1
|
Spinello Z, Besharat ZM, Mainiero F, Rughetti A, Masuelli L, Ferretti E, Catanzaro G. MiR-326: Role and significance in brain cancers. Noncoding RNA Res 2025; 12:56-64. [PMID: 40115178 PMCID: PMC11925037 DOI: 10.1016/j.ncrna.2025.02.006] [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: 10/21/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/23/2025] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that act as critical regulators of gene expression by repressing mRNA translation. The role of miRNAs in cell physiology spans from cell cycle control to cell proliferation and differentiation, both during development and in adult tissues. Accordingly, dysregulated expression of miRNAs has been reported in several diseases, including cancer, where miRNAs can act as oncogenes or oncosuppressors. Of note, miRNA signatures are also under investigation for classification, diagnosis, and prognosis of cancer patients. Brain tumours are primarily associated with poor prognosis and high mortality, highlighting an urgent need for novel diagnostic, prognostic, and therapeutic tools. Among miRNAs investigated in brain tumours, miR-326 has been shown to act as a tumour suppressor in adult and paediatric brain cancers. In this review, we describe the role of miR-326 in malignant as well as benign cancers originating from brain tissue. In addition, since miR-326 expression can be regulated by other non-coding RNA species, adding a further layer of regulation in the cancer-promoting axis, we discuss this miRNA's role in targeted therapy for brain cancers.
Collapse
Affiliation(s)
- Zaira Spinello
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Fabrizio Mainiero
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Giuseppina Catanzaro
- Department of Life Science, Health, and Health Professions, Link Campus University, 00165, Rome, Italy
| |
Collapse
|
2
|
Zehtabi M, Ghaedrahmati F, Dari MAG, Moramezi F, Kempisty B, Mozdziak P, Farzaneh M. Emerging biologic and clinical implications of miR-182-5p in gynecologic cancers. Clin Transl Oncol 2025; 27:2367-2382. [PMID: 39661239 DOI: 10.1007/s12094-024-03822-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
MicroRNAs (miRNAs) have emerged as important regulators of gene expression in various biological processes, including cancer. miR-182-5p has gained attention for its potential implications in gynecologic cancers, including breast, ovarian, endometrial, and cervical cancers. miR-182-5p dysregulation has been associated with multiple facets of tumor biology in gynecologic cancers, including tumor initiation, progression, metastasis, and therapeutic response. Studies have highlighted its involvement in key signaling pathways and cellular processes that contribute to cancer development and progression. In addition, miR-182-5p has shown potential as a diagnostic and prognostic biomarker, with studies demonstrating its correlation with clinicopathological features and patient outcomes. Furthermore, the therapeutic potential of miR-182-5p is being explored in gynecologic cancers. Strategies such as miRNA mimics or inhibitors targeting miR-182-5p have shown promise in preclinical and early clinical studies. These approaches aim to modulate miR-182-5p expression, restoring normal cellular functions and potentially enhancing treatment responses. Understanding the biologic and clinical implications of miR-182-5p in gynecologic cancers is crucial for the development of targeted therapeutic strategies and personalized medicine approaches. Further investigations are needed to unravel the specific target genes and pathways regulated by miR-182-5p. It is important to consider the emerging biologic and clinical implications of miR-182-5p in gynecologic cancers.
Collapse
Affiliation(s)
- Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farideh Moramezi
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bartosz Kempisty
- Department of Human Morphology and Embryology, Division of Anatomy, Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, Poland
- Physiology Graduate Faculty North, Carolina State University, Raleigh, NC, 27695, USA
- Center of Assisted Reproduction Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| | - Paul Mozdziak
- Physiology Graduate Faculty North, Carolina State University, Raleigh, NC, 27695, USA
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Clinical Research Development Unit, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Wawro M, Kochan J, Kasza A. Regnases play a crucial role in guarding against cancer development. Biochim Biophys Acta Rev Cancer 2025:189352. [PMID: 40360134 DOI: 10.1016/j.bbcan.2025.189352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Transcript turnover, a fundamental process in maintaining cellular homeostasis, involves intricate interactions between cis-acting sequences and trans-acting factors. Recent advancements in RNA decay research have illuminated novel ribonucleases (RNases) and regulatory elements within mRNA untranslated regions (UTRs), shedding light on the complexity of this process. Members of the Regnase/ZC3H12/MCPIP family (Regnase-1-4) emerge as multifaceted regulators in inflammation and cancer biology. Here, we focused on studies discussing the role of Regnases in cancer. Understanding the intricate roles of Regnase family proteins provides insights into cellular homeostasis and disease pathology, offering promising avenues for targeted therapeutic interventions in inflammation-related disorders and cancer.
Collapse
Affiliation(s)
- Mateusz Wawro
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Jakub Kochan
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Aneta Kasza
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
4
|
Du Y, Kajino T, Shimada Y, Takahashi T, Taguchi A. Mir-494-3p enhances aggressive phenotype of non-small cell lung cancer cells by regulating SET/I2PP2A. Sci Rep 2025; 15:15441. [PMID: 40316770 PMCID: PMC12048640 DOI: 10.1038/s41598-025-99558-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
A high level of miR-494-3p expression has been associated with poor prognosis of non-small cell lung cancer (NSCLC) patients. However, its role in NSCLC development and progression remains elusive. Analyses of the Clinical Proteomic Tumor Analysis Consortium and the Cancer Genome Atlas databases showed overexpression of miR-494-3p in both lung adenocarcinoma and lung squamous cell carcinoma cases. Furthermore, bioinformatic analysis revealed that representative pathways associated with cancer metastasis were enriched with genes positively correlated with miR-494-3p expression levels, suggesting possible involvement of miR-494-3p in the aggressive properties of NSCLC. To identify potential targets of miR-494-3p, genes inversely correlated with miR-494-3p in the mRNA expression datasets of NSCLC cell lines obtained from the Cancer Dependency Map were examined in the present study. Integration of RNA sequencing analysis of NSCLC cells with miR-494-3p inhibition and a bioinformatic search of miRNA target prediction algorithms resulted in identification of SET/I2PP2A as a direct target of miR-494-3p. The findings indicate that suppression of SET/I2PP2A by miR-494-3p promotes NSCLC cell migration and invasion, but not viability, thus indicating miR-494-3p and its downstream molecules as potential therapeutic targets for aggressive NSCLC phenotypes.
Collapse
Affiliation(s)
- Yuwen Du
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan
- Division of Advanced Cancer Diagnostics, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan
| | - Taisuke Kajino
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan
| | - Yukako Shimada
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan
| | | | - Ayumu Taguchi
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan.
- Division of Advanced Cancer Diagnostics, Nagoya University Graduate School of Medicine, Nagoya, 466-8560, Japan.
| |
Collapse
|
5
|
Cen YY, Gao XL, Feng YH, Zhou C, Li CJ, Liu F, Shen JF, Zhang YY. The Double-Edged Effect of Connexins and Pannexins of Glial Cells in Central and Peripheral Nervous System After Nerve Injury. Mol Neurobiol 2025:10.1007/s12035-025-04991-6. [PMID: 40310549 DOI: 10.1007/s12035-025-04991-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Glial cells play pivotal roles in homeostatic regulation and driving reactive pathologic changes after nerve injury. Connexins (Cxs) and pannexins (Panxs) have emerged as seminal proteins implicated in cell-cell communication, exerting a profound impact on the response processes of glial cell activation, proliferation, protein synthesis and secretion, as well as apoptosis following nerve injury. These influences are mediated through various forms, including protein monomers, hemichannel (HC), and gap junction (GJ), mainly by regulating intercellular or intracellular signaling pathways. Multiple Cx and Panx isoforms have been detected in central nervous system (CNS) or peripheral nervous system (PNS). Each isoform exhibits distinct cellular and subcellular localization, and the differential regulation and functional roles of various protein isoforms are observed post-injury. The quantitative and functional alterations of the same protein isoform in different studies remain inconsistent, attributable to factors such as the predominant mode of protein polymerization, the specific injury model, and the injury site. Similarly, the same protein isoforms have different roles in regulating the response processes after nerve injury, thus exerting a double-edged sword effect. This review describes the regulatory mechanisms and bidirectional effects of Cxs and Panxs. Additionally, it surveys the current status of research and application of drugs as therapeutic targets for neuropathic injuries. We summarize comprehensive and up-to-date information on these proteins in the glial cell response to nerve injury, providing new perspectives for future mechanistic exploration and development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yue-Yan Cen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Xin-Lin Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Yu-Heng Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renminnan Road, Chengdu, 610041, China.
| |
Collapse
|
6
|
Khazaei-Poul Y, Firouzjaei AA, Paryan M, Tafti A, Mohammadi-Yeganeh S. Evaluation of the impact of miR-3143 on the PI3K/AKT signaling pathway and its subsequent influence on the metastatic phenotype of triple-negative breast cancer cells. Exp Cell Res 2025; 448:114552. [PMID: 40203985 DOI: 10.1016/j.yexcr.2025.114552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
MicroRNAs (miRNAs) are recognized to have a pivotal role in the progression and metastatic dissemination encompassing diverse cancer varieties, such as triple-negative breast cancer (TNBC). Recent evidence has suggested that specific miRNA species can directly or indirectly influence the onset, progression, and relapse of TNBC. Previous studies have reported the frequent reduction of miR-3143 in TNBC, which appears to coincide with the activation of proliferative signaling pathways. However, the potential restorative effects of miR-3143 on TNBC cellular behavior remain unexplored. In the present study, we utilized exosome-mediated delivery to introduce miR-3143 into TNBC cells and investigated its impact on the PI3K/AKT pathway and the resulting effects on cellular proliferation, movement, and apoptosis. MDA-MB-231 TNBC cells underwent treatment with miR-3143-electroporated human umbilical cord mesenchymal stem cell (HUCMSC)-derived exosomes. RT-qPCR analysis was utilized to assess the influence of miR-3143 overexpression on the expression of its target genes, PIK3CA and AKT1, which was further validated through dual-luciferase reporter assays. Our results demonstrated that the overexpression of miR-3143 could effectively decline the level of AKT1 and PIK3CA by directly binding to their 3'-UTRs. Furthermore, the introduction of miR-3143 into TNBC cells resulted in a significant enhancement of apoptotic activities. Interestingly, the delivery of miR-3143 via HUCMSC-derived exosomes could inhibit the protumorigenic and prometastatic behaviors of TNBC cells, potentially limiting their malignant progression. Collectively, these findings enhance comprehension of the regulatory mechanisms by which miR-3143 can modulate the metastatic potential of TNBC cells. The insights gained from this study may facilitate the creation of innovative miRNA-targeting approaches to combat the aggressive nature of TNBC andstrengthen treatment effectiveness.
Collapse
Affiliation(s)
- Yalda Khazaei-Poul
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Paryan
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Tafti
- Department of Biotechnology and Molecular Medicine, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Guo X, Zhao Z, Zhu L, Liu S, Zhou L, Wu F, Fang S, Chen M, Zheng L, Ji J. The evolving landscape of biomarkers for systemic therapy in advanced hepatocellular carcinoma. Biomark Res 2025; 13:60. [PMID: 40221793 PMCID: PMC11993949 DOI: 10.1186/s40364-025-00774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and deadliest cancers. With the approval of multiple first- and second-line agents, especially the combination therapies based on immune checkpoint inhibitor (ICI) regimens, the landscape of systemic therapy for advanced HCC (aHCC) is more diverse than ever before. The efficacy of current systemic therapies shows great heterogeneity in patients with aHCC, thereby identifying biomarkers for response prediction and patient stratification has become an urgent need. The main biomarkers for systemic therapy in hepatocellular carcinoma are derived from peripheral blood, tissues, and imaging. Currently, the understanding of the clinical response to systemic therapy indicates unequivocally that a single biomarker cannot be used to identify patients who are likely to benefit from these treatments. In this review, we provide an integrated landscape of the recent development in molecular targeted therapies and ICIs-based therapies, especially focusing on the role of clinically applicable predictive biomarkers. Additionally, we further highlight the latest advancements in biomarker-driven therapies, including targeted treatments, adoptive cell therapies, and bispecific antibodies.
Collapse
Affiliation(s)
- Xinyu Guo
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingyi Zhu
- The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Shuang Liu
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingling Zhou
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Fazong Wu
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Liyun Zheng
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| | - Jiansong Ji
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
8
|
Bergantim R, Peixoto da Silva S, Pinto V, Pereira JM, Sousa D, Trigo F, Matthiesen R, Guimarães JE, Vasconcelos MH. MicroRNA-665 and its potential role in drug response and survival outcomes in multiple myeloma: a preliminary study. Front Pharmacol 2025; 16:1465814. [PMID: 40255571 PMCID: PMC12006192 DOI: 10.3389/fphar.2025.1465814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/04/2025] [Indexed: 04/22/2025] Open
Abstract
Background Multiple myeloma (MM) is a complex hematological malignancy with heterogeneous clinical and pathophysiological backgrounds that influence treatment responses and outcomes. Identifying biomarkers to predict drug response and guide treatment decisions, particularly regarding drug combinations, is essential to improve therapeutic efficacy and patient outcomes. This study explores the role of microRNAs (miRNAs/miRs) derived from bone marrow (BM) and peripheral blood (PB) in responses to treatment and survival outcomes in newly diagnosed MM (ndMM) patients. Methods This study included twenty patients with ndMM undergoing first-line treatment with bortezomib, thalidomide, and dexamethasone. The miRNAs were isolated from BM and PB, and their profiles were analyzed using Next-Generation Sequencing (NGS), followed by validation of differentially expressed miRNAs by quantitative real-time PCR (qPCR). Clinical and response data were collected to assess correlations between miRNA levels, clinical characteristics, and patient outcomes. In silico analysis for target-prediction and gene ontology (GO) enrichment was performed to explore the potential biological and functional role of the identified miRNAs. Results NGS profiling revealed several miRNAs differently expressed between treatment-refractory and sensitive patients, as well as between PB and BM. Among these, miR-665, miR-483-5p, miR-143-3p and miR-145-5p were selected for further validation by qPCR. It was observed that miR-665 was significantly elevated in treatment-refractory patients compared to treatment-sensitive patients. Additionally, miR-665 levels were higher in PB than in BM. Elevated miR-665 levels were associated with more aggressive disease characteristics and poorer clinical outcomes, including reduced overall survival. Discussion Our preliminary findings suggest that miR-665 could potentially serve as a non-invasive tool for predicting drug resistance and guiding treatment decisions in MM. These findings also highlight the potential utility of miRNAs in liquid biopsies as a predictive tool of drug response in MM and could pave the way for personalized treatment strategies, improving patient outcomes. Future research is needed to validate these results in larger cohorts and explore the underlying mechanisms of miR-665 in MM pathogenesis and drug resistance.
Collapse
Affiliation(s)
- Rui Bergantim
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Clinical Hematology, Hospital Center of São João, Porto, Portugal
- Clinical Hematology, FMUP – Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Sara Peixoto da Silva
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Vanessa Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Joana M. Pereira
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Diana Sousa
- Faculty of Dental Medicine (FMD), Universidade Católica Portuguesa, Viseu, Portugal
- Unit for Multidisciplinary Research in Biomedicine (UMIB), School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Laboratory of Integrative and Translocation Research in Population Health (ITR), Porto, Portugal
| | - Fernanda Trigo
- Clinical Hematology, Hospital Center of São João, Porto, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - José E. Guimarães
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Clinical Hematology, Hospital Center of São João, Porto, Portugal
- Clinical Hematology, FMUP – Faculty of Medicine of the University of Porto, Porto, Portugal
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário IUCSESPU, Paredes, Portugal
| | - M. Helena Vasconcelos
- i3S – Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| |
Collapse
|
9
|
Kim H, Lee YY, Kim VN. The biogenesis and regulation of animal microRNAs. Nat Rev Mol Cell Biol 2025; 26:276-296. [PMID: 39702526 DOI: 10.1038/s41580-024-00805-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/21/2024]
Abstract
MicroRNAs (miRNAs) are small, yet profoundly influential, non-coding RNAs that base-pair with mRNAs to induce RNA silencing. Although the basic principles of miRNA biogenesis and function have been established, recent breakthroughs have yielded important new insights into the molecular mechanisms of miRNA biogenesis. In this Review, we discuss the metazoan miRNA biogenesis pathway step-by-step, focusing on the key biogenesis machinery, including the Drosha-DGCR8 complex (Microprocessor), exportin-5, Dicer and Argonaute. We also highlight newly identified cis-acting elements and their impact on miRNA maturation, informed by advanced high-throughput and structural studies, and discuss recently discovered mechanisms of clustered miRNA processing, target recognition and target-directed miRNA decay (TDMD). Lastly, we explore multiple regulatory layers of miRNA biogenesis, mediated by RNA-protein interactions, miRNA tailing (uridylation or adenylation) and RNA modifications.
Collapse
Affiliation(s)
- Haedong Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Young-Yoon Lee
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea.
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Liang Y, Liang M, Yan T, Meng X, Zhou B, Gao Y. miR-185-5p May Modulate the Chemosensitivity of LUSC to Cisplatin via Targeting PCDHA11: Multi-omics Analysis and Experimental Validation. Biochem Genet 2025; 63:1734-1751. [PMID: 38613717 DOI: 10.1007/s10528-024-10795-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
Drug resistance is the major difficulty in treatment of lung squamous cell carcinoma (LUSC). This study aims to explore drug response-related miRNAs (DRmiRNAs) based on multi-omics research. We identified DRmiRNAs of LUSC with a multi-omics integrated system that combines expression data of microRNA, lncRNA, mRNA, methylation levels, somatic mutations. After identifying DRmiRNAs, we screened and validated of the target mRNAs of DRmiRNAs through Targetscan and the miRDB database. Then, Real-time PCR and Western blot assays were used to estimate the expression of DRmiRNAs and target protein, and the dual-luciferase assays were used to confirm the interaction of DRmiRNAs and target mRNA. Furthermore, CCK-8 (Cell Counting Kit-8) assays were used to evaluate cell proliferation and drug sensitivity. After integrated analysis, hsa-miR-185-5p was identified as DRmiRNA based on multi-omics data. Through Targetscan and miRDB database, the possible target mRNAs were obtained and PCDHA11 was validated as a target mRNA of miR-185-5p by real-time PCR, Western blot assays and dual-luciferase assays. CCK-8 assays and clone formation assays showed that the proliferation of miR-185-5p mimics was significantly slower than that of miR-185-5p inhibitors, which means overexpression of miR-185-5p enhanced the anticancer effects of cisplatin, whereas the downregulation of miR-185-5p reduced the effects. Furthermore, the proliferation of silencing PCDHA11 was significantly slower than that of overexpression of PCDHA11, which means PCDHA11 overexpression weakened the anticancer effects of cisplatin, and silencing PCDHA11 enhanced the effects. This study demonstrated that miR-185-5p was involved in chemoresistance of LUSC cells to cisplatin partly via down-regulating PCDHA11, which may promote understanding the underlying molecular mechanisms of drug response.
Collapse
Affiliation(s)
- Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Yan
- Department of Anesthesia, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Panjiayuan, Nanli 17, Beijing, 100021, People's Republic of China
| | - Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Sareen G, Mohan M, Mannan A, Dua K, Singh TG. A new era of cancer immunotherapy: vaccines and miRNAs. Cancer Immunol Immunother 2025; 74:163. [PMID: 40167762 PMCID: PMC11961864 DOI: 10.1007/s00262-025-04011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has transformed the treatment landscape, introducing new strategies to fight various types of cancer. This review examines the important role of vaccines in cancer therapy, focusing on recent advancements such as dendritic cell vaccines, mRNA vaccines, and viral vector-based approaches. The relationship between cancer and the immune system highlights the importance of vaccines as therapeutic tools. The discussion covers tumor cell and dendritic cell vaccines, protein/peptide vaccines, and nucleic acid vaccines (including DNA, RNA, or viral vector-based), with a focus on their effectiveness and underlying mechanisms. Combination therapies that pair vaccines with immune checkpoint inhibitors, TIL therapy, and TCR/CAR-T cell therapy show promising potential, boosting antitumor responses. Additionally, the review explores the regulatory functions of microRNAs (miRNAs) in cancer development and suppression, featuring miR-21, miR-155, the let-7 family, and the miR-200 family, among others. These miRNAs influence various pathways, such as PI3K/AKT, NF-κB, and EMT regulation, providing insights into biomarker-driven therapeutic strategies. Overall, this work offers a thorough overview of vaccines in oncology and the integrative role of miRNAs, setting the stage for the next generation of cancer immunotherapies.
Collapse
Affiliation(s)
- Gitika Sareen
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
12
|
Biswas T, Hassan H, Rohner N. Differentially expressed miRNAs offer new perspective into cave adaptation of Astyanax mexicanus. Ann N Y Acad Sci 2025; 1546:173-181. [PMID: 40082196 PMCID: PMC11998478 DOI: 10.1111/nyas.15300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Astyanax mexicanus, a species with both surface-dwelling and multiple cave-dwelling populations, offers a unique opportunity to study repeated adaptation to dark and resource-scarce environments. While previous work has identified large-scale gene expression changes between morphs under even identical laboratory conditions, the regulatory basis of these expression differences remains largely unexplored. In this study, we focus on microRNAs (miRNAs) as key regulators of gene expression. Our analysis identified 683 mature miRNAs, establishing the first comprehensive catalog of miRNAs for this species. We identified a unique subset of differentially expressed miRNAs common to all studied cave-dwelling populations, potentially orchestrating the nuanced gene expression patterns required for survival in the cave milieu. Furthermore, we performed in silico target prediction of these miRNAs, revealing possible roles in developmental and metabolic pathways pivotal for thriving in nutrient-limited cave conditions. Interestingly, we also observed that Molino, which is the "youngest" of the three cavefish analyzed in this study, exhibited the most abundant number of differentially expressed mature miRNAs among the cave morphs. The comprehensive miRNA catalog generated, along with the insight into their differential expression across different morphs, will guide future investigations into the intricate world of miRNA-mediated evolution of complex traits.
Collapse
Affiliation(s)
| | - Huzaifa Hassan
- Stowers Institute for Medical ResearchKansas CityMissouriUSA
| | - Nicolas Rohner
- Stowers Institute for Medical ResearchKansas CityMissouriUSA
- Institute for Integrative Cell Biology and PhysiologyUniversity of MünsterMünsterGermany
| |
Collapse
|
13
|
Wei Q, Lin N, Wang L. Targeting DNA Topoisomerase IIα in Retinoblastoma: Implications in EMT and Therapeutic Strategies. Biologics 2025; 19:113-123. [PMID: 40123578 PMCID: PMC11929414 DOI: 10.2147/btt.s499314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
Background This study investigates the role of DNA topoisomerase IIα (TOP2A) in retinoblastoma (RB), focusing on its involvement in epithelial-mesenchymal transition (EMT) and the potential of TOP2A inhibition as a therapeutic strategy. Methods We analyzed TOP2A expression in RB tissues using public gene expression databases (GSE97508, GSE110811, and GSE172170) and conducted functional assays in human RB cell lines (Y79 and WERI-Rb-1) modified to knock down or overexpress TOP2A. Assessments included cell proliferation, migration, invasion, and EMT marker expression via RT-PCR and Western blot. Additionally, we evaluated the effects of TOP2A modulation in subcutaneous and liver metastasis mouse xenograft models. Results TOP2A was significantly overexpressed in RB tissues (p < 0.0001). In vitro, TOP2A knockdown inhibited RB cell proliferation, migration, and invasion, and reversed EMT marker expression (p < 0.05), while TOP2A overexpression enhanced these oncogenic processes. In vivo, TOP2A knockdown or inhibition significantly reduced tumor growth and metastasis in both subcutaneous and liver metastasis models (p < 0.05). Combination therapy with TOP2A and EMT inhibitors further enhanced anti-tumor effects, significantly reducing tumor burden and metastatic lesions (p < 0.01). Conclusion TOP2A is pivotal in RB pathogenesis and progression, primarily by regulating EMT. Its inhibition not only curtails RB cell proliferation and metastasis but also reverses EMT, underscoring its potential as a therapeutic target. This study lays the groundwork for further exploration of TOP2A-targeted therapies in RB.
Collapse
Affiliation(s)
- Qingquan Wei
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Nan Lin
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Li Wang
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
14
|
Liu H, Lv MM, Li X, Su M, Nie YG, Ying ZM. Ligation-recognition triggered RPA-Cas12a cis-cleavage fluorogenic RNA aptamer for one-pot and label-free detection of MicroRNA in breast cancer. Biosens Bioelectron 2025; 272:117106. [PMID: 39740588 DOI: 10.1016/j.bios.2024.117106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/21/2024] [Accepted: 12/26/2024] [Indexed: 01/02/2025]
Abstract
"One-pot" assays which combine amplification with CRISPR/Cas12a system are in constant attracted for biosensors development. Herein, we present a one-pot isothermal assay that Ligation-recognition triggered Recombinase Polymerase Amplification (RPA)-CRISPR/Cas12a cis-cleavage (LRPA-CRISPR) fluorescent biosensor for sensitive, specific, and label-free miRNA detection. Firstly, we reveal the programmed double-stranded DNA amplicons, which utilized the ligation-recognition and polymerization to form and amplified by the RPA system. Meanwhile, we enabled exponential ligation-recognition triggered recombinase polymerase amplification of miRNA-21 sequences and exploited the cis-cleavage mechanism of Cas12a with transcription to generate functional Mango RNA for signal output. This assay can be completed within 40 min and can allow a limit of detection of 3.43 aM for miRNA-21 detection, owing to the RPA with transcription amplification and enables to product the functional Mango RNA aptamer by in vitro transcription that binds to the TO1-Biotin fluorogenic dye. Moreover, our method exhibits the advantages of self-supply crRNA, label-free, excellent specificity, and universal detection platform via the design of one-pot detection in serum and cell samples, showing tremendous potential in biomarkers diagnostics of breast cancer.
Collapse
Affiliation(s)
- Hao Liu
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Meng-Mei Lv
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan, 410000, China
| | - Xiang Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Mei Su
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Yin-Gang Nie
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Zhan-Ming Ying
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, China.
| |
Collapse
|
15
|
Diallo MT, Chen B, Yao Q, Yan Z, Sun Q, Wang D. KIF3C inhibits the progression and proliferation of colorectal cancer. BMC Gastroenterol 2025; 25:165. [PMID: 40075273 PMCID: PMC11899393 DOI: 10.1186/s12876-024-03489-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/30/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Evidence indicated that KIF3C, a member of the kinesin superfamily of motor proteins, exhibits significant upregulation across various cancer types. Consequently, its impact on cancer advancement, including cell proliferation, migration, and invasion, is evident. Nonetheless, the comprehension of KIF3C's expression and role in colorectal cancer (CRC) remains limited. METHODS Immunohistochemistry was used to evaluate the presence of KIF3C in CRC. The expression levels of KIF3C were assessed in CRC cells through western blot analysis (WB) and real-time polymerase chain reaction (RT-qPCR). KIF3C was knockdown and overexpressed using lentiviral vectors in the human CRC cell lines SW-480, HCT-116, and SW-620. In vitro experiments such as transwell assays, scratch wound healing, colony formation assays, counting cell CCK-8, and signaling pathway experiments were conducted to validate the KIF3C function in CRC cells. RESULTS We demonstrated that KIF3C is highly expressed in cells and tissues of CRC, and this expression is closely associated with tumor prognosis. It was shown that KIF3C knockdown significantly inhibited tumor cell proliferation and migration in CRC cells. Additionally, the KIF3C signaling pathway experiment in this study promoted the CRC progression by upregulating the PI3K/AKT, Bax, and Bcl-2 pathways. CONCLUSIONS KIF3C knockdown promoted CRC proliferation, as it could be a potential therapeutic target for treating CRC.
Collapse
Affiliation(s)
- Maladho Tanta Diallo
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China
| | - Bangquan Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China
| | - Qing Yao
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China
| | - Zhang Yan
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China
| | - Qiannan Sun
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China
| | - Daorong Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China.
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China.
- Medical College of Yangzhou University, Yangzhou, 225001, China.
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, Yangzhou, 225001, China.
| |
Collapse
|
16
|
Molina-Pelayo FA, Zarate-Lopez D, García-Carrillo R, Rodríguez-Beas C, Íñiguez-Palomares R, Rodríguez-Mejía JL, Soto-Guzmán A, Velasco-Loyden G, Sierra-Martínez M, Virgen-Ortiz A, Sánchez-Pastor E, Magaña-Vergara NE, Baltiérrez-Hoyos R, Alamilla J, Chagoya de Sánchez V, Dagnino-Acosta A, Chávez E, Castro-Sánchez L. miRNAs-Set of Plasmatic Extracellular Vesicles as Novel Biomarkers for Hepatocellular Carcinoma Diagnosis Across Tumor Stage and Etiologies. Int J Mol Sci 2025; 26:2563. [PMID: 40141205 PMCID: PMC11942138 DOI: 10.3390/ijms26062563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, often diagnosed at advanced stages due to insufficient early screening and monitoring. MicroRNAs (miRNAs) are key regulators of gene expression and potential biomarkers for cancer diagnosis. This study investigated the diagnostic potential of miRNAs in Extracellular Vesicles (EVs) from HCC. miRNA expression in EVs was analyzed using HCC cell lines, circulating EVs from a Diethylnitrosamine (DEN)-induced liver tumor rat model, and plasma samples from HCC patients. Receiver Operating Characteristics (ROCs) were applied to evaluate the diagnostic accuracy of circulating EV miRNAs in patients. Five miRNAs (miR-183-5p, miR-19a-3p, miR-148b-3p, miR-34a-5p, and miR-215-5p) were consistently up-regulated in EVs across in vitro and in vivo HCC models. These miRNAs showed statistically significant differences in HCC patients stratified by TNM staging and Edmondson-Steiner grading compared to healthy controls. They also differentiated HCC patients with various etiologies from the control group and distinguished HCC patients, with or without liver cirrhosis, from cirrhotic and healthy individuals. Individually and as a panel, they demonstrated high sensitivity, specificity, and accuracy in identifying HCC patients. Their consistent upregulation across models and clinical samples highlights their robustness as biomarkers for HCC diagnosis, offering the potential for early disease management and prognosis.
Collapse
Affiliation(s)
- Francisco A. Molina-Pelayo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - David Zarate-Lopez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - Rosendo García-Carrillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - César Rodríguez-Beas
- Departamento de Física, Universidad de Sonora, Hermosillo 83000, Sonora, Mexico; (C.R.-B.); (R.Í.-P.)
| | - Ramón Íñiguez-Palomares
- Departamento de Física, Universidad de Sonora, Hermosillo 83000, Sonora, Mexico; (C.R.-B.); (R.Í.-P.)
| | - José L. Rodríguez-Mejía
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - Adriana Soto-Guzmán
- Departamento de Medicina y Ciencias de la Salud, Universidad de Sonora, Hermosillo 83000, Sonora, Mexico;
| | - Gabriela Velasco-Loyden
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (G.V.-L.); (V.C.d.S.)
| | - Mónica Sierra-Martínez
- Unidad de investigación en Salud, Hospital Regional de Alta Especialidad de Ixtapaluca, Servicios de Salud del Instituto Mexicano del Seguro Social para el Bienestar (IMSS-BIENESTAR), Ciudad de México 01020, Mexico;
| | - Adolfo Virgen-Ortiz
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - Enrique Sánchez-Pastor
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
| | - Nancy E. Magaña-Vergara
- Facultad de Ciencias Químicas, Universidad de Colima, Coquimatlán 28400, Colima, Mexico;
- SECIHTI—Universidad de Colima, Colima 28045, Colima, Mexico
| | | | - Javier Alamilla
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
- SECIHTI—Universidad de Colima, Colima 28045, Colima, Mexico
| | - Victoria Chagoya de Sánchez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (G.V.-L.); (V.C.d.S.)
| | - Adán Dagnino-Acosta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
- SECIHTI—Universidad de Colima, Colima 28045, Colima, Mexico
| | - Enrique Chávez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (G.V.-L.); (V.C.d.S.)
| | - Luis Castro-Sánchez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28045, Colima, Mexico; (F.A.M.-P.); (D.Z.-L.); (R.G.-C.); (J.L.R.-M.); (A.V.-O.); (E.S.-P.); (J.A.); (A.D.-A.)
- SECIHTI—Universidad de Colima, Colima 28045, Colima, Mexico
| |
Collapse
|
17
|
Ljungström M, Oltra E. Methods for Extracellular Vesicle Isolation: Relevance for Encapsulated miRNAs in Disease Diagnosis and Treatment. Genes (Basel) 2025; 16:330. [PMID: 40149481 PMCID: PMC11942051 DOI: 10.3390/genes16030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles that facilitate intercellular communication by carrying essential biomolecules under physiological and pathological conditions including microRNAs (miRNAs). They are found in various body fluids, such as blood, urine, and saliva, and their levels fluctuate with disease progression, making them valuable diagnostic tools. However, isolating EVs is challenging due to their small size and biological complexity. Here, we summarize the principles behind the most common EV isolation methods including ultracentrifugation, precipitation, immunoaffinity, sorting, ultrafiltration, size exclusion chromatography, and microfluidics while highlighting protocol strengths and weaknesses. We also review the main strategies to identify and quantify circulating miRNAs with a particular focus on EV-encapsulated miRNAs. Since these miRNAs hold special clinical interest derived from their superior stability and therapeutic potential, the information provided here should provide valuable guidance for future research initiatives in the promising field of disease diagnostic and treatment based on EV-encapsulated miRNAs.
Collapse
Affiliation(s)
- Maria Ljungström
- Escuela de Doctorado, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain;
| | - Elisa Oltra
- Department of Pathology, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
18
|
Saxena T, Quan A, Chan E, Kozlova N, Matai L, Lee JD, Rupaimoole R, Beca F, Muranen T, Slack FJ. EGFR-induced lncRNA TRIDENT promotes drug resistance in non-small cell lung cancer via phospho-TRIM28-mediated DNA damage repair. Proc Natl Acad Sci U S A 2025; 122:e2415389122. [PMID: 40030013 PMCID: PMC11912419 DOI: 10.1073/pnas.2415389122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) play numerous roles in cellular biology and alterations in lncRNA expression profiles have been implicated in a variety of cancers. Here, we identify and characterize a lncRNA, TRIM28 Interacting DNA damage repair Enhancing Noncoding Transcript (TRIDENT), whose expression is induced upon epithelial growth factor receptor (EGFR) activation, and which exerts pro-oncogenic functions in EGFR-driven non-small cell lung cancer. Knocking down TRIDENT leads to decreased tumor-cell proliferation in both in vitro and in vivo model systems and induces sensitization to chemotherapeutic drugs. Using ChIRP-MS analysis we identified TRIM28 as a protein interactor of TRIDENT. TRIDENT promotes phosphorylation of TRIM28 and knocking down TRIDENT leads to accumulation of DNA damage in cancer cells via decreased TRIM28 phosphorylation. Altogether, our results reveal a molecular pathway in which TRIDENT regulates TRIM28 phosphorylation to promote tumor cell growth and drug resistance. Our findings suggest that TRIDENT can be developed as a biomarker or therapeutic target for EGFR mutant non-small cell lung cancer.
Collapse
Affiliation(s)
- Tanvi Saxena
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Anan Quan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Erica Chan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Nina Kozlova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Latika Matai
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jonathan D. Lee
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Rajesha Rupaimoole
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Francisco Beca
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Frank J. Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
19
|
Nor WMFSBWM, Kwong SC, Fuzi AAM, Said NABM, Jamil AHA, Lee YY, Lee SC, Lim YAL, Chung I. Linking microRNA to metabolic reprogramming and gut microbiota in the pathogenesis of colorectal cancer (Review). Int J Mol Med 2025; 55:46. [PMID: 39820715 PMCID: PMC11759585 DOI: 10.3892/ijmm.2025.5487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025] Open
Abstract
Colorectal cancer (CRC), an emerging public health concern, is one of the leading causes of cancer morbidity and mortality worldwide. An increasing body of evidence shows that dysfunction in metabolic reprogramming is a crucial characteristic of CRC progression. Specifically, metabolic reprogramming abnormalities in glucose, glutamine and lipid metabolism provide the tumour with energy and nutrients to support its rapid cell proliferation and survival. More recently, microRNAs (miRNAs) appear to be involved in the pathogenesis of CRC, including regulatory roles in energy metabolism. In addition, it has been revealed that dysbiosis in CRC might play a key role in impairing the host metabolic reprogramming processes, and while the exact interactions remain unclear, the link may lie with miRNAs. Hence, the aims of the current review include first, to delineate the metabolic reprogramming abnormalities in CRC; second, to explain how miRNAs mediate the aberrant regulations of CRC metabolic pathways; third, linking miRNAs with metabolic abnormalities and dysbiosis in CRC and finally, to discuss the roles of miRNAs as potential biomarkers.
Collapse
Affiliation(s)
| | - Soke Chee Kwong
- Centre for Population Health (CePH), Department of Social and Preventive Medicine, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Afiqah Alyaa Md Fuzi
- Office of Deputy Vice Chancellor (Research and Innovation), Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nur Akmarina Binti Mohd Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Malaysia
| | - Soo Ching Lee
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yvonne Ai-Lian Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Chhichholiya Y, Kumar V, Hlawndo L, Sangave K, Singh S, Munshi A. BreVamiR3': A comprehensive database for breast cancer-associated genetic variations in miRNA and 3' UTR of their target genes. Comput Biol Med 2025; 186:109639. [PMID: 39740512 DOI: 10.1016/j.compbiomed.2024.109639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/02/2025]
Abstract
Breast cancer (BC) is a multifactorial disease where microRNA (miRNA)-mediated dysregulated gene expression plays a pivotal role in tumorigenesis, progression, and clinical outcomes. Genetic variation, particularly SNPs in miRNA sequences and the 3' untranslated regions (3'UTRs) of their target genes, can disrupt miRNA-mRNA interactions, leading to altered gene expression. Despite several existing databases providing insights into various aspects of miRNAs and their target genes in association with the development of the disease. Still, there remains a critical gap in a unified, BC-specific repository that integrates SNPs in miRNAs, their target genes, and associated molecular mechanisms. To address this, we developed BreVamiR3' (Breast cancer-associated genetic Variations in miRNA and 3'UTR of their target genes), a novel, freely accessible database (https://brevamir3.web.app/index.html). BreVamiR3' features comprehensive and curated data on 500 miRNAs and 828 target genes associated with BC, including experimentally validated SNPs, chromosomal loci, expression profiles, BC subtypes, signaling pathways, and classifications of genes i.e. oncogene, tumor suppressor, or both. Unique features of BreVamiR3' include detailed annotations of SNPs disrupting miRNA-target interactions, subtype-specific miRNA expression, visualization of miRNA-chromosomal distributions and their experimental validation. Chromosome 19 harbours the highest number of BC-associated miRNAs, while target genes such as CCND1, KRAS, and ERBB2 demonstrate extensive miRNA regulation. By integrating genetic variations with functional and clinical relevance, BreVamiR3' provides a single platform to explore miRNA-mediated regulatory networks, their genetic variation, and downstream signaling pathways in BC. This database serves as a powerful resource for researchers, enabling exploration of miRNA-gene interactions, genetic variation, and their role in pathogenesis of BC and diverse clinical outcomes.
Collapse
Affiliation(s)
- Yogita Chhichholiya
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Vicky Kumar
- Department of Computer Science Engineering, Guru Nanak Dev DSEU Rohini Campus, Delhi Skill and Entrepreneurship University, Dwarka, Delhi, India
| | - Lalnunmawia Hlawndo
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Kshitij Sangave
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
21
|
Ruffo P, Traynor BJ, Conforti FL. Advancements in genetic research and RNA therapy strategies for amyotrophic lateral sclerosis (ALS): current progress and future prospects. J Neurol 2025; 272:233. [PMID: 40009238 PMCID: PMC11865122 DOI: 10.1007/s00415-025-12975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
This review explores the intricate landscape of neurodegenerative disease research, focusing on Amyotrophic Lateral Sclerosis (ALS) and the intersection of genetics and RNA biology to investigate the causative pathogenetic basis of this fatal disease. ALS is a severe neurodegenerative disease characterized by the progressive loss of motor neurons, leading to muscle weakness and paralysis. Despite significant research advances, the exact cause of ALS remains largely unknown. Thanks to the application of next-generation sequencing (NGS) approaches, it was possible to highlight the fundamental role of rare variants with large effect sizes and involvement of portions of non-coding RNA, providing valuable information on risk prediction, diagnosis, and treatment of age-related diseases, such as ALS. Genetic research has provided valuable insights into the pathophysiology of ALS, leading to the development of targeted therapies such as antisense oligonucleotides (ASOs). Regulatory agencies in several countries are evaluating the commercialization of Qalsody (Tofersen) for SOD1-associated ALS, highlighting the potential of gene-targeted therapies. Furthermore, the emerging significance of microRNAs (miRNAs) and long RNAs are of great interest. MiRNAs have emerged as promising biomarkers for diagnosing ALS and monitoring disease progression. Understanding the role of lncRNAs in the pathogenesis of ALS opens new avenues for therapeutic intervention. However, challenges remain in delivering RNA-based therapeutics to the central nervous system. Advances in genetic screening and personalized medicine hold promise for improving the management of ALS. Ongoing clinical trials use genomic approaches for patient stratification and drug targeting. Further research into the role of non-coding RNAs in the pathogenesis of ALS and their potential as therapeutic targets is crucial to the development of effective treatments for this devastating disease.
Collapse
Affiliation(s)
- Paola Ruffo
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Francesca Luisa Conforti
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
22
|
Qi Y, Yu J, Lou M, Yu Y, Li R, Zhang Z, Dai Y, Lao K, Cao M, Gou X. Lab on a single microbead: An enzyme-free strategy for the sensitive detection of microRNA via efficient localized catalytic hairpin assembly. Anal Chim Acta 2025; 1340:343659. [PMID: 39863312 DOI: 10.1016/j.aca.2025.343659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Accurate quantification of microRNA (miRNA) is of great significance because it provides opportunities for the accurate early diagnosis of a series of human diseases including cancers. Currently, complicated nucleic acid amplification technologies are always required for the highly sensitive miRNA detection. The introduction of nucleic acid signal amplification coupled with various enzymes will inevitably lead to tedious work and increase the complexity of the analysis process. It is still urgently desired to develop enzyme-free yet sensitive assays that enable the sensitive analysis of miRNA in complicated biological samples. RESULTS A single microbead (MB)-based localized catalytic hairpin assembly (CHA) strategy is proposed for the sensitive analysis of microRNA (miRNA). This rationally designed CHA strategy allows target miRNA to walk only on a single MB which can create a micro-amplification zone, initiating a highly efficient localized CHA reaction, generating a large number of fluorescent DNA duplexes on the surface of single MB. The efficient localized CHA on single MB can not only greatly suppress the nonspecific reaction between two hairpin probes, thus decreasing the background signal, but also greatly enhance the brightness of MB owing to the highly-concentrated fluorescence enrichment on only one MB. Therefore, highly sensitive quantification of miRNA has been achieved by measuring the fluorescence signal on MB using a confocal fluorescence microscope. This new strategy exhibits a detection limit of 1.09 pM for let-7a detection, and enables high specificity of distinguishing homologous miRNA family members. SIGNIFICANCE This is the first report by only using one single MB as a carrier to conduct localized CHA, rendering highly-concentrated fluorescence enrichment on only one MB and a dramatic increase in sensitivity. This single MB-based localized CHA strategy has been successfully applied to the accurate analysis of miRNA target in complex biological sample.
Collapse
Affiliation(s)
- Yan Qi
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Jiangtao Yu
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Ming Lou
- Stomatology College of Xi'an Medical University, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Yameng Yu
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Ruohan Li
- Yantai Yongqi Technical Consulting Service Co., Ltd., Yantai, 264003, Shandong Province, PR China
| | - Zhenhao Zhang
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Yuxuan Dai
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Kejing Lao
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Meng Cao
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China
| | - Xingchun Gou
- Institute of Basic and Translational Medicine & Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China; Engineering Research Center of Brain Diseases Drug Development, Universities of Shaanxi Province, Xi'an Medical University, Xi'an, 710021, Shaanxi Province, PR China.
| |
Collapse
|
23
|
He Q, Mi Z, Yin Z, Zheng Z, Guo B. Weighted Gene Networks Derived from Multi-Omics Reveal Core Cancer Genes in Lung Cancer. BIOLOGY 2025; 14:223. [PMID: 40136480 PMCID: PMC11939803 DOI: 10.3390/biology14030223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
Lung cancer remains the leading cause of cancer-related deaths worldwide, driven by its complexity and the heterogeneity of its subtypes, which influence pathogenesis, tumor microenvironment, and genetic alterations. We developed a novel weighted gene regulatory network reconstruction method based on maximum entropy and Markov chain entropy principles, which integrates gene expression and DNA methylation data to generate biologically informed networks. Applied to LUAD and LUSC datasets, we define a network methylation index to determine whether gene methylation acts as oncogenic or tumor-suppressive. By revealing a stable core set of pathogenic genes, we identify not only genes with significant expression changes, such as CD74 and HGF, but also pathogenic genes with stable expression, such as BRAF and KDM6A. Additionally, we uncover potential driver genes, such as CORO2B and C20orf194, associated with disease stage, gender, and smoking status. This method offers a more comprehensive understanding of NSCLC mechanisms, paving the way for improved therapeutic strategies.
Collapse
Affiliation(s)
- Qingcai He
- School of Mathematical Sciences, Beihang University, Beijing 100191, China
- LMIB and SKLCCSE, Beihang University, Beijing 100191, China
- Shen Yuan Honors College, Beihang University, Beijing 100191, China
| | - Zhilong Mi
- LMIB and SKLCCSE, Beihang University, Beijing 100191, China
- Institute of Artificial Intelligence, Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
| | - Ziqiao Yin
- LMIB and SKLCCSE, Beihang University, Beijing 100191, China
- Institute of Artificial Intelligence, Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
- Zhongguancun Laboratory, Beijing 100094, China
| | - Zhiming Zheng
- LMIB and SKLCCSE, Beihang University, Beijing 100191, China
- Institute of Artificial Intelligence, Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
- Zhongguancun Laboratory, Beijing 100094, China
| | - Binghui Guo
- LMIB and SKLCCSE, Beihang University, Beijing 100191, China
- Institute of Artificial Intelligence, Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
- Zhongguancun Laboratory, Beijing 100094, China
| |
Collapse
|
24
|
Tout I, Bougarn S, Toufiq M, Gopinath N, Hussein O, Sathappan A, Chin-Smith E, Rehaman F, Mathew R, Mathew L, Wang K, Liu L, Salhab A, Soloviov O, Tomei S, Hasan W, Da'as S, Bejaoui Y, Hajj NE, Maalej KM, Dermime S, Rasul K, Dellabona P, Casorati G, Turdo A, Todaro M, Stassi G, Ferrone S, Wang X, Maccalli C. The integrative genomic and functional immunological analyses of colorectal cancer initiating cells to modulate stemness properties and the susceptibility to immune responses. J Transl Med 2025; 23:193. [PMID: 39962504 PMCID: PMC11834280 DOI: 10.1186/s12967-025-06176-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) initiating cells (CICs) possess self-renewal capabilities and are pivotal in tumor recurrence and resistance to conventional therapies, including immunotherapy. The mechanisms underlying their interaction with immune cells remain unclear. METHODS We conducted a multi-omics analysis-encompassing DNA methylation, total RNA sequencing, and microRNAs (miRNAs; N = 800) profiling on primary CICs and differentiated tumor cell lines, including autologous pairs. Functional immunological assays were performed to assess the impact of miRNA modulation. RESULTS CICs exhibited distinct methylation patterns, transcriptomic profiles, and miRNA expressions compared to differentiated tumor cells (p < 0.05 or 0.01). Notably, miRNA-15a and -196a were implicated in regulating tumorigenic pathways, such as epithelial-to-mesenchymal transition (EMT), TGF-β signaling, and immune modulation. The transfection of CICs with miRNA mimics led to the downregulation of oncogenic EMT markers (CRKL, lncRNA SOX2-OT, JUNB, SMAD3) and TGF-β pathway, resulting in a significant reduction of the in vitro proliferation and the tumorigenicity and migration in a zebrafish xenograft model. Additionally, miRNA-15a enhanced the expression of antigen processing machinery and decreased the expression of immune checkpoints (PD-L1, PD-L2, CTLA-4) and immunosuppressive cytokines (IL-4). The co-culture of HLA-matched lymphocytes with CICs overexpressing the miRNA-15a, elicited robust tumor-specific immune responses, characterized by a shift toward central and effector memory T cell phenotypes and prevented their terminal differentiation and exhaustion. The combination of miRNA modulation with Indoleamine 2,3-dioxygenase blockade and immunomodulating agents further potentiated these effects. CONCLUSIONS Our study demonstrates that the modulation of miRNA-15a in CICs not only suppresses the tumorigenic properties but also enhances their visibility to the immune system by upregulating antigen presentation and reducing immunomodulatory molecules. These findings suggest that combining miRNA modulation with epigenetic or immunomodulatory agents holds significant promise for overcoming treatment resistance in CRC.
Collapse
Affiliation(s)
- Issam Tout
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Salim Bougarn
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Mohammed Toufiq
- Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Neha Gopinath
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Ola Hussein
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
- College of Pharmacy, Qatar University, Doha, Qatar
| | | | - Evonne Chin-Smith
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Fazulur Rehaman
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Rebecca Mathew
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Lisa Mathew
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Kun Wang
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Li Liu
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Abdulrahman Salhab
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Oleksandr Soloviov
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Sara Tomei
- Integrated Genomics Services, Research Branch, Sidra Medicine, Doha, Qatar
| | - Waseem Hasan
- Zebrafish Functional Genomics Core, Research Department, Sidra Medicine, Doha, Qatar
| | - Sahar Da'as
- Zebrafish Functional Genomics Core, Research Department, Sidra Medicine, Doha, Qatar
- College of Health and Life Science, Hamad Bin Khalifa University, Doha, Qatar
| | - Yosra Bejaoui
- College of Health and Life Science, Hamad Bin Khalifa University, Doha, Qatar
| | - Nady El Hajj
- College of Health and Life Science, Hamad Bin Khalifa University, Doha, Qatar
| | - Karama Makni Maalej
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Hamad Medical Corporation, Doha, Qatar
| | - Kakil Rasul
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Paolo Dellabona
- Experimental Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Maccalli
- Laboratory of Immune Biological Therapy, Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.
- College of Health and Life Science, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
25
|
Zhang J, Fan X, Chen Y, Han Y, Yu W, Zhang S, Yang B, Zhang J, Chen Y. An unfolded protein response (UPR)-signature regulated by the NFKB-miR-29b/c axis fosters tumor aggressiveness and poor survival in bladder cancer. Front Mol Biosci 2025; 12:1542650. [PMID: 40026699 PMCID: PMC11867963 DOI: 10.3389/fmolb.2025.1542650] [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: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Background Bladder cancer continues to pose a substantial global health challenge, marked by a high mortality rate despite advances in treatment options. Therefore, in-depth understanding of molecular mechanisms related to disease onset, progression, and patient survival is of utmost importance in bladder cancer research. Here, we aimed to investigate the underlying mechanisms using a stringent differential expression and survival analyses-based pipeline. Methods Gene and miRNA expression data from TCGA and NCBI GEO databases were analyzed. Differentially expressed genes between normal vs tumor, among tumor aggressiveness groups and between early vs advanced stage were identified using Student's t-test and ANOVA. Kaplan-Meier survival analyses were conducted using R. Functional annotation, miRNA target and transcription factor prediction, network construction, random walk analysis and gene set enrichment analyses were performed using DAVID, miRDIP, TransmiR, Cytoscape, Java and GSEA respectively. Results We identified elevated endoplasmic reticulum (ER) stress response as key culprit, as an eight-gene unfolded protein response (UPR)-related gene signature (UPR-GS) drives aggressive disease and poor survival in bladder cancer patients. This elevated UPR-GS is linked to the downregulation of two miRNAs from the miR-29 family (miR-29b-2-5p and miR-29c-5p), which can limit UPR-driven tumor aggressiveness and improve patient survival. At further upstream, the inflammation-related NFKB transcription factor inhibits miR-29b/c expression, driving UPR-related tumor progression and determining poor survival in bladder cancer patients. Conclusion These findings highlight that the aberrantly activated UPR, regulated by the NFKB-miR-29b/c axis, plays a crucial role in tumor aggressiveness and disease progression in bladder cancer, highlighting potential targets for therapeutic interventions and prognostic markers in bladder cancer management.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Urology, Shangyu People’s Hospital of Shaoxing, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xiaosong Fan
- Department of Urology, Shangyu People’s Hospital of Shaoxing, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yu Chen
- Zhejiang Hisoar Pharmaceutical Co Ltd., Hangzhou, Zhejiang, China
| | - Yichao Han
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weixing Yu
- Department of Urology, Shangyu People’s Hospital of Shaoxing, Shaoxing University, Shaoxing, Zhejiang, China
| | - Shaolin Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College(Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Bicheng Yang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junlong Zhang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanling Chen
- Digestive Endoscopy Center, The First Affiliated Hospital of Wannan Medical College(Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| |
Collapse
|
26
|
Pu W, Shen X, Fan X, Zheng Y, Liu X, Li J, Zhou JK, He J, Wei R, Gong Y, Zheng Q, Luo Y, Guo Y, Ai M, Ming Y, Ye Z, Zhao Y, Wang C, Peng Y. Structure-Guided Optimization and Preclinical Evaluation of 6- O-Benzylguanine-Based Pin1 Inhibitor for Hepatocellular Carcinoma Treatment. J Med Chem 2025; 68:2869-2889. [PMID: 39868498 DOI: 10.1021/acs.jmedchem.4c02144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths globally, and the need for effective systemic therapies for HCC is urgent. Our previous work reveals that Pin1 is a potential anti-HCC target, which regulates miRNA biogenesis and identifies API-1 as a novel Pin1 inhibitor to suppresses HCC. However, a great demand in HCC therapy as well as the limited chemical stability and pharmacokinetic feature of API-1 motivated us to find improved Pin1 inhibitors. Herein, we designed and synthesized diverse 6-O-benzylguanine derivatives and discovered API-32 as a novel Pin1 inhibitor with better stability and pharmacokinetic property over API-1. API-32 directly interacted with the Pin1 PPIase domain to inhibit Pin1 activity. API-32 significantly suppressed the cell proliferation and migration of HCC cells by blocking Pin1's downstream signal. Moreover, API-32 exhibited an enhanced inhibitory function against the HCC tumor in mice models without obvious toxicity, making it a promising drug candidate for HCC treatment.
Collapse
Affiliation(s)
- Wenchen Pu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Xianyan Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xin Fan
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Yuanyuan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Xuesha Liu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jiao Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jian-Kang Zhou
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Juan He
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Rong Wei
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yanqiu Gong
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Qingquan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yao Luo
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yingli Guo
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Min Ai
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Zixia Ye
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yun Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Chun Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
27
|
Li J, Chen M, Zhao W, Lv A, Lin S, Zheng Y, Cai M, Lin N, Xu L, Huang H. The role of miR-129-5p in regulating γ-globin expression and erythropoiesis in β-thalassemia. Hum Mol Genet 2025; 34:291-303. [PMID: 39657657 DOI: 10.1093/hmg/ddae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/04/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
The regulation of γ-globin expression is crucial due to its beneficial effects on diseases like β-thalassemia and sickle cell disease. B-cell lymphoma/leukemia 11A (BCL11A) is a significant suppressor of γ-globin, and microRNAs (miRNAs) targeting BCL11A have been shown to alleviate this suppression. In our previous high-throughput sequencing, we identified an 11.32-fold increase in miR-129-5p expression in β-thalassemia patients. However, the regulatory mechanisms of miR-129-5p in the context of erythroid differentiation remain to be elucidated. Our study aimed to elucidate the role of miR-129-5p in γ-globin regulation and erythropoiesis. We measured miR-129-5p levels in peripheral blood from β-thalassemia major and intermedia patients. Fluorescence in situ hybridization, dual-luciferase reporter assays, miRNA pull down assays and western blot analyses were conducted to examine the effects of miR-129-5p on γ-globin expression and BCL11A repression. Cell proliferation, apoptosis, and erythroid differentiation were assessed using cell counting kit-8, Wright-Giemsa, and benzidine staining, and flow cytometry assays. The expression levels of miR-129-5p were significantly elevated in β-thalassemia patients and positively correlated with γ-globin synthesis while negatively correlating with liver damage. miR-129- 5p enhanced γ-globin gene expression in K562 and HUDEP-2 cells by effectively repressing BCL11A. Overexpression of miR-129-5p inhibited cell proliferation, induced cell cycle arrest at the G1/G0 phase, promoted apoptosis and stimulated erythroid differentiation and maturation. Conversely, inhibition of miR-129-5p produced opposite cellular effects. miR-129-5p acts as a positive regulator of erythroid differentiation and γ-globin synthesis. It offers a promising miRNA target for activating the γ-globin gene and reducing ineffective erythropoiesis in β-thalassemia patients.
Collapse
Affiliation(s)
- Jingmin Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Meihuan Chen
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Wantong Zhao
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Aixiang Lv
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Siyang Lin
- The School of Medical Technology and Engineering, Fujian Medical University, 1 Xuefu North Road, Minhou District, Fuzhou 350108, China
| | - Yanping Zheng
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Meiying Cai
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Na Lin
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Hailong Huang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| |
Collapse
|
28
|
Zhand S, Goss DM, Cheng YY, Warkiani ME. Recent Advances in Microfluidics for Nucleic Acid Analysis of Small Extracellular Vesicles in Cancer. Adv Healthc Mater 2025; 14:e2401295. [PMID: 39707658 DOI: 10.1002/adhm.202401295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/28/2024] [Indexed: 12/23/2024]
Abstract
Small extracellular vesicles (sEVs) are membranous vesicles released from cellular structures through plasma membrane budding. These vesicles contain cellular components such as proteins, lipids, mRNAs, microRNAs, long-noncoding RNA, circular RNA, and double-stranded DNA, originating from the cells they are shed from. Ranging in size from ≈25 to 300 nm and play critical roles in facilitating cell-to-cell communication by transporting signaling molecules. The discovery of sEVs in bodily fluids and their involvement in intercellular communication has revolutionized the fields of diagnosis, prognosis, and treatment, particularly in diseases like cancer. Conventional methods for isolating and analyzing sEVs, particularly their nucleic acid content face challenges including high costs, low purity, time-consuming processes, limited standardization, and inconsistent yield. The development of microfluidic devices, enables improved precision in sorting, isolating, and molecular-level separation using small sample volumes, and offers significant potential for the enhanced detection and monitoring of sEVs associated with cancer. These advanced techniques hold great promise for creating next-generation diagnostic and prognostic tools given their possibility of being cost-effective, simple to operate, etc. This comprehensive review explores the current state of research on microfluidic devices for the detection of sEV-derived nucleic acids as biomarkers and their translation into practical point-of-care and clinical applications.
Collapse
Affiliation(s)
- Sareh Zhand
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dale Mark Goss
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Theranostics, Sechenov First Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
29
|
Valenzuela G, Contreras HR, Marcelain K, Burotto M, González-Montero J. Understanding microRNA-Mediated Chemoresistance in Colorectal Cancer Treatment. Int J Mol Sci 2025; 26:1168. [PMID: 39940936 PMCID: PMC11818086 DOI: 10.3390/ijms26031168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Colorectal cancer (CRC) remains the second most lethal cancer worldwide, with incidence rates expected to rise substantially by 2040. Although biomarker-driven therapies have improved treatment, responses to standard chemotherapeutics, such as 5-fluorouracil (5-FU), oxaliplatin, and irinotecan, vary considerably. This clinical heterogeneity emphasizes the urgent need for novel biomarkers that can guide therapeutic decisions and overcome chemoresistance. microRNAs (miRNAs) have emerged as key post-transcriptional regulators that critically influence chemotherapy responses. miRNAs orchestrate post-transcriptional gene regulation and modulate diverse pathways linked to chemoresistance. They influence drug transport by regulating ABC transporters and affect metabolic enzymes like thymidylate synthase (TYMS). These activities shape responses to standard CRC chemotherapy agents. Furthermore, miRNAs can regulate the epithelial-mesenchymal transition (EMT). The miR-200 family (e.g., miR-200c and miR-141) can reverse EMT phenotypes, restoring chemosensitivity. Additionally, miRNAs like miR-19a and miR-625-3p show predictive value for chemotherapy outcomes. Despite these promising findings, the clinical translation of miRNA-based biomarkers faces challenges, including methodological inconsistencies and the dynamic nature of miRNA expression, influenced by the tumor microenvironment. This review highlights the critical role of miRNAs in elucidating chemoresistance mechanisms and their promise as biomarkers and therapeutic targets in CRC, paving the way for a new era of precision oncology.
Collapse
Affiliation(s)
- Guillermo Valenzuela
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (G.V.); (H.R.C.); (K.M.)
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| | - Héctor R. Contreras
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (G.V.); (H.R.C.); (K.M.)
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| | - Katherine Marcelain
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (G.V.); (H.R.C.); (K.M.)
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| | - Mauricio Burotto
- Bradford Hill Clinical Research Center, Santiago 8380453, Chile;
| | - Jaime González-Montero
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (G.V.); (H.R.C.); (K.M.)
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
- Bradford Hill Clinical Research Center, Santiago 8380453, Chile;
| |
Collapse
|
30
|
Wilhelm ED, Dankert JT, Wiesehöfer M, Wach S, Wagner M, Spahn M, Kruithof-de Julio M, Wennemuth G. xCT as a potential marker for neuroendocrine cells in high-risk prostate cancer and the relation to AL122023.1-miR-26a/30d/30e axis. PLoS One 2025; 20:e0318213. [PMID: 39869598 PMCID: PMC11771886 DOI: 10.1371/journal.pone.0318213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/10/2025] [Indexed: 01/29/2025] Open
Abstract
Prostate cancer is the second most common type of cancer in male worldwide. Stromal-epithelial interaction is thought to have a major impact on cancer development and progression. Previous studies have shown that interaction via soluble factors lead to a reduction in the expression of xCT and AL122023.1 in the prostate carcinoma cell line LNCaP after seven days of co-culture with primary stromal p21 cells. In this study, we validated the repression of xCT and AL122023.1 at RNA level using quantitative real-time PCR and at protein level by Western Blotting. Furthermore, xCT is known to be a putative target for miRNAs miR-26a, miR-30d and miR-30e, which in turn potentially interact with AL122023.1. The lncRNA-miRNA-interaction was verified by luciferase reporter assays. However, miR-26a/-30d/-30e did not inhibit xCT expression at protein level. Nevertheless, indirect inhibitory effect of AL122023.1 on the xCT expression could be shown. Moreover, immunostaining revealed precise xCT expression in neuroendocrine cells, ranging from fetal, healthy juvenile, and adult prostate tissue to benign prostatic hyperplasia and finally advanced prostate cancer. This study explores the relevance and function of xCT and AL122023.1 in the prostate and exposes xCT as a potential marker or therapeutic target in high-risk prostate cancer.
Collapse
Affiliation(s)
- Elena D. Wilhelm
- Department of Anatomy, University Hospital Essen, Essen, Germany
| | | | - Marc Wiesehöfer
- Department of Anatomy, University Hospital Essen, Essen, Germany
| | - Sven Wach
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Mathias Wagner
- Department of General and Special Pathology, University Hospital Saarland, Homburg, Germany
| | - Martin Spahn
- Department of Urology, Lindenhofspital Bern, Bern, Switzerland
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, University Hospital Bern, Bern, Switzerland
| | | |
Collapse
|
31
|
Miyoshi J, Mannucci A, Scarpa M, Gao F, Toden S, Whitsett T, Inge LJ, Bremner RM, Takayama T, Cheng Y, Bottiglieri T, Nagtegaal ID, Shrubsole MJ, Zaidi AH, Wang X, Coleman HG, Anderson LA, Meltzer SJ, Goel A. Liquid biopsy to identify Barrett's oesophagus, dysplasia and oesophageal adenocarcinoma: the EMERALD multicentre study. Gut 2025; 74:169-181. [PMID: 39562048 PMCID: PMC11869464 DOI: 10.1136/gutjnl-2024-333364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND There is no clinically relevant serological marker for the early detection of oesophageal adenocarcinoma (EAC) and its precursor lesion, Barrett's oesophagus (BE). OBJECTIVE To develop and test a blood-based assay for EAC and BE. DESIGN Oesophageal MicroRNAs of BaRRett, Adenocarcinoma and Dysplasia (EMERALD) was a large, international, multicentre biomarker cohort study involving 792 patient samples from 4 countries (NCT06381583) to develop and validate a circulating miRNA signature for the early detection of EAC and high-risk BE. Tissue-based miRNA sequencing and microarray datasets (n=134) were used to identify candidate miRNAs of diagnostic potential, followed by validation using 42 pairs of matched cancer and normal tissues. The usefulness of the candidate miRNAs was initially assessed using 108 sera (44 EAC, 34 EAC precursors and 30 non-disease controls). We finally trained a machine learning model (XGBoost+AdaBoost) on RT-qPCR results from circulating miRNAs from a training cohort (n=160) and independently tested it in an external cohort (n=295). RESULTS After a strict process of biomarker discovery and selection, we identified six miRNAs that were overexpressed in all sera of patients compared with non-disease controls from three independent cohorts of different nationalities (miR-106b, miR-146a, miR-15a, miR-18a, miR-21 and miR-93). We established a six-miRNA diagnostic signature using the training cohort (area under the receiver operating characteristic curve (AUROC): 97.6%) and tested it in an independent cohort (AUROC: 91.9%). This assay could also identify patients with BE among patients with gastro-oesophageal reflux disease (AUROC: 94.8%, sensitivity: 92.8%, specificity: 85.1%). CONCLUSION Using a comprehensive approach integrating unbiased genome-wide biomarker discovery and several independent experimental validations, we have developed and validated a novel blood test that might complement screening options for BE/EAC. TRIAL REGISTRATION NUMBER NCT06381583.
Collapse
Affiliation(s)
- Jinsei Miyoshi
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Gastroenterology, Kawashima Hospital, Tokushima, Japan
| | - Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Scarpa
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padova, Italy
| | - Feng Gao
- Sun Yat-Sen University, The Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shusuke Toden
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Timothy Whitsett
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Landon J Inge
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yulan Cheng
- Division of Gastroenterology and Hepatology, Department Of Medicine And Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teodoro Bottiglieri
- Baylor Scott & White Research Institute, Institute of Metabolic Diseases, Dallas, TX, USA
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Martha J Shrubsole
- Department of Medicine, Division of Epidemiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ali H Zaidi
- Esophageal and Thoracic Research Laboratories, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Helen G Coleman
- Cancer Epidemiology Research Group, Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Lesley A Anderson
- Centre for Health Data Science, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Stephen J Meltzer
- Division of Gastroenterology and Hepatology, Department Of Medicine And Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ajay Goel
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
32
|
Jothimani G, Das D, Pathak S, Malayaperumal S, Zhang H, Sun XF, Banerjee A. Unraveling the mechanism of microRNA-134 in colon cancer progression: Targeting KRAS and PIK3CA for cell cycle control and histone deacetylase regulation. Exp Cell Res 2025; 444:114385. [PMID: 39746598 DOI: 10.1016/j.yexcr.2024.114385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
Colon cancer is the leading cause of cancer-related deaths worldwide. MicroRNAs (miRNAs) are key regulators of gene expression, often dysregulated in colon cancer. This study aims to elucidate the therapeutic role of miR-134-5p as a tumor suppressor miRNA in colon cancer cells. We analyzed miRNA expression profiles in primary and metastatic colon cancer cells. The clinical significance of miR-134-5p was evaluated using the TCGA database. Bioinformatics tools (HADDOCK) predicted miRNA-mRNA interactions and the molecular docking of miRNA-mRNA-AGO2 complexes. Luciferase reporter assays, cell proliferation, immunofluorescence, colony forming unit assays, and qRT-PCR analysis assessed miR-134-5p effects on KRAS, PIK3CA, and downstream signaling pathways in primary and metastatic colon cancer cells. miR-134-5p was downregulated in colon cancer cells. Bioinformatics analysis suggested KRAS, PIK3CA, EGFR, and HDAC5 as potential targets. HADDOCK analysis revealed strong binding affinity and structural stability between KRAS, PIK3CA, miR-134-5p, and AGO2. Gene-reporter assays confirmed miR-134-5p-mediated degradation of KRAS and PIK3CA. miR-134-5p transfection reduced KRAS and PI3K protein levels, suppressed EGFR/RTK signaling and its downstream targets, and inhibited HDAC expression, ultimately reducing colon cancer cell proliferation. The results of this study confirm that miR-134-5p acts as a potential tumor suppressor miRNA in colon cancer cells by inhibiting KRAS and PI3K expression through AGO2-mediated gene silencing. It deregulates downstream EGFR signaling and HDACs, thereby reducing colon cancer cell proliferation. These findings highlight miR-134-5p as a promising therapeutic target for miRNA-mediated anticancer therapy.
Collapse
Affiliation(s)
- Ganesan Jothimani
- Medical Biotechnology lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Rajiv Gandhi Salai (OMR), Kelambakkam, Chennai, Tamil Nadu, 603 103, India
| | - Diptimayee Das
- Medical Biotechnology lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Rajiv Gandhi Salai (OMR), Kelambakkam, Chennai, Tamil Nadu, 603 103, India
| | - Surajit Pathak
- Medical Biotechnology lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Rajiv Gandhi Salai (OMR), Kelambakkam, Chennai, Tamil Nadu, 603 103, India
| | - Sarubala Malayaperumal
- Medical Biotechnology lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Rajiv Gandhi Salai (OMR), Kelambakkam, Chennai, Tamil Nadu, 603 103, India
| | - Hong Zhang
- Department of Medical Sciences, School of Medicine, Orebro University, SE-701 82, Orebro, Sweden
| | - Xiao-Feng Sun
- Division of Oncology, Department of Biomedical and Clinical Sciences, O-Huset, Sjukhusvägen, Linköping University, SE-581 83, Linköping, Sweden
| | - Antara Banerjee
- Medical Biotechnology lab, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Rajiv Gandhi Salai (OMR), Kelambakkam, Chennai, Tamil Nadu, 603 103, India.
| |
Collapse
|
33
|
Zhao N, Xiong Q, Li P, Chen G, Xiao H, Wu C. TSC complex decrease the expression of mTOR by regulated miR-199b-3p. Sci Rep 2025; 15:1892. [PMID: 39806027 PMCID: PMC11730325 DOI: 10.1038/s41598-025-85706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
The TSC complex formed by TSC1 and TSC2 is the most important upstream negative regulator of mTORC1. Genetic variations in either TSC1 or TSC2 cause tuberous sclerosis complex (TSC) disease which is a rare autosomal dominant disorder resulting in impairment of multiple organ systems. In this study, besides a reported variation, c.2509_2512del (p.Asn837Valfs*11, p.N837fs) in TSC1, we found a de novo TSC2 variation c.1113delG (p.Gln371Hisfs*18, p.Q371fs), which these two mutation influence the formation of TSC complex. We found that the decrease of TSC complex with the appearance of the decreased miR-199b-3p expression. At the same time, the reduction of miR-199b-3p increased the expression of mTOR and the activation of mTORC1 and mTORC2, the additional miR-199b-3p caused the decrease the expression of mTOR and the activation of mTORC1 and mTORC2. In brief, our results may illustrate a novel mechanism of TSC caused by variations in either TSC1 or TSC2, and a new mTOR expression regulator, miR-199b-3p.
Collapse
Affiliation(s)
- Na Zhao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
- Department of Pathology, The Second Hospital of ShanXi Medical University, No.382 WuYi Road, Tai Yuan, ShanXi, Taiyuan, 030000, China
| | - Qiuhong Xiong
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Ping Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Guangxin Chen
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Han Xiao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Changxin Wu
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
34
|
Derogar R, Nejadi Orang F, Abdoli Shadbad M. Competing endogenous RNA networks in ovarian cancer: from bench to bedside. EXCLI JOURNAL 2025; 24:86-112. [PMID: 39967908 PMCID: PMC11830916 DOI: 10.17179/excli2024-7827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Epithelial ovarian cancer is responsible for the majority of ovarian malignancies, and its highly invasive nature and chemoresistant development have been major obstacles to treating patients with mainstream treatments. In recent decades, the significance of microRNAs (miRNAs), circular RNAs (circRNAs), long non-coding RNAs (lncRNAs), and competing endogenous RNAs (ceRNAs) has been highlighted in ovarian cancer development. This hidden language between these RNAs has led to the discovery of enormous regulatory networks in ovarian cancer cells that substantially affect gene expression. Aside from providing ample opportunities for targeted therapies, circRNA- and lncRNA-mediated ceRNA network components provide invaluable biomarkers. The current study provides a comprehensive and up-to-date review of the recent findings on the significance of these ceRNA networks in the hallmarks of ovarian cancer oncogenesis, treatment, diagnosis, and prognosis. Also, it provides the authorship with future perspectives in the era of single-cell RNA sequencing and personalized medicine.
Collapse
Affiliation(s)
- Roghaiyeh Derogar
- Fellowship in Gynecologic Oncology, Department of Gynecology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | | | - Mahdi Abdoli Shadbad
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Wang Z, Jiang L, Bai X, Guo M, Zhou R, Zhou Q, Yang H, Qian J. Vitamin D receptor regulates methyltransferase like 14 to mitigate colitis-associated colorectal cancer. J Genet Genomics 2025:S1673-8527(25)00002-5. [PMID: 39778713 DOI: 10.1016/j.jgg.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Colitis-associated colorectal cancer (CAC), a serious complication of ulcerative colitis (UC), is associated with a poor prognosis. The vitamin D receptor (VDR) is recognized for its protective role in UC and CAC through the maintenance of intestinal barrier integrity and the regulation of inflammation. This study demonstrates a significant reduction in m6A-related genes, particularly methyltransferase like 14 (METTL14), in UC and CAC patients and identifies an association between METTL14 and VDR. In the azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced mouse model, vitamin D treatment increases METTL14 expression and reduces tumor burden, while Vdr-knockout mice exhibit lower METTL14 levels and increased tumorigenesis. In vitro, the VDR agonist calcipotriol upregulates METTL14 in NCM460 cells, with this effect attenuated by VDR knockdown. VDR knockdown in DLD-1 colon cancer cells decreases METTL14 expression and promotes proliferation, which is reversed by METTL14 overexpression. Mechanistic studies reveal that VDR regulates METTL14 expression via promoter binding, modulating key target genes such as SOX4, DROSH, and PHLPP2. This study highlights the role of the VDR-METTL14 axis as a protective mechanism in CAC and suggests its potential as a therapeutic target for preventing and treating CAC.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Lingjuan Jiang
- Biomarker Discovery and Validation Facility, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoyin Bai
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Mingyue Guo
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Runing Zhou
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qingyang Zhou
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
36
|
Liu Q, Archilla I, Lopez‐Prades S, Torres F, Camps J, Cuatrecasas M. Polymorphic Single-Nucleotide Variants in miRNA Genes and the Susceptibility to Colorectal Cancer: Combined Evaluation by Pairwise and Network Meta-Analysis, Thakkinstian's Algorithm and FPRP Criterium. Cancer Med 2025; 14:e70621. [PMID: 39840720 PMCID: PMC11751872 DOI: 10.1002/cam4.70621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Considerable epidemiological studies have examined the correlation between polymorphic single-nucleotide variants (SNPs) in miRNA genes and colorectal carcinoma (CRC) risk, yielding inconsistent results. Herein, we sought to systematically investigate the association between miRNA-SNPs and CRC susceptibility by combined evaluation using pairwise and network meta-analysis, the FPRP analysis (false positive report probability), and the Thakkinstian's algorithm. METHODS The MEDLINE, EMBASE, WOS, and Cochrane Library databases were searched through May 2024 to find relevant association literatures. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were computed by the pairwise meta-analysis. Network meta-analysis and the Thakkinstian's method were applied for determining the potentially optimal genetic models; additionally, the FPRP was used to identify noteworthy associations. RESULTS Totally, 39 case-control trials involving 18,028 CRC cases, and 21,816 normal participants were included in the study. Eleven SNPs within nine genes were examined for their predisposition to CRC. miR-27a (rs895819) was found to significantly increase CRC risk among overall population (OR 1.58, 95% CI: 1.32-1.89) and Asians (OR 1.62, 95% CI: 1.31-2.01), with the recessive models identified as the optimal models. Furthermore, miR-196a2 (rs11614913), miR-143/145 (rs41291957), and miR-34b/c (rs4938723) were significantly related to reduced CRC risk among Asian descendants under the optimal dominant (OR 0.75, 95% CI: 0.65-0.86), recessive (OR 0.72, 95% CI: 0.60-0.85), and recessive models (OR 0.69, 95% CI: 0.56-0.85), respectively. The results were also proposed by the network meta-analysis or the Thakkinstian's method and confirmed by the FPRP criterion. CONCLUSION The miR-27a (rs895819) is correlated with elevated CRC risk among overall population and Asians, and the recessive model is found to be optimal for predicting CRC risk. Additionally, the miR-196a2 (rs11614913), miR-143/145 (rs41291957), and miR-34b/c (rs4938723), with the dominant, recessive, and recessive models identified as the optimal, might confer protective effects against CRC among Asians.
Collapse
Affiliation(s)
- Qing Liu
- Faculty of Medicine and Health Sciences, Doctoral SchoolUniversity of BarcelonaBarcelonaSpain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
| | - Ivan Archilla
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
- Pathology Department, Centre of Biomedical Diagnosis (CDB)Hospital Clinic, University of BarcelonaBarcelonaSpain
| | - Sandra Lopez‐Prades
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
- Pathology Department, Centre of Biomedical Diagnosis (CDB)Hospital Clinic, University of BarcelonaBarcelonaSpain
| | - Ferran Torres
- Department of BiostatisticsAutonomous University of Barcelona (UAB)BellaterraSpain
| | - Jordi Camps
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
- Cell Biology and Medical Genetics Unit, Department of Cell Biology, Physiology and Immunology, Faculty of MedicineAutonomous University of Barcelona (UAB)BellaterraSpain
| | - Miriam Cuatrecasas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS)BarcelonaSpain
- Pathology Department, Centre of Biomedical Diagnosis (CDB)Hospital Clinic, University of BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
- Faculty of Medicine and Health Sciences, Department of Clinical FoundationsUniversity of Barcelona (UB)BarcelonaSpain
| |
Collapse
|
37
|
Shi X, Ji Y, Wu X, Du Y, Yan X, Wang Y, Xia X. Blocking of SIRT7/FOXO3a axis by miR-152-3p enhances cisplatin sensitivity in breast cancer. Am J Med Sci 2025; 369:105-115. [PMID: 39241827 DOI: 10.1016/j.amjms.2024.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/23/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Cisplatin-based chemoresistance is a major obstacle for the treatment breast cancer (BC) including Triple-negative breast cancer (TNBC). SIRT7 is reportedly involved in the progression of BC, the underlining mechanism in Cisplatin-based chemoresistance in BC remains unclear. This work is to elucidate effects of SIRT7 on cisplatin resistance in breast cancer regulated by miR-152-3p. METHODS The RNA expression of SIRT7 and miRNAs in breast cancer were available from TCGA database. SIRT7-targeted miRNAs were predicted by TargetScan, miRanda, miRDB databases. The association of SIRT7 expression with predicted miRNA was validated by Luciferase assay. Cell apoptosis was determined by Flow cytometry. Cell viability was detected by CCK8 assay. The mRNA expression was measured by quantitative real-time polymerase chain reaction (qRT-PCR) assay. Protein expression was determined by Western blotting assay. RESULTS SIRT7 mRNA levels were dramatically enhanced in BC tissues compared to para-carcinoma tissues, also increased in BC patients with Cisplatin-based chemotherapy containing TNBC compared with those without. The increase of SIRT7 expression was obviously relevant to shorter survival time of them. Importantly, SIRT7 inhibition facilitated Cisplatin-induced cell apoptosis of TNBC (MDA-MB-231 and MDA-MB-468) and non- TNBC (MCF-7). Notably, miR-152-3p was predicted as a negative regulator of SIRT7 by overlapping downregulated miRNAs in BC patients treated with Cisplatin-based chemotherapy and miRNAs to target SIRT7. Mechanically, miR-152-3p blocked SIRT7 to stimulate an activation of FOXO3a, cleaved PARP1 and Caspase-3, sensitizing Cisplatin-induced apoptosis of BC cells. CONCLUSIONS Inhibition of SIRT7 by miR-152-3p may be a promising strategy against the resistance to cisplatin-based chemotherapy in TNBC.
Collapse
Affiliation(s)
- Xiangkui Shi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmacy, the Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yunfei Ji
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xueqing Wu
- Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yu Du
- Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaonan Yan
- Clinical Center of Reproductive Medicine, Xuzhou Central Hospital, Xuzhou, China; Clinical Center of Reproductive Medicine, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou 221000, China.
| | - Yan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmacy, the Affiliated Xuzhou Maternity and Child Health Care Hospital of Xuzhou Medical University, Xuzhou, China; Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Xiaobing Xia
- Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
38
|
Tan W, Xiao C, Ma M, Cao Y, Huang Z, Wang X, Kang R, Li Z, Li E. Role of non-coding RNA in lineage plasticity of prostate cancer. Cancer Gene Ther 2025; 32:1-10. [PMID: 39496938 DOI: 10.1038/s41417-024-00834-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 11/06/2024]
Abstract
The treatment of prostate cancer (PCa) has made great progress in recent years, but treatment resistance always develops and can even lead to fatal disease. Exploring the mechanism of drug resistance is of great significance for improving treatment outcomes and developing biomarkers with predictive value. It is increasingly recognized that mechanism of drug resistance in advanced PCa is related to lineage plasticity and tissue differentiation. Specifically, one of the mechanisms by which castration-resistant prostate cancer (CRPC) cells acquire drug resistance and transform into neuroendocrine prostate cancer (NEPC) cells is lineage plasticity. NEPC is a subtype of PCa that is highly aggressive and lethal, with a median survival of only 7 months. With the development of high-throughput RNA sequencing technology, more and more non-coding RNAs have been identified, which play important roles in different diseases through different mechanisms. Several ncRNAs have shown great potential in PCa lineage plasticity and as biomarkers. In the review, the role of ncRNA in PCa lineage plasticity and its use as biomarkers were reviewed.
Collapse
Affiliation(s)
- Wenhui Tan
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Changkai Xiao
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Min Ma
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Youhan Cao
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhenguo Huang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaolan Wang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ran Kang
- Department of Urology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhenfa Li
- Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, 421001, Hunan, China.
| | - Ermao Li
- Institute of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
39
|
Madsen S, Peluso AA, Yonamine CY, Ingerslev LR, Dall M, Petersen PSS, Plucinska K, Pradas-Juni M, Moreno-Justicia R, Gonzalez-Franquesa A, Højlund K, Kornfeld JW, Emanuelli B, Vienberg SG, Treebak JT. Rapid downregulation of DICER is a hallmark of adipose tissue upon high-fat diet feeding. Mol Cell Endocrinol 2025; 595:112413. [PMID: 39536934 DOI: 10.1016/j.mce.2024.112413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/14/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Adipose tissue regulates whole-body energy balance and is crucial for metabolic health. With energy surplus, adipose tissue expands, which may lead to local areas of hypoxia and inflammation, and consequently impair whole-body insulin sensitivity. We report that DICER, a key enzyme for miRNA maturation, is significantly lower in abdominal subcutaneous white adipose tissue of men with obesity compared with men with a lean phenotype. Furthermore, DICER is profoundly downregulated in mouse adipose tissue and liver within the first week on a high-fat diet (HFD), and remains low after prolonged HFD feeding. Downregulation of DICER in mice occurs in both mature adipocytes and stromal vascular cells. Mechanistically, chemically induced hypoxia in vitro shows DICER degradation via interaction with hypoxia-inducible factor 1-α (HIF1α). Moreover, DICER and HIF1α interact in brown adipose tissue post-HFD which may signal for DICER degradation. Finally, RNA sequencing reveals a striking time-dependent downregulation of total miRNA content in mouse subcutaneous adipose tissue after HFD feeding. Collectively, HFD in mice reduces adipose tissue DICER, likely due to hypoxia-induced interaction with HIF1α during tissue expansion, and this significantly impacts miRNA content.
Collapse
Affiliation(s)
- Søren Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - A Augusto Peluso
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars R Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Dall
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patricia S S Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kaja Plucinska
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marta Pradas-Juni
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alba Gonzalez-Franquesa
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Jan-Wilhelm Kornfeld
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark; Novo Nordisk Foundation Center for Adipocyte Signaling (Adiposign), University of Southern Denmark, Odense, Denmark
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara G Vienberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
Afsar S, Syed RU, Khojali WMA, Masood N, Osman ME, Jyothi JS, Hadi MA, Khalifa AAS, Aboshouk NAM, Alsaikhan HA, Alafnan AS, Alrashidi BA. Non-coding RNAs in BRAF-mutant melanoma: targets, indicators, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:297-317. [PMID: 39167168 DOI: 10.1007/s00210-024-03366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Melanoma, a highly aggressive skin cancer, is often driven by BRAF mutations, such as the V600E mutation, which promotes cancer growth through the MAPK pathway and contributes to treatment resistance. Understanding the role of non-coding RNAs (ncRNAs) in these processes is crucial for developing new therapeutic strategies. This review aims to elucidate the relationship between ncRNAs and BRAF mutations in melanoma, focusing on their regulatory roles and impact on treatment resistance. We comprehensively reviewed current literature to synthesize evidence on ncRNA-mediated regulation of BRAF-mutant melanoma and their influence on therapeutic responses. Key ncRNAs, including microRNAs and long ncRNAs, were identified as significant regulators of melanoma development and therapy resistance. MicroRNAs such as miR-15/16 and miR-200 families modulate critical pathways like Wnt signaling and melanogenesis. Long ncRNAs like ANRIL and SAMMSON play roles in cell growth, invasion, and drug susceptibility. Specific ncRNAs, such as BANCR and RMEL3, intersect with the MAPK pathway, highlighting their potential as therapeutic targets or biomarkers in BRAF-mutant melanoma. Additionally, ncRNAs involved in drug resistance, such as miR-579-3p and miR-1246, target processes like autophagy and immune checkpoint regulation. This review highlights the pivotal roles of ncRNAs in regulating BRAF-mutant melanoma and their contribution to drug resistance. These findings underscore the potential of ncRNAs as biomarkers and therapeutic targets, paving the way for innovative treatments to improve outcomes for melanoma patients.
Collapse
Affiliation(s)
- S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh, 517502, India.
| | - Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, 81442, Hail, Saudi Arabia.
| | - Weam M A Khojali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, 14415, Sudan
| | - Najat Masood
- Chemistry Department, Faculty of Science, University of Ha'il, P.O. Box 2440, 81451, Ha'il,, Saudi Arabia
| | - Mhdia Elhadi Osman
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - J Siva Jyothi
- Department of Pharmaceutics, Hindu College of Pharmacy, Andhra Pradesh, India
| | - Mohd Abdul Hadi
- Department of Pharmaceutics, Bhaskar Pharmacy College, Moinabad, R.R.District, Hyderabad, 500075, Telangana, India
| | - Amna Abakar Suleiman Khalifa
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 81442, Hail, Saudi Arabia
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 81442, Hail, Saudi Arabia
| | | | | | | |
Collapse
|
41
|
Gong Z, Yuan P, Gan Y, Long X, Deng Z, Tang Y, Yang Y, Zhong S. A one-pot isothermal Fluorogenic Mango II arrays-based assay for label-free detection of miRNA. Talanta 2025; 281:126920. [PMID: 39306943 DOI: 10.1016/j.talanta.2024.126920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
The capability to detect a small number of miRNAs in clinical samples with simplicity, selectivity, and sensitivity is immensely valuable, yet it remains a daunting task. Here, we described a novel Mango II aptamers-based sensor for the one-pot, sensitive and specific detection of miRNAs. Target miRNA-initiated mediated catalyzed hairpin assembly (CHA) would allow for the production of plenty of DNA duplexes and the formation of the complete T7 promoter, motivating the rolling circle transcription (RCT). Then, the subsequent RCT process efficiently generates a huge number of repeating RNA Mango II aptamers, brightened by the incorporation of fluorescent dye TO1-B for miRNA quantification, realizing label-free and high signal-to-background ratio. Moreover, this assay possesses a remarkable ability to confer high selectivity, enabling the distinction of miRNAs among family members with mere 1- or 2- nucleotide (nt) differences. By employing the proposed assay, we have successfully achieved a sensitive evaluation of miR-21 content in diverse cell lines and clinical serum samples. This offers a versatile approach for the sensitive assay of miRNA biomarkers in molecular diagnosis.
Collapse
Affiliation(s)
- Zan Gong
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Panpan Yuan
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Yuqing Gan
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Xi Long
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Zhiwei Deng
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Yalan Tang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China
| | - Yanjing Yang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China.
| | - Shian Zhong
- College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, China; Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, the "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, 410219, China.
| |
Collapse
|
42
|
Liu SP, Li YF, Zhang D, Li CY, Dai XF, Lan DF, Cai J, Zhou H, Song T, Zhao YY, He ZX, Tan J, Zhang JD. Pharmacological actions of the bioactive compounds of Epimedium on the male reproductive system: current status and future perspective. Asian J Androl 2025; 27:20-29. [PMID: 38978290 PMCID: PMC11784961 DOI: 10.4103/aja20248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/26/2024] [Indexed: 07/10/2024] Open
Abstract
ABSTRACT Compounds isolated from Epimedium include the total flavonoids of Epimedium , icariin, and its metabolites (icaritin, icariside I, and icariside II), which have similar molecular structures. Modern pharmacological research and clinical practice have proved that Epimedium and its active components have a wide range of pharmacological effects, especially in improving sexual function, hormone regulation, anti-osteoporosis, immune function regulation, anti-oxidation, and anti-tumor activity. To date, we still need a comprehensive source of knowledge about the pharmacological effects of Epimedium and its bioactive compounds on the male reproductive system. However, their actions in other tissues have been reviewed in recent years. This review critically focuses on the Epimedium , its bioactive compounds, and the biochemical and molecular mechanisms that modulate vital pathways associated with the male reproductive system. Such intrinsic knowledge will significantly further studies on the Epimedium and its bioactive compounds that protect the male reproductive system and provide some guidances for clinical treatment of related male reproductive disorders.
Collapse
Affiliation(s)
- Song-Po Liu
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Medical Genetics, Zunyi Medical University, Zunyi 563000, China
| | - Yun-Fei Li
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi 563000, China
| | - Dan Zhang
- Zunyi Medical University Library, Zunyi 563000, China
| | - Chun-Yang Li
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Xiao-Fang Dai
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Dong-Feng Lan
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Ji Cai
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - He Zhou
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Yan-Yu Zhao
- Department of Medical Genetics, Zunyi Medical University, Zunyi 563000, China
| | - Zhi-Xu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Jun Tan
- Department of Histology and Embryology, Zunyi Medical University, Zunyi 563000, China
| | - Ji-Dong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
43
|
Ciobanu OA, Herlea V, Milanesi E, Dobre M, Fica S. miRNA profile in pancreatic neuroendocrine tumors: Preliminary results. Sci Prog 2025; 108:368504251326864. [PMID: 40152231 PMCID: PMC11952036 DOI: 10.1177/00368504251326864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Our understanding of the pathophysiology of pancreatic neuroendocrine tumors (PanNETs) remains incomplete, largely due to their historically underestimated incidence and the perception of these tumors as rare and slow-growing cancers. Additionally, conventional reliance on histological examination alone is gradually being supplemented by the exploration and introduction of molecular biomarkers, such as microRNAs (miRNAs). As miRNAs modulate the expression of multiple genes and pathways involved in the tumorigenesis of PanNETs, these biomarkers hold considerable promise for diagnosis and prognosis applications. In this study, we aimed to identify miRNAs as tissue markers associated with the diagnosis of PanNETs. METHODS We conducted a case-control study including: 7 PanNETs and 19 nontumoral pancreatic tissues obtained from Romanian patients. The samples underwent miRNA profiling via quantitative RT-PCR to assess the expression of 84 miRNAs. Our results were compared with those obtained by reanalyzing a public dataset. Furthermore, we structured our miRNA expression data according to their targeted mRNAs and their roles in signaling pathways. RESULTS Fourteen miRNAs (miR-1, miR-133a-3p, miR-210-3p, miR-7-5p, miR-10a-5p, miR-92b-3p, miR-132-3p, miR-221-3p, miR-29b-3p, miR-107, miR-103a-3p, let-7b-5p, miR-148a-3p, and miR-202-3p) were identified as differentially expressed by comparing PanNETs with pancreatic nontumoral tissues, with six miRNAs (miR-7-5p, miR-92b-3p, miR-29b-3p, miR-107, miR-103a-3p, and miR-148a-3p) also found in the public dataset analyzed. Bioinformatic analysis revealed that the 14 identified miRNAs target 17 genes. Reanalyzing two public gene expression datasets, five of these genes have been found differentially expressed in PanNET compared to controls. CONCLUSIONS Our preliminary results, albeit limited by a small sample size, highlighted a specific miRNA expression pattern able to distinguish tumoral from normal pancreatic tissue. The diagnostic performance of these miRNAs, matching with circulating miRNAs and validated in more homogeneous and large cohorts, could represent a starting point for improving the diagnostic accuracy of PanNETs.
Collapse
Affiliation(s)
- Oana A Ciobanu
- Department of Endocrinology and Diabetes, Elias Hospital, Bucharest, Romania
- Department of Endocrinology and Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Vlad Herlea
- Fundeni Clinical Institute, Bucharest, Romania
- Department of Pathological Anatomy, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Elena Milanesi
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Department of Cellular, Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Maria Dobre
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Simona Fica
- Department of Endocrinology and Diabetes, Elias Hospital, Bucharest, Romania
- Department of Endocrinology and Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
44
|
Wang L, Liu H, Chen G, Wu Q, Xu S, Zhou Q, Zhao Y, Wang Q, Yan T, Cheng X. Bubble Ticket Trip: Exploring the Mechanism of miRNA Sorting into Exosomes and Maintaining the Stability of Tumor Microenvironment. Int J Nanomedicine 2024; 19:13671-13685. [PMID: 39723172 PMCID: PMC11669276 DOI: 10.2147/ijn.s498599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Exosomes are vesicles ranging from 30 to 100 nanometers in size that show great potential as carriers for therapeutic uses and drug delivery. Enriching a specific set of miRNAs in exosomes emphasizes the existence of particular sorting mechanisms that manage the targeted cargo packaging. The molecular mechanism for miRNA sorting has not been understood. It is crucial to understand the mechanism of exosome encapsulation to develop its therapeutic potential. In this review, we will explore the particular processes through which exosomes naturally encapsulate miRNA, as well as discuss the effect on tumors after encapsulation of miRNAs. We also summarize the effects of targeted drug delivery using genetic engineering and chemical methods to modify exosome-encapsulated miRNA. Finally, gaining insight into how exosome cargo is sorted could be applied in clinical settings for precise drug delivery and to hinder the progression of diseases.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Guohui Chen
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qinglu Wu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Songrui Xu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qichao Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Yadong Zhao
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qiaorong Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Ting Yan
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| |
Collapse
|
45
|
Rad SG, Orang FN, Shadbad MA. MicroRNA networks in prolactinoma tumorigenesis: a scoping review. Cancer Cell Int 2024; 24:418. [PMID: 39702128 PMCID: PMC11660578 DOI: 10.1186/s12935-024-03529-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/11/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Prolactinoma is the leading type of pituitary adenoma. Aside from the mass-like effect of prolactinoma, its hormonal effect is the main pathological cause of endocrine dysregulation and infertility. The dopamine agonist administration and surgical resection are the current mainstream anti-neoplastic treatments for affected patients; however, tumor fibrosis, tumor invasion, dopamine agonist resistance, and gain prolactinomas are clinical challenges for treating affected patients. Therefore, there is a need to develop novel treatments for these patients. Although growing evidence has highlighted the significance of dysregulated microRNA (miRNA) expression in various malignancies, no study has systematically investigated the significance of miRNA networks and their therapeutic potential in prolactinoma. For this aim, the current scoping review was performed according to the systematic reviews and meta-analyses extension for scoping reviews (PRISMA-ScR) guideline. MAIN BODY The systematic study on PubMed, Web of Science, Scopus, and Embase databases has shown that miR-200c, miR-217, miR-93a, miR-93, miR-1299, and miR-9 are the oncogenic miRNAs and miR-137, miR-145-5p, miR-197-3p, miR-29a-3p, miR-489, miR-199a-5p, miR-124, miR-212, miR-129-5p, miR-130a-3p, miR-326, miR-432, miR-548c-3p, miR-570, miR-15, miR-16, miR-26a, miR-196a2, and let-7a are tumor-suppressive miRNAs in prolactinoma tumorigenesis. CONCLUSION In summary, inhibiting the oncogenic miRNAs and ectopic expression of tumor-suppressive miRNAs can decrease prolactin secretion, reduce tumor invasion and migration, enhance dopamine agonist efficacy, and inhibit prolactinoma development. These findings can serve as a blueprint for future translational studies investigating miR-based therapeutics for prolactinoma.
Collapse
Affiliation(s)
- Sevil Ghaffarzadeh Rad
- Research Center for Evidence-based Medicine, Iranian EBM Centre: A JBI Centre of Excellence, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Abdoli Shadbad
- Research Center for Evidence-based Medicine, Iranian EBM Centre: A JBI Centre of Excellence, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
46
|
Kepsha MA, Timofeeva AV, Chernyshev VS, Silachev DN, Mezhevitinova EA, Sukhikh GT. MicroRNA-Based Liquid Biopsy for Cervical Cancer Diagnostics and Treatment Monitoring. Int J Mol Sci 2024; 25:13271. [PMID: 39769036 PMCID: PMC11678179 DOI: 10.3390/ijms252413271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Despite prevention strategies, cervical cancer remains a significant public health issue. Human papillomavirus plays a critical role in its development, and early detection is vital to improve patient outcomes. The incidence of cervical cancer is projected to rise, necessitating better diagnostic tools. Traditional screening methods like the cytological examination and human papillomavirus testing have limitations in sensitivity and reproducibility. Liquid-based cytology offers some improvements, but the need for more reliable and sensitive techniques persists, particularly for detecting precancerous lesions. Liquid biopsy is a non-invasive method that analyzes cancer-derived products in biofluids like blood, offering potential for real-time monitoring of tumor progression, metastasis, and treatment response. It can be based on detection of circulating tumor cells (CTCs), circulating free DNA (cfDNA), and microRNAs (miRNAs). This review particularly underlines the potential of microRNAs, which are transported by extracellular vesicles. Overall, this article underscores the importance of continued research into non-invasive diagnostic methods like liquid biopsy to enhance cervical cancer screening and treatment monitoring.
Collapse
Affiliation(s)
| | | | - Vasiliy S. Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov, Ministry of Healthcare of the Russian Federation, Moscow 117997, Russia (D.N.S.)
| | | | | | | |
Collapse
|
47
|
Palomino-Vizcaino G, Bañuelos-Villegas EG, Alvarez-Salas LM. The Natural History of Cervical Cancer and the Case for MicroRNAs: Is Human Papillomavirus Infection the Whole Story? Int J Mol Sci 2024; 25:12991. [PMID: 39684702 PMCID: PMC11641362 DOI: 10.3390/ijms252312991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs (ncRNAs) that negatively regulate gene expression. MiRNAs regulate fundamental biological processes and have significant roles in several pathologies, including cancer. Cervical cancer is the best-known example of a widespread human malignancy with a demonstrated viral etiology. Infection with high-risk human papillomavirus (hrHPV) has been shown to be a causative factor for cervical carcinogenesis. Despite the occurrence of prophylactic vaccines, highly sensitive HPV diagnostics, and innovative new therapies, cervical cancer remains a main cause of death in developing countries. The relationship between hrHPV infection and cervical cancer depends on the integration of viral DNA to the host genome, disrupting the viral regulator E2 and the continuous production of the viral E6 and E7 proteins, which are necessary to acquire and maintain a transformed phenotype but insufficient for malignant cervical carcinogenesis. Lately, miRNAs, the tumor microenvironment, and immune evasion have been found to be major players in cervical carcinogenesis after hrHPV infection. Many miRNAs have been widely reported as deregulated in cervical cancer. Here, the relevance of miRNA in HPV-mediated transformation is critically reviewed in the context of the natural history of hrHPV infection and cervical cancer.
Collapse
Affiliation(s)
- Giovanni Palomino-Vizcaino
- Facultad de Ciencias de la Salud, Unidad Valle de las Palmas, Campus Tijuana, Universidad Autónoma de Baja California, Tijuana 21500, Mexico;
| | - Evelyn Gabriela Bañuelos-Villegas
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México City 07360, Mexico;
| | - Luis Marat Alvarez-Salas
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México City 07360, Mexico;
| |
Collapse
|
48
|
Hossain MM, Mishra AK, Yadav AK, Ismail M, Sata TN, Sah AK, Banik A, Sharma G, Venugopal SK. Free fatty acid-induced DDX3 inhibits autophagy via miR-141 upregulation in diet-induced MASLD mice model system. Ann Hepatol 2024; 30:101758. [PMID: 39631458 DOI: 10.1016/j.aohep.2024.101758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/25/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION AND OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is one of the primary causes of chronic liver disease and may lead to liver cirrhosis and hepatocellular carcinoma. Recent reports suggested that DEAD-box RNA helicase (DDX3) acts as a sensor of free fat accumulation and may modulate the pathogenesis via miRNAs. Hence, we hypothesized that DDX3 might modulate MASLD progression via miRNA-141-mediated inhibition of Sirt-1 and autophagy. MATERIALS AND METHODS RNA and total protein were isolated from free fatty acid-treated HepG2 cells or CDAA-fed C57BL/6 mice (6 mice per group) for 6, 18, 32, or 54 weeks. The cells were transfected with DDX3 or miR-141 or siRNA to DDX3, and Western blots for autophagy markers were performed. RESULTS The FFAs induced the DDX3 and miRNA-141 expression, while downregulating Sirt-1, beclin-1, Atg7, and LC3-II. Overexpression of DDX3 resulted in increased miRNA-141. Overexpression of DDX3 or miRNA-141 downregulated Sirt-1 expression and autophagy marker proteins, while these effects were reversed with siRNA to DDX3. The expression of both DDX3 and miRNA-141 was significantly increased, while autophagy markers were downregulated in CDAA-fed mice. CONCLUSIONS These results confirmed that FFA-induced DDX3 induced the expression of miRNA-141, which in turn targeted Sirt-1 and decreased autophagy.
Collapse
Affiliation(s)
- Md Musa Hossain
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Amit K Mishra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, USA
| | - Ajay K Yadav
- Department of Medical and Molecular genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Md Ismail
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Teja Naveen Sata
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Amrendra K Sah
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Arnab Banik
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Gopal Sharma
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Senthil K Venugopal
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India.
| |
Collapse
|
49
|
Yang S, Liu Y, Zhang B, Li J, Xu F, Yu M, Chen Y, Li C, Liu T, Zhao Y, Zhao Q, Zhang J. GRHPR, Targeted by miR-138-5p, Inhibits the Proliferation and Metastasis of Hepatocellular Carcinoma Through PI3K/AKT Signaling Pathway. Cancer Biother Radiopharm 2024; 39:733-744. [PMID: 38934120 DOI: 10.1089/cbr.2023.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly aggressive cancer. This study elucidates the role of Glyoxylate reductase/hydroxypyruvate reductase (GRHPR) in HCC proliferation and metastasis, along with its molecular mechanism, and identifies miRNAs targeting GRHPR. Materials and Methods: Expression levels of GRHPR and miR-138-5p were assessed using real-time fluorescent quantitative polymerase chain reaction and Western blot techniques. Bioinformatic analysis was employed to identify miRNAs targeting GRHPR, and the results were confirmed via dual-luciferase reporter assays. HCC cell lines overexpressing GRHPR were established to investigate its roles in cell proliferation, migration, and invasion. The biological function of miR-138-5p targeting GRHPR in HCC cells was also evaluated. Furthermore, a xenograft mouse model was utilized to examine the in vivo functions of GRHPR. Results: GRHPR expression was downregulated in HCC, whereas miR-138-5p was upregulated. Overexpression of GRHPR suppressed HCC cell proliferation, migration, and invasion. Conversely, inhibition of GRHPR by miR-138-5p promoted HCC cell proliferation and invasive properties. MiR-138-5p was found to regulate Phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) phosphorylation levels by inhibiting GRHPR expression. Conclusion: This study highlights GRHPR's role as a tumor suppressor in HCC, with its function being regulated by miR-138-5p.
Collapse
Affiliation(s)
- Shuangshuang Yang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yixian Liu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Bushi Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxia Li
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fang Xu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Mengdan Yu
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Chen
- Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Chenglong Li
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ting Liu
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Zhao
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianwei Zhao
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jintao Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
50
|
Zhang T, Zhang Y, Li X, Zhang F, Cheng Z, Shi Y, Zhou X, Wang X. An anti-sense lncRNA of the A-FABP gene regulates the proliferation of hair follicle stem cells via the chi-miR-335-5p/DKK1/β-catenin axis. Int J Biol Macromol 2024; 283:137511. [PMID: 39547602 DOI: 10.1016/j.ijbiomac.2024.137511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Hair follicle development relies on both the epithelial-mesenchymal interaction (EMI) and the proliferation of hair follicle stem cells (HFSCs). This intricate process involves numerous regulatory molecules. Increasing evidence suggests that long non-coding RNAs (lncRNAs) play a crucial role in hair follicle development. However, the functions and molecular mechanisms of many lncRNAs in hair follicle development of cashmere goats remain unclear. Based on our previous lncRNA sequencing results in cashmere goats, an unannotated lncRNA differentially expressed at various stages of hair follicle development, named FABP_AS, was detected. Consequently, we aimed at exploring the function and molecular mechanisms of FABP_AS. We constructed a CRISPR/Cas9 knockout system to specifically knock down FABP_AS, providing a reference model for target lncRNA knockout in animal primary cells. Functional experiment results demonstrated that FABP_AS significantly inhibited HFSCs proliferation. Mechanism experiment results revealed that FABP_AS competitively bond to chi-miR-335-5p, promoted DKK1 gene expression, and reduced Wnt/β-catenin signaling pathway activity. In summary, our findings indicated that FABP_AS acted as a miRNA sponge, sequestering chi-miR-335-5p away from the DKK1 gene, thereby suppressing HFSCs proliferation, which would lay the groundwork for a better understanding of the molecular mechanisms of hair follicle development and provide therapeutic targets for hair loss.
Collapse
Affiliation(s)
- Tongtong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yuelang Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; Hainan Institute of Zhejiang University, Sanya 572024, China
| | - Xiang Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Fan Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zixi Cheng
- School of Electronic Science & Engineering, Southeast University, Nanjing 211000, China
| | - Yujie Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiongbo Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xin Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|