RPLP2 Human, encoded by the RPLP2 gene, is a ribosomal phosphoprotein critical for the elongation phase of eukaryotic protein synthesis. As a component of the 60S ribosomal subunit, it interacts with RPLP1 and RPLP0 to form a pentameric complex that stabilizes the ribosome’s lateral stalk, enhancing translation efficiency . Its acidic nature distinguishes it from most ribosomal proteins, which are typically basic .
RPLP2 facilitates ribosomal translocation during elongation, enabling tRNA movement and peptide bond formation. Its interaction with RPLP1 and RPLP0 forms a dynamic complex that modulates ribosomal flexibility .
RPLP2 is upregulated in hepatocellular carcinoma (HCC) and correlates with poor prognosis . Key mechanisms include:
Autocrine TLR4 activation: Secreted RPLP2 binds TLR4 on HCC cells, triggering PI3K/AKT signaling .
HIF-1α-mediated glycolysis: RPLP2 promotes nuclear translocation of HIF-1α, upregulating glycolytic enzymes (e.g., GLUT1, LDHA) .
Therapeutic target: Silencing RPLP2 reduces lactate production and tumor growth in HCC models .
Parameter | Effect of RPLP2 Knockdown | Source |
---|---|---|
Lactate Production | 50–70% reduction | |
Cell Proliferation | 40–60% inhibition (CCK-8 assay) | |
HIF-1α Nuclear Entry | Significant attenuation |
RPLP2 is an essential host factor for flaviviruses (e.g., dengue, Zika, yellow fever) and hepaciviruses (e.g., HCV). Key roles include:
Viral protein synthesis: RPLP2 depletion reduces early viral protein accumulation .
Mosquito vector dependency: Aedes aegypti requires RPLP2 for DENV replication .
Translation specificity: Minimal impact on global cellular translation but critical for viral RNA translation .
Virus | RPLP2 Dependency | Experimental Model | Source |
---|---|---|---|
DENV-2 | Essential | HuH-7 cells, Aedes mosquitoes | |
HCV | Moderate | HuH-7.5 cells | |
ZIKV | Essential | A549 cells |
RPLP2 inhibition is being explored for:
Detection of RPLP2 expression in human tissue samples typically employs multiple complementary techniques to ensure reliable results. Western blotting using antibodies specific to RPLP2 serves as a primary method for protein-level detection, while quantitative real-time PCR (qRT-PCR) is commonly used to assess RPLP2 mRNA expression. Immunohistochemistry (IHC) provides valuable information about RPLP2 localization within tissues and cells. For higher sensitivity analyses, researchers commonly use mass spectrometry-based proteomics approaches, which can detect RPLP2 in complex protein mixtures. When conducting these experiments, it is essential to include appropriate positive controls (such as recombinant RPLP2 protein) and negative controls to validate results. The recombinant human RPLP2 protein can be expressed in systems like Escherichia coli and purified using his-tag affinity chromatography for use as standards in these assays .
RPLP2 belongs to the P protein family of ribosomal proteins and has distinct structural and functional characteristics that differentiate it from other ribosomal proteins. Structurally, RPLP2 contains the Large ribosomal subunit protein P1/P2 domain and the Large ribosomal subunit protein P2 domain specific to eukaryotes, with a total length of 115 amino acids in humans . Unlike many core ribosomal proteins that are essential for basic ribosome assembly, RPLP2 localizes to the ribosomal stalk and plays a specialized role in translation elongation rather than initial ribosome assembly. RPLP2 functions by binding to elongation factors, particularly eEF2, to enhance translation efficiency. This interaction is critical for relieving ribosome pausing during translation, especially within the first ~100 codons of messenger RNAs. When RPLP2 is depleted, ribosomes accumulate at the 5' end of mRNAs, indicating its importance in translation elongation rather than initiation or termination .
For studying RPLP2 knockdown effects in cell culture models, a multi-faceted approach is recommended to obtain comprehensive and reliable results. RNA interference using siRNAs targeting RPLP2 is a widely used method, as demonstrated in studies where RPLP1/2 knockdown effects were analyzed 48 hours post-transfection . CRISPR-Cas9 genome editing provides an alternative for generating stable RPLP2 knockout cell lines for long-term studies. After knockdown, verification of reduced RPLP2 expression should be performed at both mRNA (qRT-PCR) and protein (western blot) levels. Functional assays should include pulse-chase experiments with [35S]-methionine/cysteine labeling to assess protein synthesis and turnover rates, as this method has revealed that RPLP2 knockdown increases both synthesis and degradation of certain proteins . Additionally, ribosome profiling (RIBOseq) should be conducted to analyze ribosome distribution on mRNAs, as RPLP2 depletion causes characteristic shifts in ribosome positioning toward the 5' end of open reading frames, indicating elongation defects .
RPLP2 plays a sophisticated role in promoting aerobic glycolysis in hepatocellular carcinoma (HCC) through a multistep signaling cascade. Research has demonstrated that RPLP2 is significantly upregulated in HCC tissues, and this overexpression correlates with poor patient prognosis and survival. Mechanistically, RPLP2 activates Toll-like receptor 4 (TLR4) on the surface of HCC cells through autocrine signaling, initiating a downstream cascade involving the PI3K/AKT pathway. This activation facilitates the translocation of hypoxia-inducible factor-1α (HIF-1α) into the nucleus, where it functions as a transcription factor regulating glycolytic enzymes. The enhanced HIF-1α activity directly increases the expression of key glycolytic enzymes and stimulates lactate production, hallmarks of the Warburg effect in cancer cells. This metabolic reprogramming toward aerobic glycolysis provides HCC cells with enhanced proliferation capabilities even under normoxic conditions. Experimental validation has confirmed that RPLP2 depletion reduces glycolytic enzyme expression, decreases lactate production, and inhibits HCC cell proliferation, highlighting the essential role of RPLP2 in metabolic adaptation during HCC progression .
Resolving discrepancies in RPLP2 interaction data across experimental systems requires implementing a comprehensive strategy that integrates multiple orthogonal techniques while carefully controlling experimental variables. Initially, researchers should perform systematic comparison of in vitro versus in vivo interaction studies, as RPLP2's function in translation elongation was initially identified in in vitro experiments but required high-sensitivity ribosome profiling (RIBOseq) to confirm in cellular contexts . Cross-validation should employ multiple protein-protein interaction detection methods, including co-immunoprecipitation, proximity ligation assays, and label-transfer approaches. For RNA-protein interactions, techniques such as RNA immunoprecipitation (RIP), crosslinking immunoprecipitation (CLIP), and RNA Antisense Purification (RAP) should be used complementarily. Contradictory findings often result from different cellular contexts, so researchers should directly compare RPLP2 interactions across multiple cell types and under various physiological conditions (e.g., normal growth, stress, disease states). Advanced quantitative proteomics with stable isotope labeling (SILAC) combined with mass spectrometry can provide quantitative measures of interaction strength across experimental conditions, helping to identify condition-specific interactions that might explain discrepancies .
Distinguishing between RPLP2's canonical translation role and its non-canonical functions in disease states requires sophisticated experimental design that separates these potentially overlapping activities. Researchers should employ domain mapping and mutagenesis studies to create RPLP2 variants that selectively disrupt specific functions while preserving others. This approach can identify critical residues responsible for canonical versus non-canonical activities. Temporal analysis using inducible expression systems can help separate immediate translation effects from secondary signaling effects of RPLP2. In disease contexts like hepatocellular carcinoma, specialized assays that separately quantify RPLP2's impact on translation elongation (using ribosome profiling) versus its effect on TLR4 signaling and glycolysis (using pathway-specific reporters) will help delineate these functions. Subcellular fractionation studies are crucial, as RPLP2's canonical role occurs primarily at the ribosome, while its reported activation of TLR4 suggests extracellular or membrane-associated functions that represent non-canonical activities . Additionally, comparative studies between normal cells and disease models using techniques like proximity-dependent biotin identification (BioID) can reveal disease-specific interaction partners that mediate non-canonical functions of RPLP2.
Studying RPLP2's impact on ribosome pausing and nascent protein folding requires specialized techniques that capture translation dynamics and protein maturation in real-time. Ribosome profiling (RIBOseq) with sub-codon resolution is essential for detecting site-specific ribosome pausing, as it can identify precise locations where ribosomes accumulate on mRNAs. This technique has already revealed that RPLP1/2 depletion causes ribosome accumulation within the first ~100 codons of open reading frames, suggesting elongation defects . To connect translation dynamics with protein folding outcomes, pulse-chase experiments combined with immunoprecipitation provide critical insights, as demonstrated by studies showing increased protein synthesis but faster degradation of viral proteins in RPLP2-depleted cells . Researchers should also implement nascent chain folding assays, such as limited proteolysis coupled with mass spectrometry, to assess how RPLP2 affects co-translational folding. For quantitative assessment of ribosome distribution shifts, researchers should calculate polarity scores for mRNAs, which have revealed that RPLP2 depletion causes a significant shift of ribosomes toward the 5' end of transcripts (mean difference = 0.013, P = 2.2 × 10−16) . Additionally, structural studies using cryo-electron microscopy of ribosomes with and without RPLP2 can provide molecular insights into how this protein facilitates elongation and prevents ribosome pausing.
Investigating RPLP2's role in ribosome biogenesis feedback mechanisms requires a systematic approach that captures both immediate responses and longer-term adaptations to RPLP2 depletion. Researchers should implement multi-omics studies that integrate RIBOseq, RNA-seq, and proteomics data to comprehensively assess how RPLP2 knockdown affects ribosome component synthesis. Previous studies have shown that RPLP2 depletion leads to increased ribosome density specifically on cytosolic ribosomal protein mRNAs but not mitochondrial ribosomal protein mRNAs, suggesting a targeted feedback response . Time-course experiments following RPLP2 depletion are crucial for distinguishing primary effects from compensatory responses, with special attention to early changes in eEF2K levels, which have been observed to decrease significantly in response to RPLP2 knockdown . Researchers should also investigate transcription factor activation, particularly those known to regulate ribosomal protein genes, using techniques like ChIP-seq and reporter assays. Polysome profiling combined with specific mRNA detection can reveal translation efficiency changes for ribosomal proteins and ribosome assembly factors. For functional validation, rescue experiments introducing wild-type versus mutant RPLP2 forms can confirm specific aspects of feedback regulation. These approaches will help elucidate the molecular mechanisms by which cells sense RPLP2 deficiency and trigger compensatory ribosome biogenesis.
When studying RPLP2 in disease models, a comprehensive set of control experiments is essential to ensure valid and interpretable results. Researchers must include genetic controls with both gain-of-function (overexpression of wild-type RPLP2) and loss-of-function (knockdown/knockout of RPLP2) approaches in parallel. Rescue experiments reintroducing wild-type or mutant RPLP2 into depleted cells are critical for confirming phenotype specificity. Since RPLP2 functions with RPLP1 as a complex, controls should include individual and combined knockdowns to distinguish between specific RPLP2 functions and general stalk protein effects. Temporal controls involving inducible expression systems help separate immediate from secondary effects of RPLP2 manipulation. When studying RPLP2's role in diseases like HCC, matched tissue pairs (tumor and adjacent normal tissue) from the same patients should be analyzed to control for individual variations. For functional studies like those examining RPLP2's effect on glycolysis in HCC, standard metabolic controls should include alternative metabolic pathway assessments to confirm specificity for glycolysis rather than general metabolic alterations . Additional controls should assess potential off-target effects of genetic manipulation techniques by monitoring global translation rates and cell viability markers.
When faced with contradictory findings regarding RPLP2's interactions with signaling pathways, researchers should implement a systematic approach to reconcile discrepancies. First, a comprehensive literature review should be conducted to identify methodological differences that might explain contradictions, including cell types, experimental conditions, and detection methods used across studies. Context-dependent effects should be carefully considered, as RPLP2's interactions may vary substantially between normal and disease states, different tissues, or under various stress conditions. For instance, RPLP2's activation of TLR4 signaling has been observed specifically in hepatocellular carcinoma cells, which may not occur in normal hepatocytes . Researchers should employ orthogonal methods to verify key interactions, combining biochemical approaches (co-immunoprecipitation, proximity ligation) with functional assays (reporter systems, downstream target activation). Quantitative aspects of interactions should be assessed using dose-response experiments and kinetic studies to determine whether apparent contradictions reflect threshold effects rather than absolute differences. When pathway crosstalk is suspected, inhibitor studies targeting specific nodes in the implicated pathways can help delineate direct versus indirect effects of RPLP2. Finally, genetic approaches creating RPLP2 mutants that selectively disrupt specific interaction interfaces can provide definitive evidence for direct versus coincidental associations.
Analyzing ribosome profiling data in RPLP2 studies requires sophisticated statistical approaches to accurately identify translation changes while controlling for technical variability. Researchers should implement generalized linear models, such as those used in the Riborex tool, which explicitly model the dependence of ribosome-protected fragment (RPF) abundance on RNA abundances, allowing for the identification of true translation efficiency changes rather than mRNA level differences. This approach has successfully identified significant changes in ribosome densities on just 10 genes (with 8 showing higher density) upon RPLP1/2 knockdown (q-value < 0.01, abs(log2 fold change) > 1) . For meta-gene analyses examining ribosome distribution patterns across all transcripts, researchers should calculate polarity scores that quantify 5' versus 3' ribosome distribution biases. In RPLP2 depletion studies, a small but highly significant shift toward the 5' end of open reading frames has been observed (mean difference = 0.013, P = 2.2 × 10−16) . Differential translation efficiency analysis should include appropriate multiple testing corrections (e.g., Benjamini-Hochberg procedure) to control false discovery rates. For codon-specific pause site identification, researchers should apply specialized statistical models that account for local sequence context and normalize to baseline ribosome occupancy. Finally, pathway enrichment analyses should be conducted on genes with altered translation patterns to identify biological processes most affected by RPLP2 manipulation.
RPLP2's critical role in translation elongation presents several promising therapeutic avenues for cancer treatment, particularly for hepatocellular carcinoma where RPLP2 is significantly upregulated and associated with poor prognosis . Targeting strategies should focus on disrupting the specific functions of RPLP2 that cancer cells depend on while minimizing impact on normal cells. Small molecule inhibitors designed to interfere with RPLP2's interaction with elongation factors like eEF2 could selectively impair the enhanced translation rates that cancer cells require. Alternatively, compounds that disrupt RPLP2's activation of TLR4 and subsequent PI3K/AKT/HIF-1α signaling could specifically target its role in promoting aerobic glycolysis in HCC. For increased specificity, researchers could develop bifunctional degraders (PROTACs) targeting RPLP2 for proteasomal degradation specifically in cancer cells by incorporating tumor-specific ligands. RNA-based therapeutics, including antisense oligonucleotides or siRNAs targeting RPLP2, offer another approach, though delivery specifically to cancer cells remains challenging. Combination therapies targeting both RPLP2 and downstream effectors like HIF-1α could provide synergistic effects by simultaneously disrupting both translation elongation and metabolic reprogramming. Importantly, preclinical studies should carefully evaluate potential toxicity, as complete inhibition of RPLP2 might impair normal cellular translation, whereas partial inhibition may be sufficient to selectively affect cancer cells with their higher demands on protein synthesis machinery .
Studying RPLP2's interactions within the mRNA-bound proteome requires cutting-edge methodologies that capture dynamic protein-RNA-protein complexes with high specificity and temporal resolution. Researchers should implement proximity-dependent approaches such as RNA-BioID or RNA-APEX, which involve fusing biotin ligases to RPLP2 to biotinylate nearby proteins when associated with specific mRNAs. These methods can reveal the context-specific interaction partners of RPLP2 during different phases of translation. Time-resolved crosslinking mass spectrometry (TX-MS) can capture dynamic changes in RPLP2's interaction network during translation elongation with millisecond-scale temporal resolution. For visualizing RPLP2's associations with the translational machinery in living cells, researchers should employ techniques like live-cell single-molecule imaging combined with fluorescence resonance energy transfer (FRET) to observe real-time interactions between RPLP2 and both the ribosome and elongation factors. Cryo-electron tomography of intact polysomes can provide structural insights into how RPLP2 influences ribosome conformation during different stages of elongation. For comprehensive characterization of the mRNA-bound proteome, researchers can adapt RNA antisense purification coupled with mass spectrometry (RAP-MS) to identify all proteins associated with specific mRNAs in the presence or absence of RPLP2, revealing how RPLP2 influences the composition of ribonucleoprotein complexes .
While RPLP2 is primarily known for its role in translation elongation, emerging evidence suggests broader functions in cellular stress response mechanisms that extend beyond its canonical role in protein synthesis. Researchers should investigate RPLP2's potential involvement in the integrated stress response (ISR) by examining its interactions with stress granule components and eIF2α phosphorylation pathways. The observation that RPLP2 depletion affects protein degradation rates suggests a possible connection to proteostasis networks, including the unfolded protein response (UPR) and endoplasmic reticulum-associated degradation (ERAD) pathways. RPLP2's activation of TLR4 signaling in hepatocellular carcinoma indicates potential functions in immune and inflammatory responses that warrant exploration in various stress conditions . Since RPLP2 knockdown affects eEF2K levels, its role in energy stress sensing may be significant, as eEF2K is a key regulator that suppresses translation in response to energy limitation . Systematic studies should evaluate how RPLP2 expression and localization change under various stress conditions (oxidative stress, nutrient deprivation, hypoxia, etc.) and determine whether it shuttles between different cellular compartments during stress. Interactome analyses under normal versus stress conditions could reveal stress-specific binding partners of RPLP2. Finally, researchers should investigate whether RPLP2 undergoes post-translational modifications in response to cellular stress, potentially altering its function from promoting translation to activating stress-specific pathways.
Studying RPLP2 in primary human tissues presents multiple challenges requiring specialized approaches to obtain reliable results. For tissue preservation and preparation, researchers should optimize protocols that maintain protein-RNA interactions, such as using RNase inhibitors during tissue processing and flash-freezing samples immediately after collection. Since primary tissues have limited availability and heterogeneous cell populations, researchers should implement laser capture microdissection to isolate specific cell types of interest, enabling cell type-specific analysis of RPLP2 function. Single-cell approaches, including single-cell RNA-seq and mass cytometry with RPLP2-specific antibodies, can characterize RPLP2 expression heterogeneity across different cell populations within complex tissues. For functional studies in primary tissues, ex vivo tissue slice cultures can maintain tissue architecture while allowing experimental manipulation, though optimization for each tissue type is required. Since direct genetic manipulation of primary tissues is challenging, researchers can employ adenoviral or lentiviral vectors for efficient gene delivery to primary cells. Additionally, organoid models derived from primary human tissues provide systems that better reflect in vivo conditions compared to cell lines while allowing genetic manipulation and long-term culture. Patient-derived xenograft models enable in vivo studies of RPLP2 function in human tissues within a physiologically relevant environment. Finally, for validation of findings from model systems, researchers should implement spatial transcriptomics and proteomics approaches to map RPLP2 expression patterns in intact tissue sections, correlating with histopathological features.
Distinguishing between direct and indirect effects of RPLP2 on cellular metabolic pathways requires a multi-faceted experimental strategy that isolates specific mechanisms from secondary consequences. Researchers should implement acute versus chronic manipulation studies, using inducible or rapidly acting systems (such as degron-tagged RPLP2 variants) to identify immediate metabolic changes following RPLP2 depletion or overexpression, which are more likely to represent direct effects. Temporal profiling of metabolic changes following RPLP2 manipulation can establish cause-effect relationships and separate primary from secondary adaptations. To determine whether RPLP2's effects on metabolism require its translation function, researchers should create separation-of-function mutants that selectively disrupt different RPLP2 activities and assess their impact on metabolic pathways. Direct binding studies using techniques like chromatin immunoprecipitation (ChIP) or RNA immunoprecipitation (RIP) can identify whether RPLP2 directly interacts with factors controlling metabolic gene expression. When studying RPLP2's reported role in activating TLR4 signaling and promoting glycolysis in HCC, researchers should implement pathway dissection approaches, selectively inhibiting intermediate steps (e.g., PI3K or AKT) to determine whether RPLP2's metabolic effects require these signaling components . Metabolic flux analysis using isotope-labeled metabolites provides crucial evidence for direct metabolic regulation by tracking specific pathway activities rather than steady-state metabolite levels. Finally, comparative studies across different cell types with varying metabolic dependencies can reveal context-specific versus universal effects of RPLP2 on metabolism.
Ribosomal Phosphoprotein P2 (RPLP2) is a crucial component of the ribosome, specifically part of the large 60S subunit. This protein plays a significant role in the elongation step of protein synthesis, which is essential for cellular function and growth. The human recombinant form of RPLP2 is produced using recombinant DNA technology, typically in an E. coli expression system .
RPLP2 is a single, non-glycosylated polypeptide chain consisting of 139 amino acids, with a molecular mass of approximately 14.2 kDa . It belongs to the L12P family of ribosomal proteins and is functionally equivalent to the E. coli L7/L12 ribosomal protein . Unlike most ribosomal proteins, which are basic, RPLP2 is acidic. Its C-terminal end is nearly identical to the C-terminal ends of the ribosomal phosphoproteins P0 and P1 .
RPLP2 can interact with P0 and P1 to form a pentameric complex consisting of P1 and P2 dimers and a P0 monomer . This complex is vital for the ribosome’s function in protein synthesis.
The human recombinant RPLP2 is typically produced in E. coli. The process involves cloning the gene encoding RPLP2 into an expression vector, which is then introduced into E. coli cells. These cells are cultured under conditions that induce the expression of the recombinant protein. The protein is then purified using chromatographic techniques to achieve a high level of purity .
The recombinant RPLP2 produced in E. coli is fused to a 24 amino acid His-tag at the N-terminus, which facilitates its purification . The final product is a sterile, filtered, colorless solution containing the RPLP2 protein in a buffer solution .
RPLP2 is involved in the elongation step of protein synthesis, where it plays a role in the interaction between the ribosome and elongation factors. This interaction is crucial for the accurate and efficient synthesis of proteins. The acidic nature of RPLP2 and its ability to form complexes with other ribosomal proteins are essential for its function .
The analysis of RPLP2 typically involves techniques such as SDS-PAGE to determine its purity and molecular weight . Additionally, its interaction with other ribosomal proteins can be studied using various biochemical and biophysical methods.