1
|
Chong ZX, Ho WY, Yeap SK. Deciphering the roles of non-coding RNAs in liposarcoma development: Challenges and opportunities for translational therapeutic advances. Noncoding RNA Res 2025; 11:73-90. [PMID: 39736850 PMCID: PMC11683247 DOI: 10.1016/j.ncrna.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 01/01/2025] Open
Abstract
Liposarcoma is one of the most prevalent forms of soft tissue sarcoma, and its prognosis is highly dependent on its molecular subtypes. Non-coding RNAs (ncRNAs) like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) can bind various cellular targets to regulate carcinogenesis. By affecting the expressions and activities of their downstream targets post-transcriptionally, dysregulations of miRNAs can alter different oncogenic signalling pathways, mediating liposarcoma progression. On the contrary, lncRNAs can sponge miRNAs to spare their downstream targets from translational repression, indirectly affecting miRNA-regulated oncogenic activities. In the past 15 years, multiple fundamental and clinical research has shown that different ncRNAs play essential roles in modulating liposarcoma development. Yet, there is a lack of an effective review report that could summarize the findings from various studies. To narrow this literature gap, this review article aimed to compare the findings from different studies on the tumour-regulatory roles of ncRNAs in liposarcoma and to understand how ncRNAs control liposarcoma progression mechanistically. Additionally, the reported findings were critically reviewed to evaluate the translational potentials of various ncRNAs in clinical applications, including employing these ncRNAs as diagnostic and prognostic biomarkers or as therapeutic targets in the management of liposarcoma. Overall, over 15 ncRNAs were reported to play essential roles in modulating different cellular pathways, including apoptosis, WNT/β-catenin, TGF-β/SMAD4, EMT, interleukin, and YAP-associated pathways to influence liposarcoma development. 28 ncRNAs were reported to be upregulated in liposarcoma tissues or circulation, whereas 11 were downregulated, making them potential candidates as liposarcoma diagnostic biomarkers. Among these ncRNAs, measuring the tissues or circulating levels of miR-155 and miR-195 was reported to help detect liposarcoma, differentiate liposarcoma subtypes, and predict the survival and treatment response of liposarcoma patients. Overall, except for a few ncRNAs like miR-155 and miR-195, current evidence to support the use of discussed ncRNAs as biomarkers and therapeutic targets in managing liposarcoma is mainly based on a single-center study with relatively small sample sizes or cell-based studies. Hence, more large-scale multi-center studies should be conducted to further confirm the sensitivity, specificity, and safety of ncRNAs as biomarkers and therapeutic targets. Instead of furthering investigation to confirm the translational values of all the discussed ncRNAs, which can be time- and cost-consuming, it would be more practical to focus on a few ncRNAs, including miR-155 and miR-195, to evaluate if they are sensitive and safe to be used as liposarcoma biomarkers and therapeutic agents or targets.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, 117599, Singapore
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900, Sepang, Selangor, Malaysia
| |
Collapse
|
2
|
Li X, Han Z, Li H. Hif3α Plays Key Roles in the Progression of Alzheimer's Disease Caused by Circadian Rhythm Disruption through Regulating the m 6A/KDM3A/TGF-β1 Axis. BIOLOGY 2024; 13:412. [PMID: 38927292 PMCID: PMC11201003 DOI: 10.3390/biology13060412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Disrupted circadian rhythms are associated with the onset of chronic diseases and impairments, including cancer, diabetes, and hypertension. However, whether circadian disruptions accelerate the progression of Alzheimer's disease and the respective pathway remains unclear. In this study, we constructed animal models using male C57BL/6N and APP/PS1 mice. Irregular illumination during sleeping hours was administered to the mice in our intervention groups to consistently disrupt their circadian rhythms. The impact of the intervention was evaluated through body weight tracking, cerebral index determination, histopathological staining, and biochemical marker analysis. Transcriptomic sequencing identified critical genes, with the data subsequently validated using RNA m6A detection and site analysis. The evaluations revealed that circadian disruptions impaired normal weight gain, liver and kidney functions, neuronal cells, and overall brain function. Transcriptomic sequencing data revealed a trend of elevating expression of Hif3α mRNA in the intervention groups. Further analysis of specific gene sites revealed that m6A methylation of the Hif3α gene at m6A site 3632 primarily drove the observed variations in HIF3A protein expression in our model. Furthermore, the expression of proteins in PC12 cells, N2a cells, and mice brains validated that an increase in HIF3A expression decreased KDM3A and TGF-β1 protein expression. Our study reveals a hitherto unknown pathway through which the disruption of circadian rhythms, by triggering m6A methylation at m6A site 3632 in the Hif3α gene, leads to the initiation and acceleration of AD. These findings provide valuable insights and guidelines for treating AD patients and enhancing caregiving by professionals.
Collapse
Affiliation(s)
- Xinrui Li
- Beijing National Day School, Beijing 100062, China;
| | - Zhengkun Han
- Beijing Key Laboratory of Food Processing and Safety in Forest, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China;
| | - Huiying Li
- Beijing Key Laboratory of Food Processing and Safety in Forest, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China;
| |
Collapse
|
3
|
Xu Z, Zhou H, Luo Y, Li N, Chen S. Bioinformatics analysis and validation of CSRNP1 as a key prognostic gene in non-small cell lung cancer. Heliyon 2024; 10:e28412. [PMID: 38560128 PMCID: PMC10979096 DOI: 10.1016/j.heliyon.2024.e28412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Cysteine and serine-rich nuclear protein 1 (CSRNP1) has shown prognostic significance in various cancers, but its role in non-small cell lung cancer (NSCLC) remains elusive. We investigated CSRNP1 expression in NSCLC cases using bioinformatics tools from the GEO public repository and validated our findings through RT-qPCR in tumor and adjacent normal tissues. KEGG and GO enrichment analyses were employed to unveil the significant deregulation in signaling pathways. Additionally, clinical significance of CSRNP1 in NSCLC was determined through receiver operating curve (ROC) analysis, and its impact on survival was assessed using Kaplan-Meier analysis. To explore the functional impact of CSRNP1, we silenced its expression in NSCLC cells and assessed the effects on cell viability, migration, and invasion using MTT, Transwell, and wound-healing assays, respectively. Additionally, we investigated the influence of CSRNP1 silencing on the phosphorylation patterns of critical signaling proteins such as p53, p-Akt, and p-MDM2. Our results demonstrated significantly lower CSRNP1 expression in NSCLC tumor tissues (P < 0.01). ROC analysis indicated that NSCLC patients with high CSRNP1 expression exhibited extended overall survival and disease-free survival. Furthermore, CSRNP1 silencing promoted NSCLC cells viability, migration, and invasion (P < 0.05). Mechanistically, CSRNP1 silencing led to increased phosphorylation of AKT and MDM2, along with a concurrent reduction in p53 protein expression, suggesting its impact on NSCLC through deregulated cell cycle processes. In conclusion, our study underscores the significance of CSRNP1 in NSCLC pathogenesis, offering insights for targeted therapeutic interventions of NSCLC.
Collapse
Affiliation(s)
- Zhongneng Xu
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Hao Zhou
- Department of Thoracic Surgery, Guanyun People's Hospital, Guanyun, Sichuan, 222299, China
| | - Yonggang Luo
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Nunu Li
- Department of Sanatorium 1, Air Force Health Care Center for Special Service Hangzhou Sanatorium 5, Hangzhou, Zhejiang, 310002, China
| | - Sheng Chen
- Department of Thoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| |
Collapse
|
4
|
Zaarour RF, Ribeiro M, Azzarone B, Kapoor S, Chouaib S. Tumor microenvironment-induced tumor cell plasticity: relationship with hypoxic stress and impact on tumor resistance. Front Oncol 2023; 13:1222575. [PMID: 37886168 PMCID: PMC10598765 DOI: 10.3389/fonc.2023.1222575] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Collapse
Affiliation(s)
- RF. Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - M. Ribeiro
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - B. Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - S. Kapoor
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - S. Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
5
|
Zhang T, Song X, Qiao J, Zhu R, Ren Y, Shan PF. A Novel Predictive Model for Adrenocortical Carcinoma Based on Hypoxia- and Ferroptosis-Related Gene Expression. Front Med (Lausanne) 2022; 9:856606. [PMID: 35652069 PMCID: PMC9148996 DOI: 10.3389/fmed.2022.856606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe impact of hypoxia on ferroptosis is important in cancer proliferation, but no predictive model combining hypoxia and ferroptosis for adrenocortical carcinoma (ACC) has been reported. The purpose of this study was to construct a predictive model based on hypoxia- and ferroptosis-related gene expression in ACC.MethodsWe assessed hypoxia- and ferroptosis-related gene expression using data from 79 patients with ACC in The Cancer Genome Atlas (TCGA). Then, a predictive model was constructed to stratify patient survival using least absolute contraction and selection operation regression. Gene expression profiles of patients with ACC in the Gene Expression Omnibus (GEO) database were used to verify the predictive model.ResultsBased on hypoxia-related gene expression, 79 patients with ACC in the TCGA database were divided into three molecular subtypes (C1, C2, and C3) with different clinical outcomes. Patients with the C3 subtype had the shortest survival. Ferroptosis-related genes exhibited distinct expression patterns in the three subtypes. A predictive model combining hypoxia- and ferroptosis-related gene expression was constructed. A nomogram was constructed using age, sex, tumor stage, and the predictive gene model. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that the gene signature was mainly related to the cell cycle and organelle fission.ConclusionThis hypoxia-and ferroptosis-related gene signature displayed excellent predictive performance for ACC and could serve as an emerging source of novel therapeutic targets in ACC.
Collapse
|
6
|
Wei L, Yuan N, Chen Y, Gong P. Aberrant expression of HIF3A in plasma of patients with non-small cell lung cancer and its clinical significance. J Clin Lab Anal 2021; 35:e23889. [PMID: 34245041 PMCID: PMC8373323 DOI: 10.1002/jcla.23889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Background Hypoxia‐inducible factors (HIFs) have been evaluated in various cancers and diseases. However, the specific role of hypoxia‐inducible factor 3 alpha (HIF3A) in non‐small cell lung cancer (NSCLC) remains controversial. Materials and Methods We investigated HIF3A mRNA expression in the plasma and tumor tissues of patients with NSCLC and explored its clinical significance. Plasma samples from 103 cases of lung adenocarcinoma (LUAD) and 96 cases of lung squamous cell carcinoma (LUSC), and tumor‐adjacent normal tissues from 58 LUAD and 62 LUSC cases were retrospectively evaluated at the No.8 People's Hospital of Qing Dao. HIF3A expression was explored using RT‐qPCR. The clinical significance of HIF3A was evaluated in the plasma and tumor tissues using the receiver operating curve (ROC) and the area under the curve (AUC). Results Hypoxia‐inducible factor 3 alpha expression was notably downregulated in the plasma or tumor tissues of patients with LUAD and LUSC, compared with the healthy control group or adjacent normal tissues. Furthermore, HIF3A expression had a significant positive correlation in the plasma and tumor tissues of LUAD and LUSC patients. Meanwhile, the ROC‐AUCs achieved a significantly higher range, from 0.84 to 0.93, with the plasma or tumor tissues of NSCLC patients. Thus, HIF3A expression was not only correlated with plasma and tumor tissues, but also showed potential significance in NSCLC. Conclusion Hypoxia‐inducible factor 3 alpha is aberrantly detectable in NSCLC patients in the plasma and tumor tissues. HIF3A may be involved in hypoxic responses during the development and occurrence of NSCLC.
Collapse
Affiliation(s)
- Liang Wei
- Department of Thoracic Surgery, The No.8 People's Hospital of Qingdao, Qingdao, China
| | - Na Yuan
- Department of Thoracic Surgery, The No.8 People's Hospital of Qingdao, Qingdao, China
| | - Yingying Chen
- Department of Thoracic Surgery, The No.8 People's Hospital of Qingdao, Qingdao, China
| | - Pingping Gong
- Department of Thoracic Surgery, The No.8 People's Hospital of Qingdao, Qingdao, China
| |
Collapse
|