RPS27A, or Ribosomal Protein S27a, is a protein-coding gene that plays a crucial role in the structure and function of the ribosome, specifically as a component of the 40S subunit. It is synthesized as a fusion protein with ubiquitin at its N-terminus, which is later cleaved to produce the mature ribosomal protein and free ubiquitin . This article will delve into the functions, clinical significance, and research findings related to RPS27A in humans.
RPS27A is involved in ribosome biogenesis and post-translational modifications of proteins. Beyond its role in ribosomal function, RPS27A exhibits extra-ribosomal activities, including participation in cell cycle regulation and apoptosis inhibition . It is also implicated in the induction of the tumor suppressor p53 in response to defects in ribosome biogenesis .
Ribosome Biogenesis: Essential for the production and maturation of ribosomal subunits.
Post-Translational Modifications: Involved in modifying proteins post-translationally.
Cell Cycle Regulation: Plays a role in regulating cell cycle progression.
Apoptosis Inhibition: Helps inhibit apoptosis, promoting cell survival.
RPS27A has been associated with various diseases, including cancers and ribosomopathies. High expression levels of RPS27A are linked to poor prognosis in several cancer types, such as cervical, colorectal, and leukemia . It serves as a prognostic biomarker for HPV16 cervical cancer and is involved in promoting proliferation and inhibiting apoptosis in cancer cells .
Cancer Prognosis: High expression is associated with poor prognosis in cervical, colorectal, and leukemia.
Ribosomopathies: Linked to diseases resulting from ribosomal defects.
Cancer Cell Growth: Promotes proliferation and inhibits apoptosis in cancer cells.
Recent studies have highlighted the multifaceted roles of RPS27A. For instance, its expression is significantly enriched in pathways related to cytoplasmic translation, mRNA catabolic processes, and ribosomal RNA processing . Additionally, RPS27A is proposed to be important for the induction of p53 in response to ribosome biogenesis defects .
Pathway Enrichment: Enriched in pathways related to translation and RNA processing.
p53 Induction: Important for inducing p53 in response to ribosomal defects.
Cancer Biomarker: Identified as a prognostic biomarker for certain cancers.
Cancer Type | RPS27A Expression Level | Prognostic Value |
---|---|---|
Cervical | High | Poor Prognosis |
Colorectal | High | Poor Prognosis |
Leukemia | High | Poor Prognosis |
Pathway | Description |
---|---|
Cytoplasmic Translation | Involves translation processes within the cytoplasm. |
Nuclear-Transcriptional mRNA Catabolic | Processes related to mRNA degradation in the nucleus. |
Ribosomal RNA (rRNA) Processing | Essential for ribosome maturation and function. |
MQIFVKTLTG KTITLEVEPS DTIENVKAKI QDKEGIPPDQ QRLIFAGKQL EDGRTLSDYN IQKESTLHLV LRLRGG.
RPS27A is a fusion protein consisting of ubiquitin at the N-terminus and ribosomal protein S27a at the C-terminus. When expressed, the protein undergoes post-translational processing that generates free ubiquitin monomer and ribosomal protein S27a . RPS27A is a component of the 40S subunit of the ribosome and belongs to the S27AE family of ribosomal proteins . It contains C4-type zinc finger domains and is predominantly located in the cytoplasm .
The protein serves dual critical functions:
As part of the 40S ribosomal subunit, it participates in protein synthesis
Through its ubiquitin component, it contributes to the targeting of cellular proteins for degradation by the 26S proteasome
This unique structure positions RPS27A at the intersection of protein synthesis and degradation pathways, allowing it to influence multiple cellular processes.
For comprehensive RPS27A detection and analysis, researchers should consider the following methodological approaches:
Immunohistochemistry (IHC): Effective for visualizing RPS27A protein expression in tissue sections, as demonstrated in studies of human and mouse liver samples
Public database analysis: Multiple platforms can be utilized for in silico analysis:
Coculturing assays: Particularly valuable for studying RPS27A's effects on immune cell interactions, such as macrophage polarization
Recombinant protein approaches: Using control fragments (such as Human RPS27A aa 94-155) for validation experiments in immunohistochemistry, immunocytochemistry, and Western blotting
When designing experiments, researchers should note that RPS27A shows high sequence identity across species (98% with mouse and rat orthologs), which can facilitate translational research between model organisms and humans .
RPS27A shows significant dysregulation across multiple cancer types with important implications for cancer biology:
Research indicates that RPS27A expression levels correlate with clinicopathologic features and prognosis in human cancers . The protein participates in the regulation of genomic alterations and heterogeneity, showing associations with tumor mutation burden, microsatellite instability, and neoantigen formation .
Mechanistically, RPS27A overexpression in HCC cells has been experimentally demonstrated to enhance cellular proliferation, migration, and invasion capabilities, suggesting direct oncogenic functions .
RPS27A displays complex and context-dependent relationships with immune cell populations across different cancer types:
Immune cell associations: RPS27A expression shows:
Cancer-specific immune profiles:
In hepatocellular carcinoma (LIHC): RPS27A positively associates with markers of general T cells, CD8+ T cells, B cells, monocytes, tumor-associated macrophages (TAMs), neutrophils, dendritic cells, Th1 cells, and T cell exhaustion
In stomach adenocarcinoma (STAD): RPS27A negatively correlates with markers of TAMs, M2 macrophages, and regulatory T cells (Tregs)
Spatial relationships: In breast cancer, spatial transcriptomic data demonstrates that RPS27A expression regions overlap with macrophage marker CD68 and myeloid-derived suppressor cell (MDSC) marker CSF1R
This evidence collectively suggests that RPS27A plays a significant role in shaping the tumor immune microenvironment, with important implications for immunotherapy response.
Experimental evidence has established that RPS27A significantly impacts macrophage polarization, particularly in hepatocellular carcinoma:
M2 polarization: RPS27A overexpression in HCC cells accelerates M2 polarization of macrophages, as demonstrated through coculturing assays
Methodological approach: This phenomenon can be studied using:
The promotion of M2 macrophage polarization is particularly significant as M2 macrophages typically create an immunosuppressive environment that facilitates tumor progression. This mechanism may partially explain how RPS27A contributes to cancer development and potentially resistance to certain immunotherapies.
RPS27A shows considerable promise as both a prognostic biomarker and potential immunotherapy target:
Prognostic value: RPS27A expression correlates with clinicopathologic features and survival outcomes across multiple cancer types
Immunotherapy relevance: RPS27A expression is connected to:
Potential therapeutic applications:
Research validating RPS27A as an immunotherapy target would benefit from integrating genomic, transcriptomic, and proteomic approaches with functional validation in preclinical models and correlation with clinical outcomes in patient cohorts.
RPS27A affects multiple signaling cascades critical for cancer development and progression:
DNA repair pathways: RPS27A influences DNA repair mechanisms, potentially affecting genomic stability and therapy response
Invasion and metastasis signaling: Experimental evidence shows RPS27A promotes migration and invasion capabilities in cancer cells
Immune signaling networks:
These diverse effects on signaling networks position RPS27A as a multifunctional regulator in cancer biology, suggesting that targeting RPS27A might simultaneously affect multiple cancer hallmarks.
RPS27A participates in the regulation of genomic alterations with important implications for cancer heterogeneity:
Tumor mutation burden (TMB): RPS27A expression correlates with TMB, which is a key determinant of immunotherapy response
Microsatellite instability (MSI): RPS27A shows associations with MSI status across cancer types
Neoantigen formation: RPS27A affects neoantigen profiles, which influence immune recognition of tumor cells
Understanding these genomic relationships requires integrative approaches combining next-generation sequencing technologies with functional validation studies. The associations with TMB, MSI, and neoantigens are particularly significant given the importance of these features in predicting response to immune checkpoint blockade therapies.
When designing experiments to investigate RPS27A functions, researchers should consider:
Expression analysis approaches:
Functional validation methods:
Spatial analysis techniques:
These methodological considerations ensure comprehensive characterization of RPS27A's complex functions in cancer and immune regulation.
For functional investigations of RPS27A, researchers can employ several approaches:
Overexpression systems:
Knockdown/knockout strategies:
siRNA or shRNA for transient or stable knockdown
CRISPR-Cas9 for complete knockout
Degradation-based approaches (PROTACs, etc.) for protein-level depletion
Domain-specific manipulations:
These approaches allow researchers to dissect the distinct functions of RPS27A and its contributions to cancer-related processes.
When utilizing recombinant RPS27A proteins for experimental validation:
Protein fragment selection: Consider using defined fragments like RPS27A (aa 94-155) that represent functional domains
Blocking experiments: For antibody validation, use a 100x molar excess of the protein fragment control based on concentration and molecular weight, pre-incubating the antibody-protein control fragment mixture for 30 minutes at room temperature
Cross-species considerations: Take advantage of the high sequence identity between human RPS27A and orthologs from model organisms (98% with mouse and rat), which facilitates translational research
Application-specific optimization: Adjust protocols for specific applications such as IHC/ICC and Western blotting to ensure optimal results
Proper use of recombinant proteins can significantly enhance the reliability and interpretability of experimental results in RPS27A research.
Based on current evidence, several translational research directions for RPS27A show particular promise:
Biomarker development:
Therapeutic targeting:
Mechanistic investigations:
These research directions reflect the multifaceted roles of RPS27A in cancer biology and immunology, suggesting diverse applications in cancer management.
RPS27A's connections to immune regulation suggest several potential applications in combination immunotherapy:
Macrophage-directed combinations: Given RPS27A's role in M2 macrophage polarization, combining RPS27A targeting with macrophage-reprogramming strategies (e.g., CSF1R inhibitors) may enhance anti-tumor immunity
T cell checkpoint combinations: RPS27A's associations with T cell exhaustion markers suggest potential synergy with PD-1/PD-L1 or CTLA-4 blockade
Biomarker-guided approaches: Using RPS27A expression to stratify patients for specific immunotherapy combinations based on predicted immune microenvironment characteristics
Research in this area would benefit from preclinical models evaluating combination efficacy and mechanisms, followed by carefully designed clinical trials with appropriate biomarker analyses.
Therapeutic targeting of RPS27A presents several significant challenges that researchers must address:
Dual functionality concerns: As RPS27A functions in both protein synthesis (ribosomal component) and protein degradation (ubiquitin source), complete inhibition may disrupt essential cellular processes
Specificity requirements: Strategies must selectively target cancer-specific functions of RPS27A while sparing normal cellular functions
Delivery challenges: Approaches must effectively deliver therapeutics to tumor cells while minimizing exposure in normal tissues with high protein synthesis requirements
Resistance mechanisms: Cancer cells may develop compensatory mechanisms to overcome RPS27A targeting, requiring combination approaches
Addressing these challenges will require sophisticated drug development strategies, potentially including targeted delivery systems, selective modulators of specific RPS27A functions, and carefully designed combination approaches to manage potential resistance.
Ubiquitin is a globular protein with a compact structure. It contains multiple lysine residues and two C-terminal glycines, which are crucial for its function. The primary role of ubiquitin is to tag proteins for degradation via the ubiquitin-proteasome system. This process involves the attachment of ubiquitin molecules to substrate proteins, marking them for degradation by the proteasome. This tagging process is known as ubiquitination and is mediated by a series of enzymes: ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3) .
In humans, ubiquitin is encoded by four different genes located on chromosomes 17, 2, 19, and 12. These genes produce ubiquitin either as a single protein or as part of a larger precursor protein that is subsequently processed to release free ubiquitin . The high degree of conservation in ubiquitin’s amino acid sequence across different species underscores its fundamental importance in cellular biology.
Recombinant human ubiquitin is produced using genetic engineering techniques, typically in bacterial systems such as Escherichia coli. The recombinant protein is identical to the naturally occurring human ubiquitin in terms of its amino acid sequence and functional properties. It is widely used in research to study protein degradation, signal transduction, and other cellular processes involving ubiquitin .
Recombinant ubiquitin is a valuable tool in biochemical and cell biology research. It is used to investigate the mechanisms of ubiquitination and deubiquitination, the roles of ubiquitin in various signaling pathways, and the regulation of protein turnover. Additionally, recombinant ubiquitin can be conjugated to substrate proteins in vitro to study the effects of ubiquitination on protein function and stability .