used to characterize RPL35 protein-RNA interactions:
RNA Immunoprecipitation (RIP):
This technique has successfully demonstrated RPL35 binding to both lncNB1 RNA and E2F1 mRNA
Protocol: Cells are lysed, and RPL35 protein is immunoprecipitated using a specific anti-RPL35 antibody. Associated RNAs are isolated and analyzed by RT-PCR
Control: Parallel immunoprecipitation with control IgG identifies non-specific binding
Biotin-labeled RNA pull-down:
Identifies proteins that bind to specific RNAs of interest
Protocol: RNA is in vitro-transcribed with biotin labeling, incubated with cell lysates, and protein-RNA complexes are captured using streptavidin beads. Bound proteins are identified by mass spectrometry
This method identified RPL35 as a binding partner of lncNB1 in neuroblastoma cells
Cross-linking and Immunoprecipitation (CLIP):
CLIP methods (including HITS-CLIP, PAR-CLIP, or iCLIP) provide higher resolution mapping of protein-RNA interaction sites
These techniques involve UV cross-linking of protein-RNA complexes in live cells, followed by immunoprecipitation and sequencing
Functional validation assays:
A comprehensive approach combining multiple methods provides more robust evidence for specific RPL35-RNA interactions and their functional consequences.
Several approaches are available for modulating RPL35 expression in experimental settings:
RNA interference (RNAi):
siRNA transfection: Multiple studies have successfully used siRNAs targeting RPL35. Research has employed independent siRNAs to knock down RPL35 in neuroblastoma cell lines
shRNA expression: For stable knockdown, shRNAs can be delivered via lentiviral vectors
Advantages: Relatively easy to implement, cost-effective
Limitations: Incomplete knockdown, potential off-target effects
CRISPR-Cas9 genome editing:
For complete knockout or knock-in studies
Can be used to introduce specific mutations or regulatory elements
Advantages: Complete loss of protein expression, ability to create stable cell lines
Overexpression systems:
Rescue experiments:
Combining knockdown of endogenous RPL35 with expression of siRNA-resistant RPL35 variants
Essential for confirming specificity of observed phenotypes
When modulating RPL35 expression, researchers should carefully consider potential compensatory mechanisms by other ribosomal proteins and effects on global protein synthesis that may confound interpretation of specific RPL35 functions.
To effectively investigate RPL35's role in neuroblastoma progression, a multi-faceted experimental approach is recommended:
Expression analysis in clinical samples:
Quantify RPL35 expression in neuroblastoma tumors versus normal tissue
Correlate expression with clinical parameters (stage, MYCN amplification status, patient outcome)
Methods: Immunohistochemistry, RT-qPCR, or mining public neuroblastoma datasets
Functional studies in cell lines:
Modulate RPL35 expression using siRNA or CRISPR-Cas9 in neuroblastoma cell lines (e.g., BE(2)-C, Kelly)
Assess effects on:
Proliferation: MTT/MTS assays, BrdU incorporation
Survival: Annexin V/PI staining, caspase activation assays
Colony formation: Soft agar assays, clonogenic assays
Mechanistic investigations:
Analyze RPL35's effect on the lncNB1-E2F1 axis:
Examine signaling pathway effects:
Translation-specific analyses:
In vivo studies:
This comprehensive approach enables researchers to establish RPL35's role in neuroblastoma from multiple angles, strengthening the evidence for its contribution to disease progression and therapeutic potential.
Bioinformatic approaches offer powerful tools for analyzing RPL35 expression patterns across tissues and diseases:
Dimensionality reduction techniques:
t-distributed stochastic neighbor embedding (t-SNE) has been shown to effectively identify tissue-specific and cancer-specific clusters based on ribosomal protein expression patterns, including RPL35
This approach outperforms Principal Component Analysis (PCA) for distinguishing tissue types based on ribosomal protein expression profiles
Machine learning classification models:
Artificial neural network (ANN) models based on ribosomal protein expression can classify tumors by tissue of origin with 93% accuracy
Logistic regression models can:
Correlation analysis:
RPL35 expression can be correlated with:
Expression of other genes to identify co-regulated pathways
Clinical parameters to identify prognostic associations
Molecular subtypes to understand disease heterogeneity
Multi-omics integration:
Combining RPL35 expression data with:
Genomic data (mutations, copy number alterations)
Proteomic data (protein levels, post-translational modifications)
Clinical data (survival, treatment response)
Network analysis:
Constructing protein-protein interaction networks to understand RPL35's functional connections
Pathway enrichment analysis to identify biological processes associated with RPL35 dysregulation
These bioinformatic approaches can provide insights into RPL35's tissue-specific functions, role in disease processes, and potential as a biomarker or therapeutic target.
RPL35 has been associated with Diamond-Blackfan Anemia 19 (DBA19), a rare congenital bone marrow failure syndrome characterized by red blood cell aplasia . Key aspects of this association include:
Genetic basis:
Pathophysiological mechanism:
Mutations in RPL35 impair ribosome biogenesis
This leads to nucleolar stress and activation of p53-dependent apoptosis
Erythroid progenitor cells are particularly sensitive to this stress, explaining the predominantly erythroid phenotype
Clinical implications:
Patients typically present with macrocytic anemia in infancy
Associated congenital anomalies may be present
Knowledge of the specific genetic cause (RPL35 mutation) can guide genetic counseling and family screening
Relationship to other DBA forms:
DBA19 (RPL35-associated) shares clinical features with other forms of DBA
This reinforces the central role of ribosome biogenesis in erythropoiesis
It also highlights how mutations in different ribosomal proteins can lead to similar clinical phenotypes
This association between RPL35 and DBA demonstrates how disruption of ribosomal proteins can have tissue-specific effects despite their ubiquitous expression, and underscores the critical importance of proper ribosome biogenesis in erythropoiesis.
RPL35 dysregulation has been linked to multiple cancer types:
Neuroblastoma:
Cancer type identification:
Cancer subtypes:
Some cancer cohorts show subtypes with distinct RPL35 expression patterns
These subtypes correlate with molecular markers, tumor phenotypes, and survival outcomes
For example, some tumors (143 tumors from 15 cohorts) show amplification and relative up-regulation of RPL35 along with other ribosomal proteins
Her2/Neu-amplified cancers:
The diverse roles of RPL35 across cancer types highlight how this ribosomal protein contributes to cancer biology through both canonical functions in protein synthesis and non-canonical roles in specific oncogenic pathways.
RPL35 has emerged as a significant regulator of tumor metabolism, particularly aerobic glycolysis (the Warburg effect):
The RPL35/ERK/HIF1α axis:
Metabolic reprogramming:
By enhancing glycolysis, RPL35 facilitates the metabolic shift that provides cancer cells with:
Rapid ATP production
Biosynthetic intermediates for macromolecule synthesis
Reduced reactive oxygen species generation
This metabolic adaptation supports the high proliferation rates observed in many cancers
Experimental evidence:
Therapeutic implications:
This metabolic role adds another dimension to RPL35's contributions to cancer progression, beyond its effects on protein synthesis and specific oncogenic pathways. It suggests that RPL35 helps coordinate cellular growth signals with the metabolic adaptations required to sustain rapid proliferation in cancer cells.
Several therapeutic approaches targeting RPL35 and its associated pathways show promise for cancer treatment:
Direct targeting of RPL35:
RNA interference-based therapies (siRNA, shRNA) targeting RPL35 have shown efficacy in preclinical models
Development of small molecule inhibitors that disrupt RPL35's non-canonical functions while preserving essential ribosomal functions
Targeting the lncNB1-RPL35-E2F1 axis:
Disrupting the interaction between lncNB1 and RPL35 could inhibit E2F1 translation
E2F1 inhibitors could complement RPL35-targeted approaches in neuroblastoma
Inhibiting downstream pathways:
Combination therapies:
Combining RPL35-targeted treatments with standard chemotherapy agents
Simultaneous targeting of multiple components of the RPL35/ERK/HIF1α pathway
Incorporating metabolic inhibitors that target the glycolytic phenotype promoted by RPL35
Biomarker-guided approaches:
Using RPL35 expression patterns to identify patients most likely to respond to specific therapies
Monitoring changes in RPL35-associated pathways as markers of treatment response
These approaches highlight the potential of RPL35 as a therapeutic target, particularly in cancers where its non-canonical functions contribute significantly to disease progression.
Despite growing understanding of RPL35's functions, several key aspects remain to be fully elucidated:
Tissue-specific functions:
The mechanisms underlying tissue-specific expression patterns of RPL35
Why mutations in RPL35 affect some tissues (like erythroid progenitors) more than others
The role of RPL35 in tissue development and differentiation
Regulatory mechanisms:
How RPL35 expression and activity are regulated under normal and stress conditions
The role of post-translational modifications in modulating RPL35 function
Factors that control RPL35's incorporation into ribosomes versus engagement in non-canonical functions
RNA specificity:
The full spectrum of RNAs that interact with RPL35
Structural features that determine RPL35's binding specificity
How RPL35 selectively influences translation of specific mRNAs
Cancer heterogeneity:
Why RPL35 expression patterns vary across cancer types and subtypes
The context-dependent effects of RPL35 dysregulation in different malignancies
How tumor microenvironment influences RPL35 function
Therapeutic targeting:
Strategies for selectively targeting RPL35's non-canonical functions
Approaches to overcome potential resistance mechanisms
Biomarkers predicting response to RPL35-targeted therapies
Addressing these knowledge gaps will require integrated approaches combining structural biology, functional genomics, systems biology, and translational research. Advances in these areas will enhance our understanding of RPL35's roles in normal physiology and disease pathogenesis.
Ribosomal Protein L35 (RPL35) is a crucial component of the large 60S subunit of ribosomes, which are the cellular organelles responsible for protein synthesis. This protein is encoded by the RPL35 gene and belongs to the L29P family of ribosomal proteins. It is located in the cytoplasm and plays a significant role in the translation process by facilitating the assembly and function of ribosomes .
The RPL35 gene is a protein-coding gene associated with several important biological pathways, including viral mRNA translation and nervous system development. The gene is located on chromosome 9 and has multiple processed pseudogenes dispersed throughout the genome . The protein itself is composed of approximately 80 structurally distinct proteins and 4 RNA species, forming a part of the large ribosomal subunit .
RPL35 is essential for the proper functioning of ribosomes. It is involved in the binding of RNA and mRNA, which are critical steps in the translation process. The protein’s role in ribosome biogenesis is vital, as it contributes to the processing of pre-rRNA and the assembly of ribosomal subunits . In vivo depletion of RPL35 results in a deficit of 60S ribosomal subunits and the appearance of half-mer polysomes, indicating its importance in ribosome synthesis .
Mutations or dysregulation of the RPL35 gene have been associated with certain diseases, such as Diamond-Blackfan Anemia (DBA), a rare genetic disorder characterized by failure of the bone marrow to produce red blood cells . Understanding the function and regulation of RPL35 can provide insights into the mechanisms underlying such diseases and potentially lead to the development of targeted therapies.
Recombinant RPL35 is used in various research applications to study ribosome function and protein synthesis. By expressing and purifying human recombinant RPL35, researchers can investigate its interactions with other ribosomal proteins and RNA, as well as its role in the translation process. This knowledge can contribute to the development of new therapeutic strategies for diseases related to ribosomal dysfunction.