1
|
Mukhopadhyay D, Cocco P, Orrù S, Cherchi R, De Matteis S. The role of MicroRNAs as early biomarkers of asbestos-related lung cancer: A systematic review and meta-analysis. Pulmonology 2025; 31:2416792. [PMID: 38402124 DOI: 10.1016/j.pulmoe.2024.02.002] [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: 11/18/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Asbestos is still the leading cause of occupational cancer mortality worldwide. Asbestos-related lung cancer (LC) and malignant pleural mesothelioma (MPM) prognosis is still poor especially at advanced stage, so early diagnosis biomarkers are needed. MicroRNAs (miRNAs) have been proposed as potential early diagnostic biomarkers of asbestos-related LC and MPM. AIM To evaluate the role of miRNAs as diagnostic and prognostic biomarkers of asbestos-related LC and MPM by performing a literature systematic review and meta-analysis. METHODS MEDLINE, EMBASE via Ovid, PUBMED and Cochrane library databases were systematically searched up to April 2023 to identify relevant articles. A grey literature search was also conducted using the Google Scholar platform. MeSH and free text terms for 'asbestos', 'occupational exposure', 'lung cancer', 'mesothelioma' and 'miRNAs' were used to search the literature. Our systematic review protocol was registered in the PROSPERO database. Study quality was assessed via the Newcastle-Ottawa Scale. RESULTS From the search, 331 articles were retrieved, and, after applying our selection criteria, and exclusion of one study for poor quality, 27 studies were included in the review. Most of the studies were hospital-based case-control, conducted in Europe, and evaluated MPM among men only. MiRNAs expression was measured mainly in plasma or serum. MiR-126, miR-132-3p, and miR-103a-3p were the most promising diagnostic biomarkers for MPM, and we estimated a pooled area under the curve (AUC) of 85 %, 73 %, and 50 %, respectively. In relation to MPM prognosis, miR-197‑3p resulted associated with increased survival time. MiR-126, alone and combined with miR-222, was confirmed associated also to LC diagnosis, together with miR-1254 and miR-574-5p; no miRNA was found associated to LC prognosis. CONCLUSION Based on our systematic literature review there is suggestive evidence that the expression of specific miRNAs in the blood serum or plasma are associated with asbestos-related LC and MPM diagnosis and prognosis. Further large longitudinal studies are urgently needed to validate these findings and elucidate the underlying mechanisms given the potential important implications for patients' survival.
Collapse
Affiliation(s)
- D Mukhopadhyay
- Molecular and Translational Medicine, Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, Cagliari, Italy
| | - P Cocco
- Centre for Occupational and Environmental Health, Division of Population Health, Health Services Research & Primary Care, University of Manchester, Oxford Road, Manchester, United Kingdom
| | - S Orrù
- Operative Unit of Medical Genetics, Health Agency of Sardinia, Hospital Binaghi, Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, Cagliari, Italy
| | - R Cherchi
- Operative Unit of Thoracic Surgery, Hospital G. Brotzu, Cagliari, Italy
| | - S De Matteis
- Department of Health Sciences, University of Milan, Milan, Italy
- NHLI, Imperial College London, United Knigdom
| |
Collapse
|
2
|
Malayaperumal S, Sriramulu S, Jothimani G, Banerjee A, Zhang H, Mohammed Rafi ST, Ramachandran I, NR RK, Sun XF, Pathak S. MicroRNA-122 overexpression suppresses the colon cancer cell proliferation by downregulating the astrocyte elevated gene-1/metadherin oncoprotein. Ann Med 2025; 57:2478311. [PMID: 40208016 PMCID: PMC11986857 DOI: 10.1080/07853890.2025.2478311] [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: 10/26/2023] [Revised: 11/09/2024] [Accepted: 02/17/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNAs that regulate essential cellular functions, such as cell adhesion, proliferation, migration, invasion, and programmed cell death, and therefore, alterations in miRNAs can contribute to carcinogenesis. Previous studies have shown that miRNA-122 is abundant in the liver and regulates cell proliferation, migration, and apoptosis. However, the expression pattern and mechanism of actions of miR-122 remain primarily unknown in colon cancer. METHODS In this study, we analyzed The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) database to assess the clinical significance of astrocyte elevated gene-1 (AEG-1)/metadherin (MTDH) and miR-122 in colon cancer. MiR-122 overexpression studies were performed in HCT116, SW480, and SW620 cell lines. Dual-luciferase assay was carried out to confirm the interaction between AEG-1 and miR-122. In vivo-JetPEI-transfection reagent was used for in-vivo transient transfection of miR-122 in the AOM/DSS-induced colon tumor mouse model. RESULTS Our results demonstrate that miR-122 was downregulated in colon cancer cells, and it influences the expressions of apoptotic factors and inflammatory cytokines. MiR-122 overexpression in HCT116, SW480, and SW620 cells showed upregulation of Caspase 3, Caspase 9, and BAX and decreased expression of BCL2, which are pro-apoptotic and anti-apoptotic members that maintain a ratio between cellular survival and cell death. In vivo transient transfection of miR-122 mimic in AOM/DSS induced colon tumor mouse model showed less inflammation and disease activity. The TCGA-COAD data indicated that AEG-1 expression was higher in patients with low expression of miR-122 and lower AEG-1 expression in patients with higher expression miR-122. CONCLUSION Our findings highlight the key role of miR-122 in the high grade of colonic inflammation, and possibly in colon cancer, and the use of miR-122 mimic might be a therapeutic option.
Collapse
Affiliation(s)
- Sarubala Malayaperumal
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Sushmitha Sriramulu
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Hong Zhang
- Department of Medical Sciences, School of Medicine, Orebro University, Örebro, Sweden
| | - Shabana Thabassum Mohammed Rafi
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Rajesh Kanna NR
- Department of Pathology, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Xiao-Feng Sun
- Division of Oncology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| |
Collapse
|
3
|
Wei F, Liu Y. Magnetic-plasmonic nanoparticle-based surface-enhanced Raman scattering for biomedical detection. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 338:126177. [PMID: 40220683 DOI: 10.1016/j.saa.2025.126177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Surface-enhanced Raman scattering (SERS) is a powerful spectroscopic technique that enables rapid, non-destructive, and susceptible detection of biological samples. The magnetic-plasmonic composite materials composed of magnetic and plasmonic nanoparticles have attracted extensive attention as SERS substrates in the biomedical field because of their ability to enrich, separate, and selectively identify biomolecules. In this review, the state-of-art progress of magnetic-plasmonic nanoparticle (MPNP)-based SERS substrates for biomedical detection is highlighted, covering the design and construction of MPNPs with different morphologies, organic and inorganic surface functionalization strategies adopted to improve the adaptability and applicability in biological systems for MPNPs, application development of MPNPs in biomedical detection, as well as the future challenges and issues to be addressed. It is highly expected that this review will help to fully understand the research status of MPNP-based SERS substrates and facilitate their further development and wider application in biological systems.
Collapse
Affiliation(s)
- Fengxue Wei
- Chinese Academy of Sciences Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, PR China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yaling Liu
- Chinese Academy of Sciences Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, PR China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
4
|
Oryani MA, Mohammad Al-Mosawi AK, Javid H, Tajaldini M, Karimi-Shahri M. A Bioligical Perspective on the role of miR-206 in Colorectal cancer. Gene 2025; 961:149552. [PMID: 40339768 DOI: 10.1016/j.gene.2025.149552] [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: 11/18/2024] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
MicroRNAs (miRs) have emerged as pivotal regulators in the development and progression of colorectal cancer (CRC), and MicroRNA-206 (miR-206) has garnered attention as a potentially influential factor. However, the specific biological functions and complete mechanistic understanding of miR-206 in CRC remain largely uncharacterized. This study aims to bridge this research gap by providing a comprehensive analysis of miR-206's role in CRC. An exploration of the molecular mechanisms regulated by miR-206, its intricate interplay with target genes, and its significant impact on cellular processes highlights its potential utility as both a diagnostic marker and a therapeutic target. The significance of this research lies in potentially enabling the development of innovative therapeutic approaches, ultimately aiming to improve prognosis and survival rates in CRC patients by elucidating the functions of miR-206. Critical pathways, such as c-Met and PTEN/AKT, play crucial roles within the regulatory network of miR-206 in CRC and impact various cellular processes involved in CRC pathogenesis, metastasis, and treatment response. Understanding the complex interactions between miR-206 and key signaling pathways like c-Met and PTEN/AKT is crucial for understanding the underlying mechanisms driving CRC initiation and progression. This knowledge can inform the development of targeted therapeutic interventions, potentially leading to improved patient outcomes and advances in CRC management.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences. Gorgan, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
5
|
van der Merwe M, Myburgh K, Garnis C, Towle R, Engelbrecht AM. Unravelling the role of extracellular vesicles in cervical cancer: Mechanisms of progression, resistance, and emerging therapeutic strategies. Gene 2025; 957:149467. [PMID: 40204037 DOI: 10.1016/j.gene.2025.149467] [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: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Cervical cancer remains a significant global health challenge, particularly in its advanced stages, where treatment resistance complicates effective management. Extracellular vesicles (EVs) are crucial mediators of tumor progression and resistance, primarily through the transfer of miRNA cargo. In cervical cancer, specific miRNAs, including oncogenic miRNAs such as miR-21, miR-221-3p, miR-486-5p, and miR-92a-3p are upregulated in both cells and EVs, promoting proliferation, migration, epithelial-to-mesenchymal transition (EMT), and immune evasion-all of which contribute to therapy resistance and an aggressive tumor phenotype. Conversely, tumor-suppressive miRNAs, such as miR-122-5p, miR-100, and miR-142-3p, are selectively exported from cancer cells via EVs, suggesting a protective mechanism by which cancer cells eliminate these tumor suppressors. This review focuses on the role of oncogenic and tumor-suppressive miRNAs within EVs and their implications for cervical cancer progression and treatment resistance. Additionally, it examines the dynamic interactions between the tumor microenvironment (TME) and EV cargo, as well as emerging EV-based therapeutic strategies. These include the encapsulation of chemotherapeutic agents within EVs, the use of anti-miRs to silence oncogenic miRNAs, the delivery of tumor-suppressive miRNAs, the inhibition of EV release, and the targeting of downstream miRNA-regulated proteins. While miRNA-based therapies remain in the early stages, they hold significant promise for overcoming treatment resistance and improving cervical cancer outcomes.
Collapse
Affiliation(s)
- Michelle van der Merwe
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Kathy Myburgh
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Dutta A, Chakraborty S, Bhattacharya A, Basak U, Pati S, Mukherjee S, Guha D, Banerjee S, Chaudhuri NR, Sarkar D, Jana K, Sa G, Dastidar SG, Das T. hsa-miR-5688 inhibits FOXC1-OCT4/SOX2 feedforward loop that drives chemoresistance in breast cancer stem cells. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200982. [PMID: 40330903 PMCID: PMC12051596 DOI: 10.1016/j.omton.2025.200982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 02/15/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Inherently chemotherapy-resistant breast cancer stem cells (CSCs) are responsible for tumor initiation, metastasis, and relapse. CSCs "acquire" more resistance and stemness upon chemotherapy, thereby making relapse-free survival extremely challenging. Here, we describe a novel role of FOXC1 in "acquired resistance" of breast CSCs during chemotherapy. Putative binding sites of pluripotency factors OCT4 and SOX2, but not NANOG, on FOXC1 promoter, were demonstrated by JASPAR and validated by a docking experiment. Significant decline in FOXC1 expression was noticed after OCT4 or SOX2 ablation in breast CSCs. Contrastingly, presence of putative FOXC1 binding sites on the promoters of stemness genes and drug-resistance marker ABCG2, along with downregulation of OCT4 and SOX2 in FOXC1-ablated CSCs, indicated the existence of a feedforward FOXC1-OCT4/SOX2 transactivation loop in CSCs. Chemotherapy-induced upregulation of FOXC1, stemness, as well as drug resistance in CSCs, and downregulation of the same by prior FOXC1-ablation in in-vitro and in-vivo models, endorsed the contribution of this loop in chemo-induced acquisition of stemness and drug resistance. Finally, over-expression of hsa-miR-5688 sensitized CSCs toward chemotherapy and decelerated recurrence. Accordingly, we demonstrate a hitherto unknown mechanism underpinning chemotherapy-induced resistance in breast CSCs, causing relapse and identified hsa-miR-5688 as a potential therapeutic candidate for relapse-free survival of breast cancer patients.
Collapse
Affiliation(s)
- Apratim Dutta
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sourio Chakraborty
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Apoorva Bhattacharya
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Udit Basak
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Subhadip Pati
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sumon Mukherjee
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Deblina Guha
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Shruti Banerjee
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Nibedita Ray Chaudhuri
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Diptendra Sarkar
- Department of Surgery, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - Kuladip Jana
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Gaurisankar Sa
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Shubhra Ghosh Dastidar
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Tanya Das
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| |
Collapse
|
7
|
Xu M, van de Wiel MA, Martinovičová D, Huseinovic A, van Beusechem VW, Stalpers LJ, Oei AL, Steenbergen RD, Snoek BC. High-throughput 3D spheroid screens identify microRNA sensitizers for improved thermoradiotherapy in locally advanced cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102500. [PMID: 40206659 PMCID: PMC11979520 DOI: 10.1016/j.omtn.2025.102500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Chemoradiotherapy is the standard of care for many locally advanced cancers, including cervical and head and neck cancers, but many patients cannot tolerate chemotherapy. Clinical trials have shown that radiotherapy combined with hyperthermia (thermoradiotherapy) may be equally effective, yet it yields a suboptimal overall survival of patients, emphasizing the need for improvement. MicroRNAs (miRNAs), short non-coding RNA sequences, are often dysregulated in cancer and exhibit significant potential as radiosensitizers by targeting genes associated with the DNA damage response. In this study, high-throughput miRNA screening of four cervical cancer cell lines identified 55 miRNAs with significant sensitizing potential, with 18 validated across 10 additional cancer cell lines (6 cervical and 4 head and neck). Functional studies of 6 miRNAs, including miR-16, miR-27a, miR-181c, miR-221, miR-224, and miR-1293, showed that they reduced DNA damage repair by downregulating ATM, DNA-PKcs, Ku70/80, and RAD51. Additionally, differential expression of miR-27a, miR-221, and miR-224 in treatment-sensitive versus treatment-resistant patients indicated their predictive biomarker potential for treatment response of cervical cancer patients. Conclusively, this study has identified 18 promising miRNAs for the development of sensitizers for thermoradiotherapy and may provide potential biomarkers for predicting treatment response in locally advanced cancers.
Collapse
Affiliation(s)
- MengFei Xu
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Mark A. van de Wiel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Epidemiology and Data Science, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Dominika Martinovičová
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Angelina Huseinovic
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Victor W. van Beusechem
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, Cancer Immunology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Lukas J.A. Stalpers
- Amsterdam UMC, University of Amsterdam, Radiation Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Arlene L. Oei
- Cancer Center Amsterdam, Cancer Biology and Immunology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Amsterdam UMC, University of Amsterdam, Radiation Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Renske D.M. Steenbergen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Barbara C. Snoek
- Cancer Center Amsterdam, Cancer Biology and Immunology, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
- Amsterdam UMC, University of Amsterdam, Radiation Oncology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
8
|
Aloliqi AA, Alnuqaydan AM, Albutti A, Alharbi BF, Rahmani AH, Khan AA. Current updates regarding biogenesis, functions and dysregulation of microRNAs in cancer: Innovative approaches for detection using CRISPR/Cas13‑based platforms (Review). Int J Mol Med 2025; 55:90. [PMID: 40242952 PMCID: PMC12021393 DOI: 10.3892/ijmm.2025.5531] [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: 01/15/2025] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRNAs) are short non‑coding RNAs, which perform a key role in cellular differentiation and development. Most human diseases, particularly cancer, are linked to miRNA functional dysregulation implicated in the expression of tumor‑suppressive or oncogenic targets. Cancer hallmarks such as continued proliferative signaling, dodging growth suppressors, invasion and metastasis, triggering angiogenesis, and avoiding cell death have all been demonstrated to be affected by dysregulated miRNAs. Thus, for the treatment of different cancer types, the detection and quantification of this type of RNA is significant. The classical and current methods of RNA detection, including northern blotting, reverse transcription‑quantitative PCR, rolling circle amplification and next‑generation sequencing, may be effective but differ in efficiency and accuracy. Furthermore, these approaches are expensive, and require special instrumentation and expertise. Thus, researchers are constantly looking for more innovative approaches for miRNA detection, which can be advantageous in all aspects. In this regard, an RNA manipulation tool known as the CRISPR and CRISPR‑associated sequence 13 (CRISPR/Cas13) system has been found to be more advantageous in miRNA detection. The Cas13‑based miRNA detection approach is cost effective and requires no special instrumentation or expertise. However, more research and validation are required to confirm the growing body of CRISPR/Cas13‑based research that has identified miRNAs as possible cancer biomarkers for diagnosis and prognosis, and as targets for treatment. In the present review, current updates regarding miRNA biogenesis, structural and functional aspects, and miRNA dysregulation during cancer are described. In addition, novel approaches using the CRISPR/Cas13 system as a next‑generation tool for miRNA detection are discussed. Furthermore, challenges and prospects of CRISPR/Cas13‑based miRNA detection approaches are described.
Collapse
Affiliation(s)
- Abdulaziz A. Aloliqi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Abdullah M. Alnuqaydan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Aqel Albutti
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Basmah F. Alharbi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| |
Collapse
|
9
|
Zhi Y, Zhang W, Wu Z, Chen Y, Feng L, He J, Wang F, Liu H. miR-223-3p Targets KIF4A and Promotes the Oxidative Stress-Mediated Apoptosis of Breast Cancer Cells. Cancer Biother Radiopharm 2025; 40:323-338. [PMID: 39914820 DOI: 10.1089/cbr.2024.0102] [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: 05/29/2025] Open
Abstract
Background: The abnormal expression of kinase family member 4A (KIF4A) is linked to breast cancer progression, with numerous miRNAs exhibiting abnormal expression. Thus, there is an urgent need to investigate the mechanisms of action of miRNAs and their target genes for the diagnosis and treatment of breast cancer. Materials and Methods: A bioinformatics analysis was conducted to screen for KIF4A, a key gene involved in oxidative stress in breast cancer cells. Using CCK8, EdU, cell healing, and Transwell assays, the knockdown of KIF4A was found to effectively inhibit the proliferation, migration, and invasion of breast cancer cells. Dual-luciferase assay and Western blotting confirmed that miR-223-3p targets and regulates KIF4A expression. The impact of miR-223-3p and KIF4A on oxidative stress in breast cancer cells was assessed through reactive oxygen species (ROS), superoxide dismutase (SOD), and malondialdehyde (MDA) measurements. Flow cytometry was used to evaluate tumor cell apoptosis. Results: Our results suggest that KIF4A is a downstream target of miR-223-3p. miR-223-3p inhibits the proliferation and invasion of breast cancer cells by directly targeting and downregulating KIF4A. Importantly, we found that miR-223-3p and KIF4A play important roles in regulating oxidative stress and apoptosis in breast cancer cells. Specifically, miR-223-3p promoted apoptosis by inhibiting the expression of KIF4A, increasing the accumulation level of ROS and MDA, and inhibiting the activity of SOD while KIF4A was overexpressed.
Collapse
Affiliation(s)
- Yinghui Zhi
- Department of Gland Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Wenshan Zhang
- Department of Gland Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhenyu Wu
- Department of General Surgery, The First Hospital of Hebei Medic University, Shijiazhuang, China
| | - Yan Chen
- Department of Pediatrics, The Fourth Hospital of Hebei Medic University, Shijiazhuang, China
| | - Liang Feng
- Department of Gland Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Jing He
- Department of Gland Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Feng Wang
- Department of Cardiac Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Huan Liu
- Department of Gland Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
10
|
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
|
11
|
Yan L, Quan Z, Sun T, Wang J. Autophagy signaling mediated by non-coding RNAs: Impact on breast cancer progression and treatment. Mol Aspects Med 2025; 103:101365. [PMID: 40305994 DOI: 10.1016/j.mam.2025.101365] [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: 11/26/2024] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025]
Abstract
Autophagy, a conserved cellular mechanism which detoxifies and degrades intracellular structures or biomolecules, has been identified as an important factor in the progression of human breast cancer and the development of treatment resistance. Non-coding RNAs (ncRNAs), a broad family of RNA, have the ability to influence various processes, including autophagy, due to their diverse downstream targets. ncRNAs play an important role in suppressing or activating autophagy by targeting autophagy-triggering components such as the ULK1 complex, Beclin1, and ATGs. Recent research has uncovered the intricate regulatory networks that govern autophagy dynamics, with ncRNAs emerging as key participants in this network. miRNAs, lncRNAs, and circRNAs are the three subfamilies of ncRNAs that have the most well-known interactions with autophagy, particularly macroautophagy. The high prevalence of breast cancer necessitates research into finding new biological processes that can help in early detection as well as enhance the effectiveness of treatment. The positive/negative link between autophagy and ncRNAs can be exploited as a supplementary therapy to improve sensitivity to treatment in breast cancer. This review investigates the regulatory roles of ncRNAs, particularly microRNAs (miRNAs), in modifying autophagy pathways in human breast cancer progression and treatment. However, future studies and clinical practice are needed to determine the most relevant microRNAs as biomarkers and also to better understand their role in breast cancer progression or treatment through modifying autophagy.
Collapse
Affiliation(s)
- Lei Yan
- Clinical Experimental Centre, Xi'an International Medical Center Hospital, No.777 Xitai Road, High-tech Zone, Xi'an, Shaanxi Province, 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptide, Xi'an, Shaanxi, 710100, China
| | - Zhuo Quan
- Clinical Experimental Centre, Xi'an International Medical Center Hospital, No.777 Xitai Road, High-tech Zone, Xi'an, Shaanxi Province, 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptide, Xi'an, Shaanxi, 710100, China
| | - Tiantian Sun
- Department of Oncology, Zibo Central Hospital, Shandong, 255036, China.
| | - Jiaju Wang
- Department of Hematology, Zibo Central Hospital, Shandong, 255036, China.
| |
Collapse
|
12
|
You Q, Wang P, Zhu T, Jia Z, Chang Z, Li L, Dong WF. Bifunctional MXene quantum dots-coated bimetallic Prussian blue analogues for sensitive sensing and accurate localization imaging of miRNAs in living cells. Mater Today Bio 2025; 32:101747. [PMID: 40290880 PMCID: PMC12032912 DOI: 10.1016/j.mtbio.2025.101747] [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: 02/10/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
MicroRNAs (miRNAs) are involved in multiple cellular processes and play a critical role in clinical diagnosis. In-situ spatiotemporal imaging of miRNAs in living cells is tightly linked to the carcinogenesis and development of malignant tumors. Herein, we proposed a bifunctional nanosystem-based MXene quantum dots-coated bimetallic Prussian blue analogues (Co-Mn PBA@MQDs) to execute in-vitro sensing and intracellular imaging of miRNA in living cells. The 3D nanostructures of Co-Mn PBAs were regulated to slow down the coordination reaction rate by controlling the diffusion of metal clusters and ligand precursors, thereby anchoring MQDs as the carriers of DNA probes. The resulting Co-Mn PBA@MQDs nanoparticles with miRNA recognition ability exhibit excellent electrocatalytic and photoluminescence properties for target miRNA analysis. It reached miRNA detection limit of 0.37 fM (S/N = 3) with a wide linear range of 1 fM to 1 nM, and allowed distinguish family members without additional complex modifications. Meanwhile, DNA probe adsorbed on Co-Mn PBA@MQDs can provide delivery capacity for intracellular miRNA location, resulting in the in-situ monitoring and imaging of miRNA with deregulated expression levels in cancer cells. With these advantages, the developed strategy provides a paradigm for the rational design of the miRNA analysis system, which is expected to be widely applied to disease diagnosis and further theragnostic fields.
Collapse
Affiliation(s)
- Qiannan You
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
| | - Panyong Wang
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
| | - Tongtong Zhu
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| | - Zixuan Jia
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| | - Zhimin Chang
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| | - Li Li
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| | - Wen-Fei Dong
- Department of Biomaterials and Stem Cells, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, PR China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
13
|
Aleshcheva G, Salih S, Baumeier C, Escher F, Bock CT, Schultheiss H. Discovery of miRNAs unique to actively transcribed erythroparvovirus infection in heart failure patients. ESC Heart Fail 2025; 12:1872-1882. [PMID: 39970057 PMCID: PMC12055386 DOI: 10.1002/ehf2.15194] [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: 07/01/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 02/21/2025] Open
Abstract
AIMS miRNAs, small non-coding RNAs, play key roles in gene regulation, cell differentiation and tissue development. They influence viral infection outcomes by directly interacting with viral genomes or modifying the host microenvironment. This study demonstrates miRNAs' ability to selectively suppress transcriptionally active erythroparvovirus, highlighting their potential in antiviral therapies. METHODS AND RESULTS Seventy-five endomyocardial biopsy (EMB) specimens from patients with unexplained heart failure were analysed. The samples included 19 with dilated cardiomyopathy and inflammation (DCMi), 12 with dilated cardiomyopathy (DCM), 25 with inflammation and active erythroparvovirus infection, 13 with active erythroparvovirus infection only and 6 from undiagnosed patients as controls. miRNA expression was measured using TaqMan assays. miR-98, miR-222, miR-106b and miR-197 were significantly upregulated in patients with transcriptionally active erythroparvovirus infection, independent of inflammation (P < 0.005). These miRNAs differentiated these patients from all other groups with over 90% specificity. CONCLUSIONS These specific miRNAs offer a novel diagnostic tool for active erythroparvovirus infections and hold promise as therapeutic targets, providing safer alternatives to traditional antiviral treatments.
Collapse
Affiliation(s)
- Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT)BerlinGermany
| | - Sara Salih
- BHT – Berliner Hochschule für TechnikBerlinGermany
| | | | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT)BerlinGermany
- DHZC (German Heart Centre of Charité)BerlinGermany
- DZHK (German Centre for Cardiovascular Research), partner site BerlinBerlinGermany
| | - C. Thomas Bock
- Department of Infectious Diseases, Division of Viral Gastroenteritis and Hepatitis Pathogens and EnterovirusesRobert Koch InstituteBerlinGermany
| | | |
Collapse
|
14
|
Lobo J, Tavares NT, Fonseca D, Jerónimo C, Henrique R, Wyvekens N, Yang Y, Snuderl M, Maclean F, Gordetsky J, Fletcher CDM, Hirsch MS, Hornick JL, Idrees MT, Collins K, Warmke L, Ulbright TM, Acosta AM. MicroRNA-371-373 cluster and methylome analysis suggests that a subset of 'somatic-type' malignancies arising in germ cell tumors may originate in yolk sac tumor components. J Pathol 2025; 266:160-176. [PMID: 40152072 PMCID: PMC12056289 DOI: 10.1002/path.6412] [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: 07/03/2024] [Revised: 12/28/2024] [Accepted: 02/04/2025] [Indexed: 03/29/2025]
Abstract
Somatic-type malignancies (SMs) arising in germ cell tumors (GCTs) are aggressive neoplasms resistant to systemic treatment. Most are diagnosed in metastatic sites after chemotherapy; however, they have also been well-documented in primary testicular GCTs. Historically, SMs were thought to originate in components of teratoma that acquire molecular alterations equivalent to those that characterize their true somatic counterparts. However, recent studies have shown that SMs typically lack the hallmark molecular alterations seen in similar somatic tumors. Additionally, clinicopathologic and molecular data suggest that a subset may derive from yolk sac tumor (YST) rather than teratoma. In this study, we evaluated the relationship between conventional histological types of GCTs and SMs by comparing expression of microRNA (miR)-371-373 and genomic methylation profiles. A total of 96 samples (including multiple paired conventional GCT-SM samples from individual tumors) were assessed for miR-371-373 expression by RT-qPCR and genomic DNA methylation using a clinically validated assay. Expression of miR-371-373 was higher in conventional GCTs than in SMs (considered as a single category encompassing all histological subtypes). However, miR-371-373 expression was heterogeneous among SMs, with significantly higher levels in sarcomatoid YST (SYST) and glandular neoplasms than in other SMs. Genomic DNA methylation analysis showed that SMs (considered as a single category) did not form a distinct cluster. Instead, they grouped into multiple clusters that did not show perfect correspondence with histology and often included conventional GCTs. Genome-wide methylation assessment showed a higher abundance of hypermethylated regions in SMs than in conventional GCTs. Analysis of paired conventional GCT and 'somatic-type' components that did not meet size criteria for SMs dissected from individual tumors demonstrated separation according to histology, suggesting that epigenetic processes play a role in the transition from conventional GCT to 'somatic-type' phenotypes. Gene-level and pathway-level analyses identified MAPK/RAS signaling, mitosis/proliferation, differentiation towards neural tissue/neuroectoderm, epithelial-to-mesenchymal transition, and DNA repair as key differentially regulated processes in components with somatic-type histology, suggesting mechanisms of progression from conventional to 'somatic' phenotypes in GCT. These results support the hypothesis that a subset of SMs derive from YST and suggest that some subtypes (such as SYST) may represent 'intermediate' phenotypes. Additionally, analysis of differentially methylated promoter regions in SM identified genes and biologic processess that may underlie 'somatic tranformation' in GCTs. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- João Lobo
- Department of PathologyPortuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC)PortoPortugal
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI‐IPOP)Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & CI‐IPOP@RISE (Health Research Network)PortoPortugal
- Department of Pathology and Molecular Immunology, ICBAS – School of Medicine and Biomedical SciencesUniversity of PortoPortoPortugal
| | - Nuno Tiago Tavares
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI‐IPOP)Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & CI‐IPOP@RISE (Health Research Network)PortoPortugal
- Doctoral Programme in Biomedical Sciences, ICBAS – School of Medicine and Biomedical SciencesUniversity of PortoPortoPortugal
| | - Diana Fonseca
- Department of PathologyPortuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC)PortoPortugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI‐IPOP)Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & CI‐IPOP@RISE (Health Research Network)PortoPortugal
- Department of Pathology and Molecular Immunology, ICBAS – School of Medicine and Biomedical SciencesUniversity of PortoPortoPortugal
| | - Rui Henrique
- Department of PathologyPortuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC)PortoPortugal
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI‐IPOP)Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & CI‐IPOP@RISE (Health Research Network)PortoPortugal
- Department of Pathology and Molecular Immunology, ICBAS – School of Medicine and Biomedical SciencesUniversity of PortoPortoPortugal
| | - Nicolas Wyvekens
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Yiying Yang
- Department of PathologyNew York University, Langone HealthNew YorkNYUSA
| | - Matija Snuderl
- Department of PathologyNew York University, Langone HealthNew YorkNYUSA
| | - Fiona Maclean
- Department of Anatomical PathologyDouglass Hanly Moir PathologyMacquarie ParkNSWAustralia
- Cancer Diagnosis and Pathology GroupKolling Institute of Medical Research, Royal North Shore HospitalSt LeonardsNSWAustralia
- Department of Clinical Medicine, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNSWAustralia
| | - Jennifer Gordetsky
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
| | | | - Michelle S Hirsch
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Jason L Hornick
- Department of PathologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMAUSA
| | - Muhammad T Idrees
- Department of PathologyIndiana University School of MedicineIndianapolisINUSA
| | - Katrina Collins
- Department of PathologyIndiana University School of MedicineIndianapolisINUSA
| | - Laura Warmke
- Department of PathologyIndiana University School of MedicineIndianapolisINUSA
| | - Thomas M Ulbright
- Department of PathologyIndiana University School of MedicineIndianapolisINUSA
| | - Andres M Acosta
- Department of PathologyIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
15
|
Tavares NT, Lourenço C, Constâncio V, Fernandes-Pontes F, Fonseca D, Silva-Santos R, Braga I, Maurício J, Henrique R, Liu M, Weiss RS, Bagrodia A, Jerónimo C, Lobo J. MicroRNA-371-373 cluster extracellular vesicle-based communication in testicular germ cell tumors. Cell Commun Signal 2025; 23:252. [PMID: 40448114 DOI: 10.1186/s12964-025-02250-8] [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: 03/30/2025] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most common type of cancer in young adults. The cluster of microRNAs 371-373 is highly upregulated in TGCTs, and detection of miR-371a-3p specifically is currently being developed for clinical implementation as a sensitive and specific biomarker for TGCT, except for teratoma. Extracellular vesicles (EVs) are nano-sized particles used for cell communication, being increasingly regarded as potential sources of cancer biomarkers. Thus, the aim of this study was to characterize EVs from a wide range of TGCT samples, including cell lines, tissue explants and matched plasma samples from patients and healthy donors, and then use these samples to assess microRNA expression (miR-371-373 cluster and let-7e). TGCT-derived EVs were successfully isolated and characterized according to MISEV guidelines. TGCT cell lines showed different levels of EV-derived miR-371-373 cluster and let-7e. Upon differentiation of NT2 cells with ATRA, both cellular and EV-derived miR-371-373 cluster were downregulated, whereas let-7e was upregulated. TGCT patient samples presented high levels of EV-derived miR-371-373, except for the teratoma samples. We conclude that a significant portion of the circulating miR-371-373 cluster used as a TGCT biomarker in the clinic is secreted into EVs, and that this cluster and the let-7 family of microRNAs may be related with TGCT intercellular communication and differentiation.
Collapse
Affiliation(s)
- Nuno Tiago Tavares
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Catarina Lourenço
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
- i3S - Instituto de Investigação e Inovação Em Saúde - University of Porto, Porto, 4200-135, Portugal
- Instituto Nacional de Engenharia Biomédica, Porto, 4200-135, Portugal
| | - Vera Constâncio
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Fernanda Fernandes-Pontes
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
| | - Diana Fonseca
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
| | - Rui Silva-Santos
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Isaac Braga
- Department of Urology, Urology Clinic, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Joaquina Maurício
- Department of Medical Oncology, Urology Clinic, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Michelle Liu
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal.
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal.
| |
Collapse
|
16
|
Shirzad S, Eterafi M, Karimi Z, Barazesh M. MicroRNAs involved in colorectal cancer, a rapid mini-systematic review. BMC Cancer 2025; 25:934. [PMID: 40413456 DOI: 10.1186/s12885-025-14343-1] [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: 11/29/2024] [Accepted: 05/16/2025] [Indexed: 05/27/2025] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) involves the uncontrolled proliferation of glandular epithelial cells in the colon or rectum. The high mortality rate associated with CRC has driven extensive research into innovative diagnostic and therapeutic strategies. Among these, microRNAs (miRNA) have gained attention for their crucial role in regulating various cellular processes that contribute to the initiation, progression, and metastasis of CRC. METHOD This systematic review aimed to assess the roles of various miRNAs in CRC by analyzing multiple studies. The PICO framework was followed to structure the study regarding miRNA involved in CRC development and progression compared to normal cases. The outcomes were measured according miRNAs impact on CRC progression, survival rates, and treatment response. Systematic review of studies published from 2000 to November 2023 were included. Data were collected from prominent databases, including Google Scholar, PubMed, ScienceDirect, Irandoc, SID, and Magiran, covering studies from 2000 to November 2023. Studies were managed using EndNote for citation management, and duplicates were removed. The remaining studies were evaluated based on predefined inclusion and exclusion criteria. RESULTS In our review, we categorized 28 miRNAs based on their potential tumor suppressor or oncogenic effects in CRC progression. Among them, 14 miRNAs were highlighted as important based on the assessment using TCGA data, with miR-200a also showing a significant effect on patient survival. CONCLUSION This study compiled and analyzed validated miRNAs associated with CRC progression. The findings suggest the potential of these miRNAs as non-invasive biomarkers, which may be used alone or in combination with traditional tumor markers for improved diagnostic and prognostic applications in CRC. This review contributes novel insights by updating the current understanding and offering a comprehensive evaluation of miRNAs in CRC.
Collapse
Affiliation(s)
- Sogol Shirzad
- Students Research Committee, Gerash University of Medical Sciences, Gerash, Iran
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran
| | - Majid Eterafi
- Students Research Committee, Gerash University of Medical Sciences, Gerash, Iran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zeinab Karimi
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran.
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran.
| | - Mahdi Barazesh
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
17
|
Javankiani S, Bolandi S, Soleimani A, Meigoli MSS, Parsafar M, Safaei S, Esmailpour M, Nadimi S, Avval NA, Fazayel SMA, Zahed Z, Sharafi M. MAPK signaling mediates tamoxifen resistance in estrogen receptor-positive breast cancer. Mol Cell Biochem 2025:10.1007/s11010-025-05304-0. [PMID: 40410609 DOI: 10.1007/s11010-025-05304-0] [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: 02/22/2025] [Accepted: 05/04/2025] [Indexed: 05/25/2025]
Abstract
Tamoxifen is a cornerstone in the treatment of estrogen receptor (ER)-positive breast cancer, yet resistance to this therapy remains a significant clinical challenge. In most cases, the resistance phenotype is not caused by loss or mutation of the ER, but by changes in multiple proliferative and survival pathways. The mitogen-activated protein kinase (MAPK) signaling pathways regulate various cellular processes such as cell growth, proliferation, and apoptosis. This review provides a comprehensive analysis of molecular mechanisms that sustain MAPK activation and promote tamoxifen resistance. We evaluated molecular factors that promote the survival of tamoxifen-resistant cells through the regulation of MAPK signaling, including growth factors, RNA-binding proteins, non-genomic ER variants, and microRNAs. Mitochondrial dynamics and their regulation by MAPK highlight novel adaptive mechanisms employed by resistant cells to survive. Furthermore, MAPK-mediated phosphorylation of ERα enhances resistance through ligand-independent activation and sustained cellular proliferation. MAPK and parallel oncogenic pathways, including PI3K/AKT and receptor tyrosine kinases (EGFR, IGF-1R, and FGFR), function synergistically to enhance signaling redundancy and compensatory survival mechanisms. Therapeutic interventions targeting MAPK signaling-ranging from small-molecule inhibitors to RNA-based therapies-offer promising avenues for overcoming tamoxifen resistance.
Collapse
Affiliation(s)
- Sepide Javankiani
- General Surgery Department, Tehran University of Medical Sciences, Tehran, Iran
- International Surgical Research Association (ISRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Soheil Bolandi
- Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anvar Soleimani
- Department of Medical Microbiology, College of Health Sciences, Cihan University Sulaimaniya, Kurdistan, Sulaimaniya City, Iraq
| | | | - Mahdis Parsafar
- Department of Biomedical Engineering, College of Medical Sciences and Technologies, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Safaei
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojgan Esmailpour
- Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sogol Nadimi
- Department of Chemistry, University of Bonn, Bonn, Germany
| | | | - Seyed Mohammad Ali Fazayel
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Zahra Zahed
- Department of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Malihe Sharafi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran.
| |
Collapse
|
18
|
Kim S, Han M, Hwang HJ, Ahn YH, Im HJ, Hwang SH, Koh KN, Kim N. MicroRNA-196a increases apoptosis in B cells through downregulation of FOXO1. Mol Cells 2025:100223. [PMID: 40403879 DOI: 10.1016/j.mocell.2025.100223] [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: 12/09/2024] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025] Open
Abstract
MicroRNAs (miRNAs) are key regulators of cancer pathogenesis, and their expression is often dysregulated in cancer cells. The role of miR-196a-5p has been investigated in various types of cancers, however it is relatively less understood in B-cell malignancies. This study aimed to investigate the role of miR-196a-5p in B cells by using a human diffuse large B-cell lymphoma (DLBCL) cell line, SU-DHL-6 and mouse B lymphocytes. The enforced expression of miR-196a in SU-DHL-6 cells increased daunorubicin-mediated apoptosis. Luciferase assay revealed that FOXO1 was a direct target of miR-196a-5p in SU-DHL-6 cells. The mRNA and protein expression of FOXO1 was downregulated in miR-196a-overexpressing SU-DHL-6 cells. In addition, miR-196a-5p was highly expressed in mouse bone marrow (BM) cells, compared with that of splenic (SP) B cells, and FOXO1 expression was negatively correlated with miR-196a-5p level. miR-196a-5p was upregulated by B cell receptor (BCR)-stimulation, which was inversely correlated with FOXO1 expression in SP B cells. Apoptosis was increased when miR-196a-5p was upregulated in murine primary B cells. These results identify miR-196a-5p as a post-transcriptional regulator of FOXO1 and indicate its importance in regulating B cell malignancies and activation.
Collapse
Affiliation(s)
- Soyoung Kim
- Asan Institute for Life Sciences and Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mina Han
- Asan Institute for Life Sciences and Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyun Ju Hwang
- Asan Institute for Life Sciences and Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Ho Ahn
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Hyun Hwang
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung-Nam Koh
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Nayoung Kim
- Asan Institute for Life Sciences and Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Perez-Moreno E, Ortega-Hernández V, Zavala VA, Gamboa J, Fernández W, Carvallo P. Suppression of breast cancer metastatic behavior by microRNAs targeting EMT transcription factors. A relevant participation of miR-196a-5p and miR-22-3p in ZEB1 expression. Breast Cancer Res Treat 2025:10.1007/s10549-025-07723-5. [PMID: 40382762 DOI: 10.1007/s10549-025-07723-5] [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: 07/08/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
PURPOSE Metastasis, the leading cause of cancer-associated deaths, is promoted by transcription factors SNAIL, SLUG, ZEB1 and TWIST through the activation of epithelial-mesenchymal transition (EMT). MicroRNAs can suppress EMT, emerging as candidate molecular biomarkers and novel therapeutic targets. Herein, we evaluated microRNAs downregulated in breast cancer (BC) tissues expressing EMT transcription factors, to find new potential regulators of EMT. METHODS Candidate microRNAs were selected from microarray data by their inversely correlated expression with SNAIL, SLUG, ZEB1 and TWIST, evaluated in BC tissues through immunohistochemistry. We selected eight microRNAs predicted in silico as probable modulators of SNAIL, SLUG, ZEB1 and TWIST, and validate their interaction through the 3'UTR region in luciferase reporter gene assays. MDA-MB-231 cells were transfected with selected microRNAs to perform migration, invasion and cell proliferation assays, and western blot was used to evaluate protein levels. RESULTS MiR-30a-5p, miR-1271-5p, miR-196a-5p, miR-202-3p, miR-210-3p, miR-22-3p and miR-331-3p decreased luciferase activity through SNAIL, SLUG, ZEB1 and/or TWIST 3'UTR. These microRNAs, including miR-34b-3p, decreased migration, invasion and cell proliferation in MDA-MB-231 cells. MiR-30a-5p, miR-202-3p and miR-22-3p decreased vimentin expression, whereas miR-196a-5p and miR-22-3p decreased endogenous ZEB1 levels. MiR-196a-5p, miR-202-3p and miR-30a-5p also decreased CCR7 expression, a chemokine receptor involved in lymph node metastasis. CONCLUSION microRNAs selected in this work can regulate gene expression trough 3'UTR region of EMT-transcription factors. In BC cells, miR-196a-5p and miR-22-3p decrease ZEB1 levels, being novel modulators of EMT. Also, the eight evaluated microRNAs, reduced the metastatic hallmarks in BC cells.
Collapse
Affiliation(s)
- Elisa Perez-Moreno
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Victoria Ortega-Hernández
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina A Zavala
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Gamboa
- Unidad de Patología Mamaria, Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Wanda Fernández
- Unidad de Anatomía Patológica, Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Pilar Carvallo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Maurya N, Meena A, Luqman S. Role of microRNAs in lung oncogenesis: Diagnostic implications, resistance mechanisms, and therapeutic strategies. Int J Biol Macromol 2025:144261. [PMID: 40381781 DOI: 10.1016/j.ijbiomac.2025.144261] [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: 07/28/2024] [Revised: 04/16/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Lung cancer continues to pose a significant global health concern, presenting a formidable challenge on a worldwide scale, necessitating a deeper understanding of molecular mechanisms underlying its pathogenesis and treatment responses. microRNA (miRNA) modulation in the context of lung cancer therapeutics aims to unravel the complexities of miRNA-mediated regulatory networks. This comprehensive review elucidates microRNA's diverse roles in lung cancer, encompassing their involvement in key signaling pathways, cellular processes, the regulation of oncogenic or tumor-suppressive targets, and drug sensitivity. Moreover, this review critically examines the potential of miRNAs as diagnostic and prognostic biomarkers and their implications in therapeutic interventions for lung cancer. microRNAs are effective in making lung cancer therapy more efficient. They can make tumor cells more responsive to chemotherapy, radiation, and targeted therapies. microRNAs can target the drug efflux mechanism, increasing the effectiveness of chemotherapy agents and decreasing resistance. Furthermore, microRNAs play a crucial role in developing and inhibiting the resistance mechanisms against conventional treatments; improving the dysregulated expression of microRNAs enhances the therapeutic efficacy of existing therapies. By compiling knowledge on miRNA-mediated processes related to lung cancer, this review offers a comprehensive resource for researchers to understand and address the complexities of oncogenesis, diagnostics, resistance mechanisms, and therapeutic strategies.
Collapse
Affiliation(s)
- Nidhi Maurya
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226 015, Uttar Pradesh, India.; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 001, Uttar Pradesh, India
| | - Abha Meena
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226 015, Uttar Pradesh, India.; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 001, Uttar Pradesh, India
| | - Suaib Luqman
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226 015, Uttar Pradesh, India.; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 001, Uttar Pradesh, India.
| |
Collapse
|
21
|
Banerjee S, Bose D, Johnson S, Liu J, Virgin H, Robertson ES. Novel small non-coding RNAs of Epstein-Barr virus upregulated upon lytic reactivation aid in viral genomic replication and virion production. mBio 2025; 16:e0406024. [PMID: 40197026 PMCID: PMC12077129 DOI: 10.1128/mbio.04060-24] [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: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Epstein-Barr virus (EBV) employs various strategies for long-term survival, including the expression of non-coding RNAs (ncRNAs). This study uncovers and characterizes two novel EBV-encoded ncRNAs, p7 and p8, which are upregulated during lytic reactivation and interact with both viral and host genomes. These ncRNAs bind to cellular RNA transcripts, significantly reducing ARMCX3 mRNA levels, while p8 also influences PTPN6 and RPL24 expressions. Although p7 does not directly bind to LMP1 RNA but both ncRNAs found to downregulate LMP1 expression. Furthermore, these ncRNAs interact with the OriLyt region of EBV genome, promoting viral DNA replication. Functional assays indicate that p7 and p8 enhance cell proliferation and inhibit apoptosis by modulating the p53 pathway and suppressing pro-apoptotic proteins. These findings highlight the role of p7 and p8 in supporting EBV persistence by regulating viral replication, cell survival, and immune evasion, making them promising targets for therapeutic strategies in EBV-related diseases.IMPORTANCEEpstein-Barr virus (EBV) employs diverse strategies for long-term persistence in the host, including the expression of viral non-coding RNAs (ncRNAs) that manipulate key cellular pathways to promote viral replication and immune evasion. This study identifies two novel EBV-encoded ncRNAs, p7 and p8, which are upregulated during lytic reactivation and interact with both viral and host genes to regulate viral DNA replication and promote host cellular survival. By modulating apoptotic and proliferative pathways, p7 and p8 facilitate viral reactivation while promoting host cell survival, highlighting their potential as critical regulators in EBV-driven oncogenesis. This discovery expands our understanding of EBV-host interactions, suggesting p7 and p8 as targets for novel therapeutic strategies in EBV-associated malignancies.
Collapse
Affiliation(s)
- Sagarika Banerjee
- Departments of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dipayan Bose
- Departments of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steve Johnson
- Department of Pathology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jie Liu
- Departments of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Herbert Virgin
- Department of Pathology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Erle S. Robertson
- Departments of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Xia S, Fan H, Xiao J, Shen C, Yan Y, Wang M, Tang T, Sun W, Wang J, Jia X, Lai S. MiR- 223 alleviates the heat-stress-induced inhibition of cell proliferation by targeting PRDM1. BMC Genomics 2025; 26:470. [PMID: 40355855 PMCID: PMC12067916 DOI: 10.1186/s12864-025-11567-0] [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: 06/07/2024] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Heat stress, exacerbated by global warming, has emerged as a significant concern for both the health of dairy cattle and the quality of milk production. In vitro investigations suggest that primary bovine mammary epithelial cells exhibit enhanced levels of programmed cell death when subjected to elevated ambient temperatures, potentially resulting in a reduction in the total number of mammary epithelial cells within the mammary gland, thereby partially elucidating the diminished milk yield in lactating cows under heat stress. In vivo, heat stress affects both milk synthesis and secretion by directly acting on mammary epithelial cells and by altering hormonal levels and metabolic pathways, which can lead to long-term effects on mammary growth. Future research should focus on elucidating the molecular mechanisms by which heat stress regulates mammary development. Previous studies have demonstrated that heat stress induction results in a significant downregulation of miR- 223 in MAC-T cells; therefore, miR- 223 may play a crucial role in the response to heat stress. Nevertheless, the mechanism by which miR- 223 confers resistance to heat stress in MAC-T remains unclear. METHODS Here, to investigate how miR- 223 regulates the proliferation of MAC-T cells, we performed a combination of miRNA- 223 overexpression and inhibition strategies. We transfected MAC-T cells with miR- 223 mimics or inhibitors and evaluated the impact on cell proliferation using CCK- 8 assay, EdU assay, and RT-qPCR. Additionally, MAC-T cells subjected to heat stress were used to investigate how miR- 223 and its target gene regulate cell proliferation under heat stress, either by promoting or alleviating the inhibition of cell proliferation, as assessed by EdU assay, CCK- 8 assay, and RT-qPCR. RESULTS In this study, we investigated the effects of heat stress on MAC-T cell proliferation and gene expression. Bioinformatics analysis identified PRDM1 as a key regulator of proliferation, and it was selected for further investigation. RT-qPCR validated the upregulation of PRDM1 under heat stress, confirming its role in regulating cell proliferation. The results revealed that miR- 223 mimic promoted cell proliferation, with PRDM1 identified as its target gene. Importantly, after heat stress, the miR- 223 mimic or the knockdown of PRDM1 in MAC-T was proven to partially reverse the inhibition of proliferation. CONCLUSION Consequently, the miR- 223 targeting PRDM1 might be important in alleviating heat-stress-induced inhibition of cell proliferation. This would potentially alleviate heat stress-induced damage to the mammary gland, thereby improving milk production in dairy cows.
Collapse
Affiliation(s)
- Siqi Xia
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Huimei Fan
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Jianghai Xiao
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Ci Shen
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Yongping Yan
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Meigui Wang
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Tao Tang
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Wenqiang Sun
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Jie Wang
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Xianbo Jia
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Songjia Lai
- State Key Laboratory of Swine and Poultry Breeding Industrycollege of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China.
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, People's Republic of China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, People's Republic of China.
| |
Collapse
|
23
|
Kausar N. Machine learning and explainable artificial intelligence reveals the MicroRNAs associated with survival of head and neck squamous cell carcinoma patients. Comput Biol Chem 2025; 118:108503. [PMID: 40378655 DOI: 10.1016/j.compbiolchem.2025.108503] [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: 11/07/2024] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Dysregulated microRNAs (miRNAs) play a significant role in cancer development and metastasis. In literature, miRNAs have been used for the survival prediction of different types of cancers using AI. Although AI is useful for diagnosis and prognosis prediction of cancer, however, a major criticism of incorporating it into medical fields is that it is essentially a mechanistically uninterpretable opaque "black box", and hence it may not have the required level of accountability, transparency, and reliability in decisions of cancer diagnosis and prognosis for their adoption in clinical settings. Therefore, there is need to develop intelligent models which may explain their prediction so that they may be reliably used by the clinicians. As dysregulated miRNAs are reported to cause cancer metastasis hence, they can play role in survival of patient. Therefore, there is needed to develop ML based techniques which may automatically indicate specific miRNAs involved in survival of patients. In this research, Machine Learning and Explainable AI (XAI) based models have been developed for survival prediction of Head and Neck Squamous Cell Carcinoma (HNSC) patients using miRNA sequences and clinical datasets. miRNAs dataset contains the data of 485 HNSC patients and clinical dataset contains data of 528 patients. The proposed XAI based model explains its prediction by showing the specific miRNA sequences involved in survival of the patients to demonstrate its reliability to be used by clinicians for therapeutic decisions. In this study, it has been shown that explainable ML can provide explicit knowledge of how models make their predictions, which is necessary for increasing the trust and adoption of innovative ML techniques in oncology and healthcare.
Collapse
Affiliation(s)
- Nabeela Kausar
- Department of Software Engineering and Artificial Intelligence, Iqra University, Islamabad, Pakistan.
| |
Collapse
|
24
|
Martinelli C, Ercoli A, Vizzielli G, Burk SR, Cuomo M, Satasiya V, Kacem H, Braccia S, Mazzarotti G, Miriello I, Tchamou MN, Restaino S, Arcieri M, Poli A, Tius V, Parisi S, Pergolizzi S, Iatì G, Nibali CC, Pizzimenti C, Pepe L, Ieni A, Cortellino S, Giordano A. Liquid biopsy in gynecological cancers: a translational framework from molecular insights to precision oncology and clinical practice. J Exp Clin Cancer Res 2025; 44:140. [PMID: 40340939 PMCID: PMC12060497 DOI: 10.1186/s13046-025-03371-1] [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: 01/06/2025] [Accepted: 03/17/2025] [Indexed: 05/10/2025] Open
Abstract
Liquid biopsy offers a noninvasive method to identify and monitor tumor-derived biomarkers, including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, microRNAs, and tumor-educated platelets, that provide real-time insights into the biological behavior of gynecological cancers. The detection of these markers has the potential to revolutionize cancer management by enabling earlier detection, providing novel data to personalize treatments, and predicting disease recurrence before clinical imaging and predicting disease recurrence before clinical imaging can confirm progression, thereby also guiding complex clinical decision-making. However, because this new "omics" layer introduces additional complexity, it must be fully understood, from its biological rationale to technical development and clinical integration, to prevent confusion or misapplication. That is why, focusing on 14 critical fields of inquiry, our goal is to map the current state of liquid biopsy from bench to bedside while highlighting practical considerations for clinical integration. Each topic integrates recent advances in assay sensitivity, biomarker variability, and data interpretation, underscoring how standardized protocols and robust analytical methods are pivotal for reliable results. We then translate these findings into disease-specific insights, examining how liquid biopsy could refine early detection, minimal residual disease assessment, and therapy guidance in endometrial, cervical, and ovarian cancers. Although several FDA-approved assays and promising commercial tests illustrate the field's rapid evolution, many translational hurdles remain, including the need for harmonized protocols, larger prospective clinical trials, and cost-effectiveness analyses. Crucially, our synthesis clarifies the pivotal role of interdisciplinary collaboration. Oncologists, laboratory scientists, and industry partners must align on standardized procedures and clinically relevant endpoints. Without such coordination, promising biomarkers may remain confined to research settings, limiting their practical benefit. Taken together, our review offers a translational view designed to contextualize liquid biopsy in gynecological oncology.
Collapse
Affiliation(s)
- Canio Martinelli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Alfredo Ercoli
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Giuseppe Vizzielli
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Sharon Raffaella Burk
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Maria Cuomo
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Vrunda Satasiya
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Housem Kacem
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simone Braccia
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Giulio Mazzarotti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Irene Miriello
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
| | - Manuela Nana Tchamou
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Stefano Restaino
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Martina Arcieri
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Alice Poli
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Veronica Tius
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Giuseppe Iatì
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Chiara Conti Nibali
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Cristina Pizzimenti
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Ludovica Pepe
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Antonio Ieni
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Salvatore Cortellino
- Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Naples, Italy.
- Laboratory of Molecular Oncology, Research Hospital, Campobasso, 86100, Italy.
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy.
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA.
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
25
|
Jelski W, Okrasinska S, Mroczko B. microRNAs as Biomarkers of Breast Cancer. Int J Mol Sci 2025; 26:4395. [PMID: 40362631 PMCID: PMC12072494 DOI: 10.3390/ijms26094395] [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: 04/09/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Breast cancer (BC) is the most common type of cancer found in women. Detection of this cancer at an early stage is essential for effective treatment and a favorable prognosis. Potential early breast cancer biomarkers useful for diagnosing these tumors are microRNAs. These are small single-stranded RNA chains that can regulate the post-transcriptional expression of many different oncogenes. Cancer cells contain miRNAs that play a special role in the etiology of cancer development. The role of microRNAs in the initiation and development of breast cancer gives us great hope for the creation of molecular tools for early cancer detection. MicroRNAs are characterized by a high stability due to RNase, which protects them from degradation and enables their detection in various biological fluids. Researchers have described multiple serum microRNA signatures useful for detecting breast cancer. This review discusses the importance and potential usefulness of microRNAs in detecting breast cancer at an early stage, predicting the course of the disease, and assessing the effectiveness of treatment.
Collapse
Affiliation(s)
- Wojciech Jelski
- Department of Biochemical Diagnostics, Medical University, Waszyngtona 15 A, 15-269 Bialystok, Poland;
| | - Sylwia Okrasinska
- Department of Biochemical Diagnostics, University Hospital, Waszyngtona 15 A, 15-269 Bialystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University, Waszyngtona 15 A, 15-269 Bialystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University, Waszyngtona 15 A, 15-269 Bialystok, Poland
| |
Collapse
|
26
|
Li Z, Zhang T, Yang X, Peng Y. Role of noncoding RNA and protein interaction in pancreatic cancer. Chin Med J (Engl) 2025; 138:1019-1036. [PMID: 40205638 PMCID: PMC12068769 DOI: 10.1097/cm9.0000000000003587] [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: 01/01/2025] [Indexed: 04/11/2025] Open
Abstract
ABSTRACT Noncoding RNAs (ncRNAs) are a class of RNA molecules with little or no protein-coding potential. Emerging evidence indicates that ncRNAs are frequently dysregulated and play pivotal roles in the pathogenesis of pancreatic cancer. Their aberrant expression can arise from chromosomal abnormalities, dysregulated transcriptional control, and epigenetic modifications. ncRNAs function as protein scaffolds or molecular decoys to modulate interactions between proteins and other biomolecules, thereby regulating gene expression and contributing to pancreatic cancer progression. In this review, we summarize the mechanisms underlying ncRNA dysregulation in pancreatic cancer, emphasize the biological significance of ncRNA-protein interactions, and highlight their clinical relevance. A deeper understanding of ncRNA-protein interactions is essential to elucidate molecular mechanisms and advance translational research in pancreatic cancer.
Collapse
Affiliation(s)
- Zhang Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tingting Zhang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaojuan Yang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
27
|
Suri K, Hosur V, Panchakshari R, Amiji MM. A Multimodal Therapeutic Strategy for Inflammatory Bowel Disease Using MicroRNA-146a Mimic Encapsulated in Lipid Nanoparticles. Mol Pharm 2025. [PMID: 40324972 DOI: 10.1021/acs.molpharmaceut.5c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Dysregulated microRNAs (miRNAs) have significant potential as diagnostic tools for various chronic diseases; however, their therapeutic applications remain largely unexplored. Given their capacity to regulate multiple pathways, miRNAs are promising candidates for treating pleiotropic diseases, such as inflammatory bowel disease (IBD). In our study, we conducted a comprehensive review of the literature of miRNA-146 levels in the inflamed tissues of IBD patients and murine colitis models. Initially, we quantified the expression of miRNA-146a and miRNA-146b in the colons of mice using the dextran sodium sulfate (DSS)-inducedacute model of IBD. We selected miRNA-146a for further study due to its anti-inflammatory properties and potential relevance in IBD treatment. We hypothesized that a macrophage model of inflammation would be well-suited to studying the effects of this miRNA. Subsequently, we investigated the use of lipid nanoparticles (LNPs) for the targeted delivery of miRNA-146a to macrophages, which play a key role in IBD. Our results indicated that miRNA-146a levels increased in the DSS model and LNP-mediated delivery effectively downregulated genes associated with inflammation. These findings highlight the critical role of miRNA-146a in modulating IBD and suggest that LNP-based delivery could be a promising therapeutic strategy for managing inflammatory responses.
Collapse
Affiliation(s)
- Kanika Suri
- Takeda Development Center Americas, Cambridge, Massachusetts 02142, United States
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, Massachusetts 02120, United States
| | - Vishnu Hosur
- The Jackson Laboratory, Bar Harbor, Maine 04609, United States
| | - Rohit Panchakshari
- Takeda Development Center Americas, Cambridge, Massachusetts 02142, United States
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
28
|
Azar BKY, Vakhshiteh F. The Pre-metastatic Niche: How Cancer Stem Cell-Derived Exosomal MicroRNA Fit into the Puzzle. Stem Cell Rev Rep 2025; 21:1062-1074. [PMID: 40095238 DOI: 10.1007/s12015-025-10866-z] [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] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Cancer metastasis is a complicated biological process that critically affects cancer progression, patient outcomes, and treatment plans. A significant step in metastasis is the formation of a pre-metastatic niche (PMN). A small subset of cells within tumors, known as cancer stem cells (CSCs), possess unique characteristics including, differentiation into different cell types within the tumor, self-renewal, and resistance to conventional therapies, that enable them to initiate tumors and drive metastasis. PMN plays an important role in preparing secondary organs for the arrival and proliferation of CSCs, thereby facilitating metastasis. CSC-derived exosomes are crucial components in the complex interplay between CSCs and the tumor microenvironment. These exosomes function as transporters of various substances that can promote cancer progression, metastasis, and modulation of pre-metastatic environments by delivering microRNA (miRNA, miR) cargo. This review aims to illustrate how exosomal miRNAs (exo-miRs) secreted by CSCs can predispose PMN and promote angiogenesis and metastasis.
Collapse
Affiliation(s)
- Behjat Kheiri Yeghaneh Azar
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
29
|
Singh P, Chaturvedi R, Somvanshi P. Network-Based Integrative Analysis to Identify Key Genes and Corresponding Reporter Biomolecules for Triple-Negative Breast Cancer. Cancer Med 2025; 14:e70674. [PMID: 40287845 PMCID: PMC12034156 DOI: 10.1002/cam4.70674] [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: 09/23/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The malignant neoplasm of the TNBC is the leading cause of death among Indian women. Recent studies identified the global burden of TNBC affecting approximately more than 40 percent of all BC cases in women worldwide. The absence of expression of receptors such as ER, PR, and HER2 characterizes TNBC. OBJECTIVES Due to the lack of specific targets, standard treatment options for TNBC are limited. This integrative study aims to identify key genes and provide insights into the underlying molecular mechanisms of TNBC, which can potentially lead to the development of more effective therapeutic strategies. MATERIAL AND METHODOLOGY This study integrates PPI and WGCNA analysis of TNBC-related datasets (GSE52194 and GSE58135) to identify key genes. Subsequently, downstream analysis is conducted to explore potential therapeutic targets for TNBC. RESULTS The present study renders the potential 13 key genes (PLCG2, CXCL10, CDK1, STAT1, IL6, PLK1, CCNB1, AURKA, NDC80, EGFR, 1L1B, FN1, BUB1B), along with their associated 6 TFs and 20 miRNAs, as reporter biomolecules around which the most significant changes occur. There were some miRNAs hsa-mir-449b-5p, hsa-let-7b-5p, hsa-mir-26a-5p, hsa-mir-155-5p, hsa-mir-24-3p, hsa-mir-212-3p, hsa-mir-21-5p, hsa-mir-210-3p and hsa-mir-20a-5p whose association with other cancers and other BC subtypes have been reported but their association with TNBC need to be explored. Further, enrichment and cumulative survival analysis support the disease association of identified key genes with TNBC. CONCLUSION This integrative analysis could be regarded for experimental inspection as it provides the platform for future researchers in drug designing and biomarker discovery for TNBC diagnosis and treatment.
Collapse
Affiliation(s)
- Pooja Singh
- School of Computational & Sciences (SCIS)Jawaharlal Nehru UniversityNew DelhiIndia
| | - Rupesh Chaturvedi
- School of Biotechnology (SBT)Jawaharlal Nehru UniversityNew DelhiIndia
| | - Pallavi Somvanshi
- School of Computational & Sciences (SCIS)Jawaharlal Nehru UniversityNew DelhiIndia
| |
Collapse
|
30
|
Wang Y, Qi J, Ding D, Deng R, Jin F, Ren J, Di H. Biomimetic fluorescence-enhanced platform based on photonic crystals and DNAzyme walker for visualization and quantification of miRNA-21. Talanta 2025; 286:127440. [PMID: 39732101 DOI: 10.1016/j.talanta.2024.127440] [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: 10/25/2024] [Revised: 12/07/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Developing a fluorescence sensing platform for point-of-care detection of low abundance biomarkers is highly valuable for early diagnosis of disease. Herein, a biomimetic fluorescence-enhanced platform based on photonic crystals and DNAzyme walker was constructed and further applied to visualize and quantify the miRNA-21 in biological samples. The DNAzyme walker was orthogonally activated by the target miRNA-21, which enabled the unlocking of the DNAzyme walker strand and the subsequently repeated substrate cleavage, thus generating enhanced fluorescence signals. Meanwhile, a biomimetic photonic crystals substrate was employed to physically boost the emitted fluorescence signals again, enabling visual detection by naked eyes and quantitative analysis by smartphone. A limit of detection as low as 84.8 pM was obtained and the single base variation in miRNA-21 could also be discriminated. Furthermore, the potential application of this platform was demonstrated in spiked-urine sample and total RNA extracts, showing good agreement with RT-qPCR results. Therefore, the multiple signal amplification strategy-based fluorescent platform has the advantages of high sensitivity, good specificity, good portability and high-throughput analysis, providing a powerful tool for convenient disease diagnosis and health assessment.
Collapse
Affiliation(s)
- Yingqian Wang
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China.
| | - Jia'en Qi
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| | - Dianxing Ding
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| | - Rong Deng
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| | - Fujing Jin
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| | - Jiamin Ren
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| | - Huixia Di
- Institute of Biomedical Precision Testing and Instrumentation, College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, Shanxi, 030600, PR China
| |
Collapse
|
31
|
Ramilo Amor A, Enlund S, Sinha I, Jiang Q, Hermanson O, Nilsson A, Shirazi Fard S, Holm F. A distinct alternative mRNA splicing profile identifies the oncogenic CD44 transcript variant 3 in KMT2A-rearranged pediatric T-cell acute lymphoblastic leukemia cells. Exp Hematol 2025; 145:104712. [PMID: 39793727 DOI: 10.1016/j.exphem.2025.104712] [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: 09/04/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 01/13/2025]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL), which constitutes of 10-15% of all pediatric acute lymphoblastic leukemia (ALL) cases, is known for its complex pathology due to pervasive genetic and chromosomal abnormalities. Although most children are successfully cured, chromosomal rearrangements involving the KMT2A gene is considered a poor prognostic factor. In a cohort of 171 pediatric T-ALL samples, we have studied differences in gene and splice variant patterns in KMT2A-rearranged (KMT2A-r) T-ALL compared with KMT2A-negative (KMT2A-wt) T-ALL samples. Our results have identified a distinct gene expression and splice variant expression pattern in pediatric KMT2A-r patient samples including significant expression of splicing regulatory markers ESRP1 and MBNL3. Additionally, the prosurvival long transcript variant of BCL2 were upregulated in KMT2A-r compared with KMT2A-wt T-ALL samples. Lastly, increased levels of activating methylation in the promoter region of CD44 were identified followed by an upregulation of the oncogenic transcript variant CD44v3 in KMT2A-r T-ALL. Together, this suggests that CD44v3 could play a potential role as gene expression-based risk stratification of KMT2A-r T-ALL and could possibly serve as a therapeutic target using splicing modulators.
Collapse
Affiliation(s)
- Amanda Ramilo Amor
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sabina Enlund
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Indranil Sinha
- Department of Molecular Biosciences, Wenner-Gren Institutet, Stockholm University, Stockholm, Sweden
| | - Qingfei Jiang
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Nilsson
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shahrzad Shirazi Fard
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Frida Holm
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
32
|
Sharma R, Yadav J, Bhat SA, Musayev A, Myrzagulova S, Sharma D, Padha N, Saini M, Tuli HS, Singh T. Emerging Trends in Neuroblastoma Diagnosis, Therapeutics, and Research. Mol Neurobiol 2025; 62:6423-6466. [PMID: 39804528 DOI: 10.1007/s12035-024-04680-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025]
Abstract
This review explores the current understanding and recent advancements in neuroblastoma, one of the most common extracranial solid pediatric cancers, accounting for ~ 15% of childhood cancer-related mortality. The hallmarks of NBL, including angiogenesis, metastasis, apoptosis resistance, cell cycle dysregulation, drug resistance, and responses to hypoxia and ROS, underscore its complex biology. The tumor microenvironment's significance in disease progression is acknowledged in this study, along with the pivotal role of cancer stem cells in sustaining tumor growth and heterogeneity. A number of molecular signatures are being studied in order to better understand the disease, with many of them serving as targets for the development of new therapeutics. This includes inhibitor therapies for NBL patients, which notably concentrate on ALK signaling, MDM2, PI3K/Akt/mTOR, Wnt, and RAS-MAPK pathways, along with regulators of epigenetic mechanisms. Additionally, this study offers an extensive understanding of the molecular therapies used, such as monoclonal antibodies and CAR-T therapy, focused on both preclinical and clinical studies. Radiation therapy's evolving role and the promise of stem cell transplantation-mediated interventions underscore the dynamic landscape of NBL treatment. This study has also emphasized the recent progress in the field of diagnosis, encompassing the adoption of artificial intelligence and liquid biopsy as a non-intrusive approach for early detection and ongoing monitoring of NBL. Furthermore, the integration of innovative treatment approaches such as CRISPR-Cas9, and cancer stem cell therapy has also been emphasized in this review.
Collapse
Affiliation(s)
- Rishabh Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Jaya Yadav
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Sajad Ahmad Bhat
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
- Department of Biochemistry, NIMS University, Rajasthan, Jaipur, 303121, India
| | - Abdugani Musayev
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
| | | | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Nipun Padha
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Department of Zoology, Cluster University of Jammu, Jammu, 180001, India
| | - Manju Saini
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India.
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, (INMAS-DRDO), New Delhi, Delhi, 110054, India.
| |
Collapse
|
33
|
Rodemann MM, Dreschmann V, Dörner E, Sommer A, Kraetzschmar J, Klein-Hitpass L, Nagae G, Hiyama E, von Schweinitz D, Kappler R, Vokuhl C, Pietsch T. Identification of a Growth-Promoting Gene Cluster in the Region 2q24 as a Driver of Tumorigenesis in Childhood Hepatoblastoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00147-6. [PMID: 40316217 DOI: 10.1016/j.ajpath.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 05/04/2025]
Abstract
Hepatoblastoma (HB) represents the most common primary malignancy of the liver in childhood. Cytogenetic studies uncovered characteristic copy number alterations in HB. The frequent gain of chromosome 2q and particularly the recurrent 2q24 amplification suggest the presence of a so far unidentified oncogenic driver within this amplicon. High-resolution copy number profiles from 76 patients with HB were generated by using molecular inversion probe array technology. 2q gain was present in 63.2%, and 2q24 high-gain/amplification was present in 14.5% of patients analyzed. In the smallest overlapping region at 2q24.2q24.3, spanning >5.2 Mbp, 22 protein-coding genes, 2 long noncoding RNA genes, and one miRNA gene were mapped. RNA expression analysis of these smallest overlapping region genes identified RBMS1, BAZ2B, MARCH7, DPP4, FIGN, and TANK as overexpressed in 2q24 high-gain/amplified HB cases. Accordingly, these six genes were selected for further investigation. In situ, immunohistochemical staining showed higher protein expression of these genes in 2q24 high-gain HB tissue sections. In vitro, functional analyses were performed in established human HB cell lines carrying a 2q (high-)gain. Knockdown of these genes by specific siRNAs resulted in reduced proliferation and marked reduction of Wnt pathway activity. These genes located within the 2q24 amplicon might collaborate in driving cellular growth by interaction with the Wnt pathway that is known to be activated pathologically in HB.
Collapse
Affiliation(s)
- Martin M Rodemann
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Verena Dreschmann
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Evelyn Dörner
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | | | | | | | - Genta Nagae
- Genome Science Laboratory, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Eiso Hiyama
- Department of Pediatric Surgery, Hiroshima University Hospital, Hiroshima, Japan; Department of Biomedical Science, Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | | | - Roland Kappler
- Department of Pediatric Surgery, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Christian Vokuhl
- Pediatric Pathology, Department of Pathology, University of Bonn Medical Center, Bonn, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany.
| |
Collapse
|
34
|
Ali FEM, Badran KSA, El-Maksoud MSA, Ibrahim IM, Althagafy HS, Hassanein EHM. The role of Wnt/β-catenin signaling in lung cancer progression and therapy: a comprehensive review. Med Oncol 2025; 42:183. [PMID: 40289194 DOI: 10.1007/s12032-025-02709-1] [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: 12/20/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Most instances of lung cancer (LC), which is the primary cause of cancer-related death worldwide, are non-small-cell lung cancer (NSCLC). Genetic predispositions, environmental exposures, and smoking are risk factors that lead to the development of LC, and the ineffectiveness of existing treatments emphasizes the need for innovative approaches to therapy. Through its regulation of cell proliferation, apoptosis, epithelial-to-mesenchymal transition (EMT), and cancer stem cell maintenance, the Wnt/β-catenin signaling system is essential to advancing LC. This study offers a thorough examination of Wnt/β-catenin signaling in LC, emphasizing how miRNAs, long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), protein-coding genes, enzymes, and both natural and synthetic drugs affect this signaling. Recent research supports the dual function of Wnt/β-catenin signaling in tumor development and repression, which we describe. We also emphasize the therapeutic potential of Wnt/β-catenin inhibitors despite issues including off-target effects and bioavailability. This study highlights the potential of focusing on Wnt/β-catenin signaling to enhance LC patient outcomes by combining computational studies with molecular insights. It also lays the groundwork for further research and treatment development.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt.
- Faculty of Pharmacy, Michael Sayegh, Aqaba University of Technology, Aqaba, 77110, Jordan.
| | - Khalid S A Badran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| | - Mostafa S Abd El-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| |
Collapse
|
35
|
Bamfield-Cummings S, Silva J, Karim ZA. A thematic analysis of prognostic, diagnostic, and therapeutic of circulating miRNA biomarkers in bortezomib-resistant multiple myeloma. SAGE Open Med 2025; 13:20503121251328486. [PMID: 40297788 PMCID: PMC12035079 DOI: 10.1177/20503121251328486] [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: 08/22/2024] [Accepted: 03/03/2025] [Indexed: 04/30/2025] Open
Abstract
Objective The increasing demand for precision medicine has spurred molecular diagnostic investigations to emphasize the utility of miRNA as significant biomarkers. Recent studies have underscored miRNA's role as prognostic, diagnostic, and therapeutic biomarkers in managing and monitoring multiple myeloma patients. This review aims to present the latest insights on the potential of circulating miRNA as prognostic, diagnostic, and therapeutic biomarkers in bortezomib-resistant multiple myeloma. Methods For this purpose, a comprehensive thematic literature review from January 2014 and August 2024 was conducted utilizing the databases CINAHL, Pubmed, and Google Scholar. Twenty pertinent studies were meticulously analyzed and categorized into the following sections: Bortezomib (BTZ) resistance in multiple myeloma, the predictive role of miRNAs in BTZ resistance, the impact of circulating miRNAs in multiple myeloma, and the potential of circulating miRNA as prognostic, diagnostic, and therapeutic biomarkers. Results Of note, eight studies identified circulating miRNAs as diagnostic miRNA biomarkers (i.e., miR-744, miR-130a, let-7d, let-7e, miR-34a, etc.). In comparison, nine studies identified several circulating miRNAs that can be used as prognostic biomarkers (i.e., miR-20a, miR-483-5p, mir-1246, let-7a, let-7e, etc.). Moreover, five studies identified circulating miRNAs as promising therapeutic biomarkers (i.e., mir-15a, mir-92a, mir-19a, etc.). This discovery can significantly enhance early detection, accurate diagnosis, prognosis, overall survival rates, and quality of life for patients with multiple myeloma. Conclusion Based on this evidence, exploring circulating miRNAs as a potential noninvasive biomarker for multiple myeloma represents a noteworthy advancement. This is attributed to the abundance of miRNAs in plasma or serum, which exhibits remarkable stability against enzymatic degradation.
Collapse
Affiliation(s)
| | - Jeane Silva
- Department of Health Management, Economics, and Policy, Augusta University, GA, USA
| | - Zubair A. Karim
- Department of Nutrition and Dietetics, College of Allied Health Science, Augusta University, GA, USA
| |
Collapse
|
36
|
Wu H, Ling X, Huang S, Zhao Q, Zhang D, Wang H. Direct Fluorescence Anisotropy Detection of miRNA Based on Duplex-Specific Nuclease Signal Amplification. Anal Chem 2025; 97:8574-8580. [PMID: 40205322 DOI: 10.1021/acs.analchem.5c00723] [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: 04/11/2025]
Abstract
The dysregulation of microRNAs (miRNAs) is associated with various diseases, including cancer, so miRNAs are considered a potential biomarker candidate for disease diagnosis and therapy. However, the direct, rapid, sensitive, and specific detection of miRNAs remains quite challenging due to their short length, sequence homology, and low abundance. Herein, we propose a simple and homogeneous fluorescence anisotropy (FA) strategy for the direct and rapid (∼35 min) quantification of miRNA-21 based on duplex-specific nuclease (DSN)-assisted signal amplification. In the presence of target miRNA-21, the complementary single-stranded DNA (ssDNA) probes labeled with a single fluorophore, tetramethylrhodamine (TMR), are specifically hydrolyzed into small fragments by endonuclease DSN upon formation of the DNA/RNA hybrid, which leads to a reduction in FA due to the decrease in molecular size. However, the target miRNA remains intact during the enzymatic digestion process and is released in solution for the next round of binding, hydrolysis, and release for recycling. It is observed that the ssDNA probe labeled with TMR at the 5'-end, in which the fluorophore is nine nucleotides away from the nearest dG base to eliminate/reduce photoinduced electron transfer interaction between TMR and the dG base, exhibits the maximum FA change in response to the target miRNA-21. The change in FA enables the sensitive detection of miRNA-21 ranging from 0.050 to 2.0 nM, with a detection limit of 40 pM. In addition, this amplification strategy exhibits high selectivity and can even discriminate single-base mutations between miRNA family members. We further applied this method to detect miRNA-21 in the extract of various cancer cell lines. Therefore, this method holds great potential for miRNA analysis in tissues or cells, providing valuable information for biomedical research, clinical diagnostics, and therapeutic applications.
Collapse
Affiliation(s)
- Huilan Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoting Ling
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shoulong Huang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiang Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dapeng Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hailin Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- College of Resource and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
37
|
Bresesti C, Carito E, Notaro M, Giacca G, Breggion S, Kerzel T, Mercado CM, Beretta S, Monti M, Merelli I, Canu T, Naldini L, Squadrito ML. Reprogramming liver metastasis-associated macrophages toward an anti-tumoral phenotype through enforced miR-342 expression. Cell Rep 2025; 44:115592. [PMID: 40253698 DOI: 10.1016/j.celrep.2025.115592] [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: 08/13/2024] [Revised: 01/24/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025] Open
Abstract
Upon metastatic seeding in the liver, liver macrophages, including Kupffer cells, acquire a transcriptional profile typical of tumor-associated macrophages (TAMs), which support tumor progression. MicroRNAs (miRNAs) fine-tune TAM pro-tumoral functions, making their modulation a promising strategy for macrophage reprogramming into an anti-tumoral phenotype. Here, we analyze the transcriptomic profiles of liver and splenic macrophages, identifying miR-342-3p as a key regulator of liver macrophage function. miR-342-3p is highly active in healthy liver macrophages but significantly downregulated in colorectal cancer liver metastases (CRLMs). Lentiviral vector-engineered liver macrophages enforcing miR-342-3p expression acquire a pro-inflammatory phenotype and reduce CRLM growth. We identify Slc7a11, a cysteine-glutamate antiporter linked to pro-tumoral activity, as a direct miR-342-3p target, which may be at least partially responsible for TAM phenotypic reprogramming. Our findings highlight the potential of in vivo miRNA modulation as a therapeutic strategy for TAM reprogramming, offering an approach to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Chiara Bresesti
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Eleonora Carito
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Marco Notaro
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giovanna Giacca
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Sara Breggion
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Thomas Kerzel
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Carl Mirko Mercado
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Beretta
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Monti
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ivan Merelli
- BioInformatics Core, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Tamara Canu
- Preclinical Imaging Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luigi Naldini
- Vita-Salute San Raffaele University, 20132 Milan, Italy; Targeted Cancer Gene Therapy Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mario Leonardo Squadrito
- Vector Engineering and In vivo Tumor Targeting Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy.
| |
Collapse
|
38
|
Akand SK, Rahman A, Masood M, Tabrez S, Saleem M, Ahmed MZ, Akhter Y, Haque MM, Rub A. hsa-miR-330-5p regulates serine palmitoyltransferase long chain base subunit 1 and augments host protective immune response against Leishmania donovani infection. Arch Microbiol 2025; 207:123. [PMID: 40237871 DOI: 10.1007/s00203-025-04325-z] [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: 01/16/2025] [Revised: 03/09/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Leishmaniasis, caused by the protozoan parasites of the genus Leishmania, poses a significant global health challenge, particularly in the resource-limited regions where it causes high mortality. Regardless in the progress of treatment strategies, the emergence of drug resistance and limited efficacy requires the search of novel therapy and therapeutic targets. MicroRNAs, the crucial post-transcriptional regulators of gene expression, play critical roles in host-pathogen interactions. Here, we screened the miRNAs dysregulated during Leishmania donovani infection through literature search. hsa-miR-330-5p, one of the miRNAs which through human KEGG 2021 and Human Cyc 2016 analysis was found to be involved in multiple pathways including sphingolipid signaling pathway. Sphingolipids are important class of lipids involved in different cellular processes and therefore are the targets of many pathogens including Leishmania. hsa-miR-330-5p was found downregulated after 24 h of Leishmania donovani infection in THP-1 derived human macrophages. Target prediction of sphingolipid biosynthetic genes through in silico prediction tools showed 3/ UTR of serine palmitoyltransferase long chain base subunit 1 to be a target of hsa-miR-330-5p. The in silico target prediction of hsa-miR-330-5p was validated by cloning the 3/ UTR target sequence of gene, transfecting and performing luciferase assay in HEK 293 T cell line. Transfection of mimic of hsa-miR-330-5p reduced the luciferase activity which validated the in silico target prediction. Further, mimic of hsa-miR-330-5p inhibited the expression of the target gene, serine palmitoyltransferase long chain base subunit 1 and augmented the expression of pro-inflammatory cytokines in L. donovani infected THP-1 derived macrophages. Mimic of hsa-miR-330-5p also led to a significant reduction in the intracellular parasite burden in both THP-1 derived as well as primary human macrophages. This study has not only identified the sphingolipid biosynthesis regulatory miRNA but will also help in the development of novel and effective treatment strategy against leishmaniasis in future.
Collapse
Affiliation(s)
- Sajjadul Kadir Akand
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Areeba Rahman
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Mohammad Masood
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Shams Tabrez
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - Mohammad Saleem
- Faculty of Dentistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Mohammad Z Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Kingdom of Saudi Arabia
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Mohammad Mahfuzul Haque
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India
| | - Abdur Rub
- Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| |
Collapse
|
39
|
Liu W, Wang Y, Peng W, Zeng X, Jiang P, Xiao W, Chen J, Chen P. Cu 2+-encapsulated DNA nanosphere and filter enable sensitive and rapid analysis of miRNA-155. Biosens Bioelectron 2025; 274:117203. [PMID: 39889434 DOI: 10.1016/j.bios.2025.117203] [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: 12/12/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/03/2025]
Abstract
MicroRNAs (miRNAs) are critical regulators of gene expression, with aberrant levels linked to diseases such as breast cancer. Notably, they are challenging to detect due to their low abundance in complex sample matrices. In this study, a Cu2⁺-encapsulated DNA nanosphere system capable of homogeneous, enzyme-free, one-pot detection of microRNA-155 (miRNA-155) in human plasma was developed. The system employed self-assembled DNA nanospheres loaded with copper ions as specific recognizers, coupled with a filter membrane-assisted reaction to separate free-Cu2+ from residual components. Moreover, dual quantum dots (QDs) were utilized for signal output, using competitive binding to enhance sensitivity for detecting ultra-low and subtle changes in miRNA levels in complex samples. The method achieved an excellent detection performance, with a limit of detection (LOD) at the low-aM level. Additionally, it demonstrated high specificity in distinguishing between different miRNAs and single nucleotide polymorphisms (SNPs). Validation using 38 clinical plasma samples achieved over 95% accuracy in identifying breast cancer patients, demonstrating 100% sensitivity and approximately 90% clinical consistency compared to imaging, pathology, and quantitative real-time polymerase chain reaction (qRT-PCR). The method required minimal sample pre-treatment and was completed within 1 h. Overall, the developed method offers a reliable tool for breast cancer diagnosis and establishes a mode for extending its application to other biomarkers in the clinical setting.
Collapse
Affiliation(s)
- Weijing Liu
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Breast Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wu Peng
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xianghu Zeng
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Pengjun Jiang
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wei Xiao
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510632, China.
| | - Jie Chen
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Breast Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Piaopiao Chen
- Department of Laboratory Medicine, Med+X Center for Manufacturing, Department of General Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
40
|
Ribeiro AL, Dallagiovanna B. The Role of Long Non-Coding RNAs in Human Endoderm Differentiation. Noncoding RNA 2025; 11:29. [PMID: 40278506 PMCID: PMC12029278 DOI: 10.3390/ncrna11020029] [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: 02/26/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The human genome sequencing revealed a vast complexity of transcripts, with over 80% of the genome being transcribed into non-coding RNAs. In particular, long non-coding RNAs (lncRNAs) have emerged as critical regulators of various cellular processes, including embryonic development and stem cell differentiation. Despite extensive efforts to identify and characterize lncRNAs, defining their mechanisms of action in state-specific cellular contexts remains a significant challenge. Only recently has the involvement of lncRNAs in human endoderm differentiation of pluripotent stem cells begun to be addressed, creating an opportunity to explore the mechanisms by which lncRNAs exert their functions in germ layer formation, lineage specification, and commitment. This review summarizes current findings on the roles of lncRNAs in endoderm differentiation, highlighting the functional mechanisms and regulatory aspects underlying their involvement in cell fate decisions leading to endoderm development. The key lncRNAs implicated in endoderm differentiation are discussed, along with their interaction with transcription factors and RNA-binding proteins and modulation of signaling pathways essential for endoderm development. Gaining insight into the regulatory roles of lncRNAs in endoderm differentiation enhances the understanding of developmental biology and provides a foundation for discovering novel lncRNAs involved in cell fate determination.
Collapse
Affiliation(s)
| | - Bruno Dallagiovanna
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute—FIOCRUZ/PR, Curitiba 81350-010, Brazil;
| |
Collapse
|
41
|
Magazova A, Ashirbekov Y, Abaildayev A, Satken K, Utegenova G, Belkozhayev A, Balmukhanova A, Dzhumatayeva Z, Beissova A, Shargorodska I, Balmukhanova A, Sharipov K. Circulating microRNAs demonstrate limited diagnostic potential for diabetic retinopathy in the population of Kazakhstan. PeerJ 2025; 13:e19259. [PMID: 40231069 PMCID: PMC11995893 DOI: 10.7717/peerj.19259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Background Diabetic retinopathy (DR) is the most common complication of diabetes, leading to blindness. The asymptomatic onset and the existing difficulties in diagnosing warrant the search for biomarkers that can facilitate the early diagnosis of DR. The aim of this study was to evaluate the potential of plasma microRNAs (miRNAs), which have previously been shown to be involved in the pathogenesis of DR and differentially expressed in plasma/serum of patients, as biomarkers for DR in the Kazakhstani population. Materials and Methods Using quantitative RT-PCR, we compared the levels of ten candidate miRNAs in plasma among three groups: type 2 diabetes mellitus (T2DM) patients with DR (DR patients, N = 100), T2DM patients without DR (noDR patients, N = 98), and healthy controls (N = 30). Results Level of miR-423-3p was significantly reduced in DR patients compared to noDR patients (pFDR = 5.4 × 10-3). Levels of miR-423-3p and miR-221-3p were significantly reduced in DR patients compared to controls (pFDR = 5.4 × 10-3 and 0.024, respectively ), level of miR-23a-3p was significantly reduced in noDR patients compared to controls (pFDR = 0.047), levels of miR-221-3p and miR-23a-3p were significantly reduced in T2DM patients (combined group) compared to controls (pFDR = 0.047, and 0.049, respectively). Also, there were several significant differences between groups formed based on clinical-pathological characteristics, but none of these results remained significant after adjustment for multiple comparisons. Correlation analysis revealed weak associations between the levels of miR-423 and miR-221-3p and DR staging (pFDR = 1.3 × 10-3 and 0.026, respectively), and fair associations between the levels of miR-29b-3p and miR-328-3p and diabetes duration in noDR patients (pFDR = 8.8 × 10-3 and 0.016, respectively). According to receiver operating characteristic (ROC) analysis, only miR-23a-3p can be considered a potential biomarker with moderate informativeness for diagnosing proliferative DR (PDR); however, a larger sample size is needed to verify this finding. Furthermore, the small magnitude of observed changes in miRNA levels between groups significantly complicates classification. Conclusions Due to the low specificity and small magnitude of deviations from the norm, the studied miRNAs have low potential in the diagnosis of DR.
Collapse
Affiliation(s)
- Aizhan Magazova
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Almaty Multidisciplinary Clinical Hospital, Almaty, Kazakhstan
- Department of Ophthalmology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Yeldar Ashirbekov
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Arman Abaildayev
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Kantemir Satken
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Gulzhakhan Utegenova
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Department of Biology, South Kazakhstan Pedagogical University named after Ozbekali Zhanibekov, Shymkent, Kazakhstan
| | - Ayaz Belkozhayev
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Department of Chemical and Biochemical Engineering, Geology and Oil-Gas Business Institute named after K. Turyssov, Satbayev University, Almaty, Kazakhstan
| | - Altynay Balmukhanova
- Department of Health Policy and Organization, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Zaure Dzhumatayeva
- Kazakh Scientific Research Institute of Eye Diseases, Almaty, Kazakhstan
| | - Ainagul Beissova
- Department of Public Health, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Iryna Shargorodska
- Ophthalmology and Optometry department of Postgraduate Education, Bogomolets National Medical University, Kyiv, Ukraine
| | | | - Kamalidin Sharipov
- Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Department of Biochemistry, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| |
Collapse
|
42
|
Wiśnik A, Jarych D, Krawiec K, Strzałka P, Potocka N, Czemerska M, Sałagacka-Kubiak A, Pluta A, Wierzbowska A, Zawlik I. Role of MicroRNAs in Acute Myeloid Leukemia. Genes (Basel) 2025; 16:446. [PMID: 40282406 PMCID: PMC12026923 DOI: 10.3390/genes16040446] [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: 03/07/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
MicroRNA (miRNA), a significant class of regulatory non-coding RNA (ncRNA), can regulate the expression of numerous protein-coding messenger RNAs (mRNAs). miRNA plays an important part in shaping the human transcriptome. So far, in the human genome, about 2500 miRNAs have been found. Acute myeloid leukemia (AML) belongs to a malignant clonal disorder of hematopoietic stem cells and is characterized by the uncontrolled clonal proliferation of abnormal progenitor cells in the bone marrow and blood. For the past several years, significant scientific attention has been attracted to the role of miRNAs in AML, since alterations in the expression levels of miRNAs may contribute to AML development. This review describes the main functions of non-coding RNA classes and presents miRNA biogenesis. This study aims to review recent reports about altered microRNA expression and their influence on AML cell survival, cell cycle, and apoptotic potential. Additionally, it summarizes the correlations between miRNAs and their target mRNAs in AML and outlines the role of particular miRNAs in AML subtypes according to ELN recommendations.
Collapse
Affiliation(s)
- Aneta Wiśnik
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | - Dariusz Jarych
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 93-232 Lodz, Poland
| | - Kinga Krawiec
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | - Piotr Strzałka
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | - Natalia Potocka
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Magdalena Czemerska
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | | | - Agnieszka Pluta
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | - Agnieszka Wierzbowska
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
- Copernicus Memorial Multi-Specialist Oncology and Trauma Center, 93-510 Lodz, Poland
| | - Izabela Zawlik
- Laboratory of Molecular Biology, Centre for Innovative Research in Medical and Natural Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland
- Department of General Genetics, Faculty of Medicine, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland
| |
Collapse
|
43
|
Nair M, Samidurai A, Das A, Kakar SS, Kukreja RC. Ovarian cancer and the heart: pathophysiology, chemotherapy-induced cardiotoxicity, and new therapeutic strategies. J Ovarian Res 2025; 18:72. [PMID: 40188339 PMCID: PMC11971845 DOI: 10.1186/s13048-025-01636-z] [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: 01/05/2025] [Accepted: 02/21/2025] [Indexed: 04/07/2025] Open
Abstract
Ovarian Cancer (OC) is recognized as the most lethal gynecologic malignancy, characterized by numerous genetic mutations that trigger uncontrolled cellular growth and replication. Emerging evidence suggests that non-coding RNAs including miRNAs and lncRNAs significantly influence OC through their multiple roles including tumor initiation, progression, metastasis, immune evasion, and chemoresistance, making them promising diagnostic markers and therapeutic targets. The primary approach to treating OC typically involves cytoreductive surgery followed by chemotherapy. However, the chemotherapeutic agents, particularly the anthracyclines such as doxorubicin (DOX), are known for their cardiotoxic effects, which can range from acute to chronic, potentially leading to heart failure and death. To enhance the overall treatment response and to minimize cardiotoxicity, alternative strategies have been explored. These include the use of liposomal doxorubicin (DOXIL) as a substitute for DOX, various radiotherapies, immunotherapies, and the co-administration of angiotensin-converting enzyme inhibitors and/or beta-blockers. Phosphodiesterase-5 inhibitors (PDE5i) have also demonstrated efficacy in reducing cardiotoxicity linked to cancer treatments and in promoting apoptosis in cancer cells across multiple cancer types. Although there is no current clinical trial directly examining the impact of PDE5i on reducing cardiotoxicity in OC, however emerging therapies such as Withaferin A, PARP inhibitors, and nanoparticle combination therapy show promise. Additional research is essential to develop treatments that are both effective against OC and less harmful to the heart.
Collapse
Affiliation(s)
- Megha Nair
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Arun Samidurai
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Anindita Das
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville, Louisville, KY, USA
| | - Rakesh C Kukreja
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
44
|
Chen YC, Bazewicz CG, Dinavahi SS, Huntington ND, Schell TD, Robertson GP. Emerging Role of the p53 Pathway in Modulating NK Cell-Mediated Immunity. Mol Cancer Ther 2025; 24:523-535. [PMID: 39470047 DOI: 10.1158/1535-7163.mct-24-0325] [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: 05/10/2024] [Revised: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
The p53 pathway plays an important role in role in cancer immunity. Mutation or downregulation of the proteins in the p53 pathway are prevalent in many cancers, contributing to tumor progression and immune dysregulation. Recent findings suggest that the activity of p53 within tumor cells, immune cells, and the tumor microenvironment can play an important role in modulating NK cell-mediated immunity. Consequently, efforts to restore p53 pathway activity are being actively pursued to modulate this form of immunity. This review focuses on p53 activity regulating the infiltration and activation of NK cells in the tumor immune microenvironment. Furthermore, the impact of p53 and its regulation of NK cells on immunogenic cell death within solid tumors and the abscopal effect are reviewed. Finally, future avenues for therapeutically restoring p53 activity to improve NK cell-mediated antitumor immunity and optimize the effectiveness of cancer therapies are discussed.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Christopher G Bazewicz
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- oNKo-Innate Pty Ltd. Moonee Ponds, Victoria, Australia
| | - Todd D Schell
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
45
|
Tulimilli SV, Karnik M, Bettadapura ADS, Sukocheva OA, Tse E, Kuppusamy G, Natraj SM, Madhunapantula SV. The tumor suppressor role and epigenetic regulation of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in cancer and tumor microenvironment (TME). Pharmacol Ther 2025; 268:108826. [PMID: 39971253 DOI: 10.1016/j.pharmthera.2025.108826] [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: 10/16/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Oxidative stress and inflammation may initiate carcinogenesis and facilitate metastasis via activation of pro-inflammatory signaling network. The side product of arachidonic acid processing by cyclooxygenase-2 (COX-2), the prostaglandin E2 (PGE2), plays a key role in various metabolic disorders and during inflammation-mediated tumorigenesis. It has been demonstrated that PGE2 increases the proliferation, migration, invasion, metastasis, and resistance of cancer cells to apoptosis and other forms of programmed cell death. The expression level of PGE2 metabolizing enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is often decreased in various malignancies. However, the role of 15-PGDH and PGE2 in the regulation of carcinogenesis remains controversial. Numerous cancer cell lines and mouse models have demonstrated the role of 15-PGDH as a tumor suppressor. Downregulation of 15-PGDH increased cancer cell proliferation, migration, anchorage independent growth, colony formation while overexpression reversed these effects, by inducing apoptosis and cell cycle arrest in vitro and in vivo. The expression of 15-PGDH is regulated by various mechanisms, including (a) epigenetic alterations (methylation of promoter region, histone deacetylases, microRNAs (miR-21, miR-26a/b, miR-106b-5p, miR-146b-3p, miR-155, miR-218-5p, and miR-620)); and (b) dysregulated oxidative stress and associated mediators (elevated levels of growth factors and proinflammatory cytokines (such as IL1β and TNFα)). Several transcription factors, such as HNF3β, β-catenin, Snail, Slug, can bind to 15-PGDH promoter region and downregulate the enzyme expression. In contrast, the expression of 15-PGDH can be upregulated by several anti-inflammatory cytokines and anti-cancer agents, such as IL10 and vitamin D. The functional activity of 15-PGDH protein can be modulated by signaling effectors and oxidative stress, including increased production of reactive oxygen species (ROS). However, the role of oxidative stress regulator protein, i.e., nuclear factor erythroid 2-related factor 2 (Nrf2), in the control of 15-PGDH expression remains unclear. This article provides insights and comprehensive overview of the tumor suppressor role of 15-PGDH in various cancers. Epigenetic and post-translational mechanisms regulating 15-PGDH expression and the role of novel ROS-Nrf2-15-PGDH axis were discussed and accented as potential drug targets.
Collapse
Affiliation(s)
- SubbaRao V Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST supported center and ICMR Collaborating Center of Excellence - ICMR-CCoE), Department of Biochemistry (DST-FIST supported department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Medha Karnik
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST supported center and ICMR Collaborating Center of Excellence - ICMR-CCoE), Department of Biochemistry (DST-FIST supported department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Anjali Devi S Bettadapura
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST supported center and ICMR Collaborating Center of Excellence - ICMR-CCoE), Department of Biochemistry (DST-FIST supported department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Olga A Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, CALHN, Port Rd, Adelaide, SA 5000, Australia.
| | - Edmund Tse
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, CALHN, Port Rd, Adelaide, SA 5000, Australia.
| | - Gowthamarajan Kuppusamy
- Department of Pharmaceutics (DST-FIST supported department), JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Ooty, Nilgiris, Tamil Nadu, India.
| | - Suma M Natraj
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST supported center and ICMR Collaborating Center of Excellence - ICMR-CCoE), Department of Biochemistry (DST-FIST supported department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - SubbaRao V Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST supported center and ICMR Collaborating Center of Excellence - ICMR-CCoE), Department of Biochemistry (DST-FIST supported department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India; Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| |
Collapse
|
46
|
Arun B, John G, Raman R. MicroRNA Signatures: Illuminating Minimal Residual Disease Monitoring in Juvenile Myelomonocytic Leukemia - A Review. J Hematol 2025; 14:43-55. [PMID: 40336920 PMCID: PMC12056752 DOI: 10.14740/jh1384] [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: 11/22/2024] [Accepted: 01/20/2025] [Indexed: 05/09/2025] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is an aggressive pediatric myelodysplastic/myeloproliferative neoplasm characterized by RAS pathway mutations and significant heterogeneity. Minimal residual disease (MRD) monitoring is crucial for evaluating treatment response and predicting relapse risk. MicroRNA (miRNAs), small non-coding RNAs with pivotal roles in gene regulation, have emerged as promising biomarkers for JMML MRD detection. This review explores the mechanistic role of miRNAs in JMML pathogenesis, emphasizing their diagnostic, prognostic, and therapeutic potential. Dysregulated miRNA profiles correlate with distinct JMML subgroups and disease progression, suggesting utility in non-invasive MRD monitoring. Emerging evidence highlights miR-150-5p as a tumor suppressor targeting STAT5b and its therapeutic potential in ameliorating JMML's aberrant signaling pathways. We compare traditional MRD methods, such as flow cytometry and polymerase chain reaction (PCR), with miRNA-based techniques, underscoring the latter's superior sensitivity, specificity, and non-invasiveness. Recent advances in miRNA profiling technologies, including next-generation sequencing and digital PCR, enable precise detection of residual leukemic cells and support personalized treatment approaches. Despite significant progress, challenges persist in standardizing miRNA-based assays and validating their clinical utility. Ethical considerations, including patient privacy and informed consent, remain critical for integrating miRNA diagnostics into routine care. This review provides a comprehensive synthesis of current knowledge on miRNA signatures in JMML, illuminating their transformative potential in MRD monitoring and paving the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Bhavyadharshini Arun
- Hasan Lab, Department of Medical Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Geofrey John
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Rajeshkumar Raman
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, Ooty, The Nilgiris, India
| |
Collapse
|
47
|
Zamani Sani M, Mirzaei M, Mota A, Mohammadian J, Aboutalebi Vand Beilankouhi E, Rahmati M. MicroRNAs' Significance in Retinoblastoma Diagnosis and Treatment: The Little Heroes. Biochem Genet 2025; 63:1176-1197. [PMID: 39862293 DOI: 10.1007/s10528-024-10976-2] [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] [Accepted: 11/09/2024] [Indexed: 01/27/2025]
Abstract
One in 16, 000 live births is affected by the retinal tumor RB (retinoblastoma), which is frequently found in a child's early years. Both of the RB1 alleles that have been locally mutated in the affected retina are present in 60 percent of cases. Retinoblastoma (RB) can be detected using a variety of techniques, including imaging of the brain and orbits, eye examinations under anesthesia (EUAs), and the discovery of cell-free tumor DNA in samples of aqueous humor or plasma. In addition to the conventional surgical, chemotherapy, and radiotherapy approaches to treating retinoblastoma, new approaches have also been developed. Oncogenes, genes of tumor suppressors, and other molecular elements involved in cell growth and division interact complexly during the pathogenesis of retinoblastoma. The development of new therapies depends on comprehending the function of these molecular components. As a small class of non-coding RNAs capable of altering gene expression, microRNAs (miRNA) are understood to represent potential targets for the treatment of cancer. This study aimed to describe the changes in microRNA expression in some types of cancer, with a particular focus on retinoblastoma.
Collapse
Affiliation(s)
- Maryam Zamani Sani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mirzaei
- Department of Ophthalmology, Nikoukari Eye Hospital, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mota
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jamal Mohammadian
- School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Rahmati
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
48
|
Guang S, Liu MF. The discovery of tiny RNAs that have crucial roles. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1183-1185. [PMID: 39792332 DOI: 10.1007/s11427-024-2819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Affiliation(s)
- Shouhong Guang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The USTC RNA Institute, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Life Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230027, China.
| | - Mo-Fang Liu
- New Cornerstone Science Laboratory, Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
49
|
Solaimani M, Hosseinzadeh S, Abasi M. Non-coding RNAs, a double-edged sword in breast cancer prognosis. Cancer Cell Int 2025; 25:123. [PMID: 40170036 PMCID: PMC11959806 DOI: 10.1186/s12935-025-03679-0] [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: 05/07/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Cancer is a rising issue worldwide, and numerous studies have focused on understanding the underlying reasons for its occurrence and finding proper ways to defeat it. By applying technological advances, researchers are continuously uncovering and updating treatments in cancer therapy. Their vast functions in the regulation of cell growth and proliferation and their significant role in the progression of diseases, including cancer. This review provides a comprehensive analysis of ncRNAs in breast cancer, focusing on long non-coding RNAs such as HOTAIR, MALAT1, and NEAT1, as well as microRNAs such as miR-21, miR-221/222, and miR-155. These ncRNAs are pivotal in regulating cell proliferation, metastasis, drug resistance, and apoptosis. Additionally, we discuss experimental approaches that are useful for studying them and highlight the advantages and challenges of each method. We then explain the results of these clinical trials and offer insights for future studies by discussing major existing gaps. On the basis of an extensive number of studies, this review provides valuable insights into the potential of ncRNAs in cancer therapy. Key findings show that even though the functions of ncRNAs are vast and undeniable in cancer, there are still complications associated with their therapeutic use. Moreover, there is an absence of sufficient experiments regarding their application in mouse models, which is an area to work on. By emphasizing the crucial role of ncRNAs, this review underscores the need for innovative approaches and further studies to explore their potential in cancer therapy.
Collapse
Affiliation(s)
- Maryam Solaimani
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Sahar Hosseinzadeh
- Faculty of Pharmacy and Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mozhgan Abasi
- Immunogenetics Research Center, Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, PO Box: 48175/861, Sari, Iran.
| |
Collapse
|
50
|
Lim SY, Boyd SC, Diefenbach RJ, Rizos H. Circulating MicroRNAs: functional biomarkers for melanoma prognosis and treatment. Mol Cancer 2025; 24:99. [PMID: 40156012 PMCID: PMC11951542 DOI: 10.1186/s12943-025-02298-7] [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: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
MicroRNAs (miRNAs) hold significant promise as circulating cancer biomarkers and unlike many other molecular markers, they can provide valuable insights that extend beyond tumour biology. The expression of circulating miRNAs may parallel the cellular composition and dynamic activity within the tumour microenvironment and reveal systemic immune responses. The functional complexity of miRNAs-where a single miRNA can regulate multiple messenger RNAs (mRNAs) to fine tune fundamental processes, and a single mRNA can be targeted by multiple miRNAs-underscores their broad significance and impact. However, this complexity poses significant challenges for translating miRNA research into clinical practice. In melanoma, specific miRNA signatures have shown notable diagnostic, prognostic and predictive value, with lineage-specific and immune-related miRNAs frequently identified as valuable markers. In this review, we explore the role of circulating miRNAs as potential biomarkers in melanoma, and highlight the current status and advances required to translate miRNA research into therapeutic opportunities.
Collapse
Affiliation(s)
- Su Yin Lim
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Suzanah C Boyd
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Russell J Diefenbach
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Helen Rizos
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|