ORIGINAL RESEARCH article
Front. Immunol.
Sec. Cancer Immunity and Immunotherapy
Volume 16 - 2025 | doi: 10.3389/fimmu.2025.1602898
This article is part of the Research TopicIntegrating Molecular Mechanisms, Immunotherapy, and Drug Sensitivity in Cancer Immunology and OncologyView all 28 articles
Multi-omics analysis reveals the role of ribosome biogenesis in malignant clear cell renal cell carcinoma and the development of a machine learning-based prognostic model
Provisionally accepted- 1Affiliated Meizhou Hospital of Shantou University Medical College, Meizhou, China
- 2Department of Urology, Meizhou People's Hospital (Meizhou Academy of Medical Sciences), Meizhou, Guangdong Province, China
Select one of your emails
You have multiple emails registered with Frontiers:
Notify me on publication
Please enter your email address:
If you already have an account, please login
You don't have a Frontiers account ? You can register here
Background: Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cancer, marked by high molecular heterogeneity and limited responsiveness to targeted or immune therapies.Ribosome biogenesis (Ribosis), a central regulator of cell growth and metabolism, has emerged as a driver of tumor aggressiveness. However, its role in ccRCC pathogenesis and prognosis remains poorly defined.Methods: We integrated bulk RNA sequencing, single-cell RNA sequencing, and spatial transcriptomics sequencing data to dissect the biological functions and clinical relevance of Ribosisrelated genes in ccRCC. Through pseudotime trajectory analysis and metabolic flux inference, we examined malignant progression and metabolic reprogramming. A prognostic model based on a Ribosis-related signature (RBRS) was built using 118 machine learning algorithm combinations and validated in internal and external cohorts. A web-based calculator was also developed. We further analyzed immune infiltration, genomic alterations, tumor microenvironment features, and drug sensitivity. Expression of five core Ribosis-related genes (RPL38, RPS2, RPS14, RPS19, RPS28) was validated by qRT-PCR.We identified a Ribosis-high malignant subpopulation with enhanced stemness, poor prognosis, and elevated oxidative phosphorylation. These cells showed increased metabolic activity, especially in the pyruvate-lactate axis, potentially facilitating immune evasion. The RBRS model outperformed 32 published signatures (C-index = 0.68). High-risk patients exhibited an "immuneactivated yet immunosuppressed" microenvironment, with increased CD8⁺ T-cell infiltration and elevated regulatory T cells, myeloid-derived suppressor cells, and immune checkpoint expression (e.g., PDCD1, CTLA-4). Despite active antigen presentation and immune cell recruitment, terminal tumor-killing capacity was impaired. High-risk tumors also showed higher mutation burden, frequent copy number loss of tumor suppressor genes, and resistance to common targeted therapies. The five RBRS genes were significantly upregulated in tumor tissues, consistent with bulk RNA-seq data.We reveal Ribosis as a key driver of ccRCC progression. The RBRS model demonstrates robust prognostic value and translational utility, linking Ribosis to metabolism, immune dysfunction, and therapy resistance, offering new insights for risk stratification and precision treatment in ccRCC.
Keywords: Ribosome biogenesis, Clear cell renal cell carcinoma, Multi-omics analysis, Malignant cells, machine learning, Prognostic model
Received: 30 Mar 2025; Accepted: 13 Jun 2025.
Copyright: © 2025 Xie, Peng, Wang, Huang, Zhou, Zhang, Jiang, Zhong, Feng and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
* Correspondence: Nanhui Chen, Affiliated Meizhou Hospital of Shantou University Medical College, Meizhou, China
Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.