1
|
Zhou S, Zhang L, Duan X, Liu K, Yingnan Y, Ma M, Han B. MiR-425-5p intervenes in autoimmune myocarditis by regulating Treg cell differentiation through NRAS. Front Cell Dev Biol 2025; 13:1600103. [PMID: 40433545 PMCID: PMC12106460 DOI: 10.3389/fcell.2025.1600103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Aim Our Previous research revealed significant differences in exosome-mediated intercellular miR-425a-5p between normal children and those with fulminant myocarditis. We sought to elucidate the molecular underpinnings and functional implications of miR-425a-5p in the context of myocarditis progression. Methods Bioinformatics techniques were employed to predict NRAS as the target gene of miR-425a-5p. We constructed a cellular myocarditis paradigm through LPS-mediated provocation of AC16 cardiomyocyte cultures. MiR-425a-5p was overexpressed, and the expressions of NRAS, cell apoptosis, and proinflammatory cytokine profiles, encompassing IL-1β, IL-6, and TNF-α, were comprehensively quantified. An experimental autoimmune myocarditis (EAM) mouse model was created using adeno-associated virus (AAV) for miR-425a-5p overexpression. Comprehensive histopathological analyses were conducted utilizing multiple staining techniques, including hematoxylin-eosin (HE), immunohistochemical, and Masson trichrome methodologies to characterize tissue responses. Results The study demonstrated that miR-425a-5p alleviated the inflammatory response in both AC16 cells and EAM mice through NRAS mediation. Single-cell data analysis of cardiac immune cells revealed that miR-425a-5p promoted Treg cell differentiation and improved cardiac function. Conclusion MiR-425a-5p plays a crucial role in modulating inflammatory responses in myocarditis, potentially offering a novel therapeutic strategy for managing the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
2
|
Zhou S, Han B. Biological disturbance of MiR-425 and its application prospects in cardiovascular diseases. Front Cell Dev Biol 2025; 13:1593241. [PMID: 40417179 PMCID: PMC12098596 DOI: 10.3389/fcell.2025.1593241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
MiR-425 is a biological molecule that has potential applications in cardiovascular diseases. It can regulate biological functions by combining with LncRNAs, binding with proteins, and changing the differentiation of immune cells. MiR-425 also has a role as a biomarker of disease. In cardiovascular diseases, it has clinical significance in reducing inflammation and heart repair, inducing angiogenesis, improving the prediction of atherosclerosis, reducing cardiac fibrosis, and regulating atrial natriuretic peptide to affect cardiovascular function. Target gene prediction and KEGG enrichment analysis are also mentioned.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Biomedical Engineering and Interdisciplinary Studies), Shandong First Medical University, jinan, China
| |
Collapse
|
3
|
Bayat M, Nahid-Samiei R, Sadri Nahand J, Naghili B. Interferon and immunity: the role of microRNA in viral evasion strategies. Front Immunol 2025; 16:1567459. [PMID: 40416980 PMCID: PMC12101089 DOI: 10.3389/fimmu.2025.1567459] [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: 01/27/2025] [Accepted: 03/26/2025] [Indexed: 05/27/2025] Open
Abstract
Interferons (IFNs) are indispensable innate antiviral cytokines that orchestrate the vertebrate immune response against viral incursions. Nearly every cell possesses the remarkable ability to release IFNs upon detecting viral threats, triggering a robust signaling cascade that alerts neighboring cells and halts viral propagation via paracrine communication. The intricate influence of IFNs is mediated by an extensive network of proteins activated through the Jak-STAT pathways, facilitating the swift transcription of over 300 interferon-stimulated genes (ISGs) that fortify cellular defenses against replication. However, the cunning nature of viruses has led to the evolution of sophisticated evasion strategies, notably through the manipulation of host microRNAs (miRNAs) that disrupt vital components of the IFN signaling machinery. This review delves into the intricate interplay between viral infections and both host- and viral-derived miRNAs, exploring their potent roles in modulating RIG-I-like receptors, Toll-like receptors, IFN receptors, and the JAK/STAT pathway, ultimately shaping the landscape of antiviral immunity.
Collapse
Affiliation(s)
- Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahil Nahid-Samiei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrouz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Liu W, Rao X, Sun W, Chen X, Yu L, Zhang J, Chen J, Zheng X. The neuroinflammatory role of microRNAs in Alzheimer's disease: pathological insights to therapeutic potential. Mol Cell Biochem 2025; 480:2689-2706. [PMID: 39567427 DOI: 10.1007/s11010-024-05164-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the most common cause of dementia, contributing to around 60-80% of cases. The main pathophysiology of AD is characterized by an abnormal accumulation of protein aggregates extracellularly (beta-amyloid plaques) and intracellularly (neurofibrillary tangles of hyperphosphorylated tau). However, an increasing number of studies have also suggested neuroinflammation may have a crucial role in precipitating the cascade reactions that result in the development of AD neuropathology. In particular, several studies indicate microRNAs (miRNAs) can act as regulatory factors for neuroinflammation in AD, with potential to affect the occurrence and/or progression of AD inflammation by targeting the expression of multiple genes. Therefore, miRNAs may have potential as therapeutic targets for AD, which requires more research. This article will review the existing studies on miRNAs that have been identified to regulate neuroinflammation, aiming to gain further insights into the specific regulatory processes of miRNAs, highlight the diagnostic and therapeutic potential of miRNAs as biomarkers in AD, as well as current challenges, and suggest the further work to bridge the gap in knowledge to utilize miRNAs as therapeutic targets for AD.
Collapse
Affiliation(s)
- Wenjia Liu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Xin Rao
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Wen Sun
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Xiaodong Chen
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Liyang Yu
- School of Electronics and Information, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Jiangtao Zhang
- Department of Geriatrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China.
| | - Jiong Chen
- Department of Geriatrics, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Xiaorong Zheng
- Blood Purification Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| |
Collapse
|
5
|
Wei Z, Li X, Zhou J, Zhou Y, Xiao Z, Yang Q, Liu X, Peng Y, Yang Y, Ding Y, Ru Z, Wang Y, Yang M, Yang X. Inhibition of miRNA-365-2-5p Targeting SIRT1 Regulates Functions of Keratinocytes to Enhance Wound Healing. FASEB J 2025; 39:e70560. [PMID: 40261275 DOI: 10.1096/fj.202401124rrr] [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: 05/19/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
The development of drugs to accelerate wound healing is an important area of clinical research. Recent advancements have highlighted the prospects of microRNAs as therapeutic targets for various disorders, although their involvement in mice wound healing remains unclear. Peptides have been proved to be unique and irreplaceable molecules in the elucidation of competing endogenous RNAs mechanisms (ceRNA) involved with skin wound healing. In the present work, CyRL-QN15, a peptide characterized by its minimal length and maximal wound healing efficacy, was applied as a probe to explore the ceRNA mechanism in regard to accelerated wound healing. Results showed that the use of CyRL-QN15 significantly reduced the expression of miRNA-365-2-5p at the wound in mice. In mouse keratinocytes, miRNA-365-2-5p inhibition increased SIRT1 and FOXO1 protein expression and decreased STAT2 protein expression, promoting cell proliferation, migration, and reducing inflammatory factors. Similarly, inhibiting miRNA-365-2-5p at mouse wounds promoted Full-thickness injured skin wounds healing, increased SIRT1 and FOXO1 protein expression, decreased STAT2 protein expression, and reduced inflammatory factors. Overall, these findings demonstrate that miRNA-365-2-5p serves a crucial function in the biological processes underlying cutaneous wound healing in mice, offering a novel target for future therapeutic interventions in wound healing.
Collapse
Affiliation(s)
- Ziqi Wei
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xingguo Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyi Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuxuan Zhou
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoxun Xiao
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Qian Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xin Liu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Peng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yuliu Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Yujing Ding
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Zeqiong Ru
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, Yunnan, China
| | - Meifeng Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
6
|
Wang X, Fu F, Li Z, Wang X, Shu Y, Wang J. Entropy-driven DNA circuit induced rolling circle transcription generates fluorescent light-up RNA aptamer for one-pot and label-free detection of miRNA-133a. Talanta 2025; 294:128173. [PMID: 40262346 DOI: 10.1016/j.talanta.2025.128173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/15/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
MiRNA-133a is a marker for the early diagnosis of acute myocardial infarction. Specific detection of miRNA-133a is of great significance for early diagnosis and treatment of heart disease. Here, a cascade amplification strategy based on entropy-driven DNA circuit (EDC) and rolling circle transcription (RCT) was constructed to generate repeated fluorescent light-up RNA aptamers for one-pot, and label-free detection of miRNA-133a. In this study, target miRNA-initiated EDC to produce single-stranded DNA would allow for the ligation of RCT's padlock into a loop and the formation of the complete T7 promoter. Subsequently the RCT was triggered, and effectively generated large number of repeated Spinach sequences. The aptamer combined with the corresponding fluorophore DFHBI-1T and formed RNA aptamer-fluorophore complexes to generate enhanced fluorescence signals. The developed biosensor exhibits sensitive detection of miRN-133a, with a linear range of 50 pM to 50 nM and a detection limit of 38.3 pM. The recovery of the actual sample was 99.1 %-103.0 %, and the relative standard deviation (RSD) was 1.6 %-3.6 %. In addition, the detection has high selectivity and can distinguish RNA sequences with single base mutation. This work provides a general method for the sensitive detection of miRNA biomarkers in molecular diagnosis.
Collapse
Affiliation(s)
- Xue Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Fan Fu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Zhihao Li
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Xiaojuan Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| | - Jianhua Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| |
Collapse
|
7
|
Saadh MJ, Saeed TN, Alfarttoosi KH, Sanghvi G, Roopashree R, Thakur V, Lakshmi L, Kubaev A, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomes and MicroRNAs: key modulators of macrophage polarization in sepsis pathophysiology. Eur J Med Res 2025; 30:298. [PMID: 40247413 PMCID: PMC12007276 DOI: 10.1186/s40001-025-02561-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] [Received: 01/26/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Sepsis is a highly dangerous and complex condition that can result in death. It is characterized by a strong reaction to an infection, causing dysfunction in multiple bodily systems and a high risk of mortality. The transformation of macrophages is a vital stage in the procedure as they possess the capability to interchange between two separate types: M1, which promotes inflammation, and M2, which inhibits inflammation. The choice greatly affects the immune response of the host. This analysis underscores the rapidly expanding roles of exosomes and microRNAs (miRNAs) in regulating the trajectory of macrophage polarization during episodes of sepsis. Exosomes, extremely small extracellular vesicles, facilitate cellular communication by transferring biologically active compounds, including miRNAs, proteins, and lipids. We investigate the impact of changes in exosome production and composition caused by sepsis on macrophage polarization and function. Unique microRNAs present in exosomes play a significant role in controlling crucial signaling pathways that govern the phenotype of macrophages. Through thorough examination of recent progress in this area, we clarify the ways in which miRNAs derived from exosomes can either aggravate or alleviate the inflammatory reactions that occur during sepsis. This revelation not only deepens our comprehension of the underlying mechanisms of sepsis, but it also reveals potential new biomarkers and targets for treatment. This assessment aims to amalgamate diverse research investigations and propose potential avenues for future investigations on the influence that exosomes and miRNAs have on macrophage polarization and the body's response to sepsis. These entities are essential for controlling the host's reaction to sepsis and hold important functions in this mechanism.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Tamara Nazar Saeed
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - L Lakshmi
- Department of Nursing, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
8
|
Telkoparan-Akillilar P, Chichiarelli S, Tucci P, Saso L. Integration of MicroRNAs with nanomedicine: tumor targeting and therapeutic approaches. Front Cell Dev Biol 2025; 13:1569101. [PMID: 40260417 PMCID: PMC12009947 DOI: 10.3389/fcell.2025.1569101] [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: 01/31/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a pivotal role in the post-transcriptional regulation of gene expression. Over the past decade, they have emerged as key regulators in cancer progression, influencing different cellular processes such as proliferation, apoptosis, metastasis, and immune evasion. Their unique ability to target multiple genes simultaneously makes miRNAs highly attractive as potential therapeutic agents in oncology. However, several challenges have hindered their direct clinical application, most notably their inherent instability in biological fluids, rapid degradation by nucleases, and inefficient delivery to specific tumor sites. Additionally, off-target effects and the potential for toxicity further complicate the therapeutic use of miRNAs. Nanomedicine offers a promising solution to these challenges by enabling the development of advanced platforms for the stable, safe, and targeted delivery of miRNAs. Nanoparticle-based delivery systems, such as liposomes, polymeric nanoparticles, and inorganic nanocarriers, can protect miRNAs from degradation, improve their bioavailability, and allow for precise tumor targeting through passive or active targeting mechanisms. These nanocarriers can also be engineered to release miRNAs in response to specific stimuli within the tumor microenvironment, enhancing therapeutic efficacy while minimizing side effects. This review will explore the integration of miRNAs with nanotechnology, focusing on various nanoparticle formulations and their roles in enhancing miRNA stability, specificity, and function in cancer treatment. In addition, we will discuss current advances in preclinical and clinical applications, highlight promising tumor-targeting strategies, and address the remaining challenges such as toxicity, immunogenicity, and scalability. Future research should focus on overcoming these barriers, ultimately paving the way for the widespread adoption of personalized miRNA-based nanomedicine in cancer therapy.
Collapse
Affiliation(s)
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, Rome, Italy
| |
Collapse
|
9
|
Luo Y, Ren Q, He J, Wu M. miR-126-3p Serves as a Biomarker for Hepatitis B Virus-Associated Chronic Acute Liver Failure and Regulates Inflammation by Regulating ERRFI1. J Biochem Mol Toxicol 2025; 39:e70252. [PMID: 40227026 DOI: 10.1002/jbt.70252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/14/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Hepatitis B virus-associated chronic acute liver failure (HBV-ACLF) is the leading cause of ACLF, affecting approximately 90% of patients with ACLF. The objective of this study was to investigate the clinical relevance of miR-126-3p on HBV-ACLF as well as the regulatory impact of ERRFI1 and miR-126-3p on the inflammatory response caused by ACLF via in vitro experimental methodologies. RT-qPCR was utilized to quantify the expression levels of miR-126-3p, ERRFI1, NLRP3, caspase 1, and IL-1β. The clinical function of miR-126-3p was assessed using ROC analysis or Kaplan-Meier curve. Cell proliferation was quantified via the CCK-8 assay, while the dual-luciferase reporter assay was employed to confirm the specific binding interaction between miR-126-3p and ERRFI1. In patients with HBV-ACLF, a significant downregulation of miR-126-3p expression was observed; The level of miR-126-3p served as a prognostic indicator for the progression of HBV-ACLF, with reduced expression being associated with an unfavorable clinical outcome. In addition, miR-126-3p was found to modulate LPS-induced cell proliferation, and inflammation in THLE-2 cells through the regulation of ERRFI1 expression. Therefore, miR-126-3p might serve as a biomarker for HBV-ACLF.
Collapse
Affiliation(s)
- Yiping Luo
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiuping Ren
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jun He
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Menghang Wu
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Yang Y, Ning H, Zhu H, Du J, Sun W, Song K, Cheng YY, Fan J, Peng X. A Supramolecular Nanoengine Generates Nanomechanical Force on Demand for Precise Cytosolic Delivery of Anti-miRNAs and Synergistic TNBC Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2419651. [PMID: 40025859 DOI: 10.1002/adma.202419651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/28/2025] [Indexed: 03/04/2025]
Abstract
Although anti-microRNA (miRNA) is capable of silencing target miRNA and regulating multiple mRNAs in diverse signaling pathways, RNA medicines still encounter numerous challenges, especially in terms of poor delivery, inefficient endo/lysosomal escape, and suboptimal treatment. Herein, we have developed a carrier-free supramolecular nanoengine, AMGA (anti-miRNA/GEM2-Azo), which significantly enhances the cytosolic delivery of anti-miRNA without requiring light irradiation, thereby facilitating precise targeting and synergistic chemo-gene therapy for triple-negative breast cancer (TNBC). AMGA can be rapidly internalized by cancer cells and specifically generate nanomechanical force to promote the efficient escape of anti-miRNAs from the endo/lysosome to the cytoplasm, simultaneously downregulating miR-21 and miR-10b. In comparison to Lipofectamine 2000, AMGA demonstrated superior efficacy in inhibiting the proliferation, migration, and invasion of cancer cells. Significantly, AMGA exhibited profound antitumor and gene silencing effects in an orthotopic human TNBC mouse model. This novel supramolecular nanoengine presents a promising strategy for cytosolic delivery of anti-miRNAs.
Collapse
Affiliation(s)
- Yuxin Yang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Haijun Ning
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Hao Zhu
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518071, China
| |
Collapse
|
11
|
Kandettu A, Kuthethur R, Chakrabarty S. A detailed review on the role of miRNAs in mitochondrial-nuclear cross talk during cancer progression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167731. [PMID: 39978440 DOI: 10.1016/j.bbadis.2025.167731] [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: 09/08/2024] [Revised: 01/11/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that are associated with biochemical pathways through the post-transcriptional regulation of gene expression in different cell types. Based on their expression pattern and function, miRNAs can have oncogenic and tumor suppressor activities in different cancer cells. Altered mitochondrial function and bioenergetics are known hallmarks of cancer cells. Mitochondria play a central role in metabolic reprogramming during cancer progression. Cancer cells exploit mitochondrial function for cell proliferation, invasion, migration and metastasis. Genetic and epigenetic changes in nuclear genome contribute to altered mitochondrial function and metabolic reprogramming in cancer cells. Recent studies have identified the role of miRNAs as major facilitators of anterograde and retrograde signaling between the nucleus and mitochondria in cancer cells. Detailed analysis of the miRNA-mediated regulation of mitochondrial function in cancer cells may provide new avenues for the diagnosis, prognosis, and therapeutic management of the disease. Our review aims to discuss the role of miRNAs in nuclear-mitochondrial crosstalk regulating mitochondrial functions in different cancer types. We further discussed the potential application of mitochondrial miRNAs (mitomiRs) targeting mitochondrial biogenesis and metabolism in developing novel cancer therapy.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS) Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Raviprasad Kuthethur
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS) Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS) Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
12
|
Sahoo B, Mishra DD, Tiwari S. MiR-192-5p targets cell cycle regulation in diabetic kidney disease via cyclin-dependent kinase inhibitor 3. Noncoding RNA Res 2025; 11:60-72. [PMID: 39736853 PMCID: PMC11683246 DOI: 10.1016/j.ncrna.2024.11.003] [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: 07/20/2024] [Revised: 10/08/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Diabetic kidney disease (DKD), a.k.a diabetic nephropathy, is a leading cause of end-stage renal disease. However, in a fair percentage of patients with type-2 diabetes, renal involvement also occurs due to non-diabetic reasons (non-diabetic kidney disease, NDKD). In this study, we identified miRNA-mRNA regulatory networks specific to human DKD pathogenesis. miRNA profiling of the renal biopsy from cases (DKD, n = 5), disease controls (T2DM with NDKD, n = 6), and non-diabetic, non-CKD controls (patients undergoing nephrectomy for renal cancer, n = 3) revealed 68 DKD-specific miRNA regulation. Sixteen target mRNAs of these DKD-miRNAs were found to have a negative association with the estimated glomerular filtration rate (eGFR) in patients with DKD. The renal gene expression and eGFR data of DKD patients (n = 10-18) in the NephroSeq database were used. Based on these findings, 11 miRNA-mRNA regulatory networks were constructed for human DKD pathogenesis. Of these, in-vitro validation of miR-192-5p- CDKN3 (Cell cycle-dependent kinase inhibitor 3) network was done as miR-192-5p exhibited a maximum number of target genes in the identified DKD regulatory networks, and CDKN3 appeared as a novel target of miR-192-5p in our study. We demonstrated that miR-192-5p overexpression or knockdown of CDKN3 attenuated high glucose-induced apoptosis, fibrotic gene expression, cell hypertrophy, and cell cycle dysregulation and improved viability of proximal tubular cells. Moreover, miR-192-5p overexpression significantly inhibited CDKN3 mRNA and protein expression in proximal tubular cells. Overall, 11 miRNA-mRNA regulatory networks were predicted for human DKD pathogenesis; among these, the association of miR-192-5p- CDKN3 network DKD pathogenesis was confirmed in proximal tubular cell culture.
Collapse
Affiliation(s)
- Biswajit Sahoo
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Deendayal Das Mishra
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| |
Collapse
|
13
|
AbdElneam AI, Al-Dhubaibi MS, Bahaj SS, Mohammed GF, Atef LM. Assessment of miR-19b-3p, miR-182-5p, and miR-155-5p expression and its relation. Arch Dermatol Res 2025; 317:619. [PMID: 40119951 DOI: 10.1007/s00403-025-04043-y] [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: 11/03/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/25/2025]
Abstract
Alopecia areata (AA) is an autoimmune disorder characterized by non-scarring hair loss. Despite the growing understanding of its immune-related pathogenesis, biomarkers for early diagnosis and disease severity assessment remain limited. Recent studies have suggested that microRNAs (miRNAs) play a crucial role in regulating immune responses and inflammation in autoimmune diseases. This study aimed to investigate the expression levels of three miRNAs, miR-19b-3p, miR-182-5p, and miR-155-5p, in AA patients and their potential as diagnostic markers and indicators of disease severity. A total of 67 AA patients and 62 healthy controls were included in this case-control study. The severity of AA was evaluated using the Severity of Alopecia Tool (SALT) score, categorizing patients into mild, moderate, and severe groups. Plasma miRNA extraction was performed using the Direct-zol™ RNA MiniPrep kit, and qRT-PCR analysis was conducted to quantify the expression levels of miR-19b-3p, miR-182-5p, and miR-155-5p. Diagnostic accuracy was assessed using Receiver Operating Characteristic (ROC) curve analysis, and correlation analysis was performed to examine the relationship between miRNA expression and disease severity. The results revealed that the expression of miR-19b-3p, miR-182-5p, and miR-155-5p was significantly higher in AA patients compared to healthy controls (p = 0.001 for all three miRNAs). ROC curve analysis demonstrated high diagnostic accuracy, with AUC values of 0.99 for miR-19b-3p, 0.95 for miR-182-5p, and 0.97 for miR-155-5p. These miRNAs showed high sensitivity and specificity, indicating their strong potential as diagnostic biomarkers. Moreover, correlation analysis revealed a significant association between miR-155-5p expression and the severity of AA (p < 0.001), suggesting its potential as a marker of disease progression. This study highlights the significant upregulation of miR-19b-3p, miR-182-5p, and miR-155-5p in AA patients, indicating their potential as minimally invasive diagnostic markers. Furthermore, the correlation between miRNA expression and disease severity provides valuable insights into the molecular mechanisms underlying AA. These findings suggest that miRNAs, particularly miR-155-5p, may serve as promising biomarkers for diagnosing and monitoring the progression of AA, potentially aiding in the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ahmed Ibrahim AbdElneam
- Department of Clinical Biochemistry, Department of Basic Medical Sciences, College of Medicine, Shaqra University, Dawadmi, Saudi Arabia
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Center, 33 El Bohouth St. (Former El Tahrir St.), Dokki 12622, Cairo, Egypt
| | | | - Saleh Salem Bahaj
- Department of Microbiology and Immunology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Yemen
| | - Ghada Farouk Mohammed
- Department of Dermatology, Venereology, and Sexology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Lina Mohammed Atef
- Department of Dermatology, Venereology, and Sexology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
14
|
Popa ML, Ichim C, Anderco P, Todor SB, Pop-Lodromanean D. MicroRNAs in the Diagnosis of Digestive Diseases: A Comprehensive Review. J Clin Med 2025; 14:2054. [PMID: 40142862 PMCID: PMC11943142 DOI: 10.3390/jcm14062054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
MicroRNAs (miRNAs) have emerged as crucial regulators in digestive pathologies, including inflammatory bowel disease (miR-31, miR-155, and miR-21), colorectal cancer (miR-21, miR-598, and miR-494), and non-alcoholic fatty liver disease (miR-21, miR-192, and miR-122). Their capacity to modulate gene expression at the post-transcriptional level makes them highly promising candidates for biomarkers and therapeutic interventions. However, despite considerable progress, their clinical application remains challenging. Research has shown that miRNA expression is highly dynamic, varying across patients, disease stages, and different intestinal regions. Their dual function as both oncogenes and tumor suppressors further complicates their therapeutic use, as targeting miRNAs may yield unpredictable effects. Additionally, while miRNA-based therapies hold great potential, significant hurdles persist, including off-target effects, immune activation, and inefficiencies in delivery methods. The intricate interplay between miRNAs and gut microbiota adds another layer of complexity, influencing disease mechanisms and treatment responses. This review examined the role of miRNAs in digestive pathologies, emphasizing their diagnostic and therapeutic potential. While they offer new avenues for disease management, unresolved challenges underscore the need for further research to refine their clinical application.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (M.L.P.); (S.B.T.); (D.P.-L.)
| | - Paula Anderco
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (M.L.P.); (S.B.T.); (D.P.-L.)
| | | | | |
Collapse
|
15
|
Di Leva F, Arnoldi M, Santarelli S, Massonot M, Lemée MV, Bon C, Pellegrini M, Castellini ME, Zarantonello G, Messina A, Bozzi Y, Bernier R, Zucchelli S, Casarosa S, Dassi E, Ronzitti G, Golzio C, Morandell J, Gustincich S, Espinoza S, Biagioli M. SINEUP RNA rescues molecular phenotypes associated with CHD8 suppression in autism spectrum disorder model systems. Mol Ther 2025; 33:1180-1196. [PMID: 39741407 PMCID: PMC11897779 DOI: 10.1016/j.ymthe.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/01/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Loss-of-function mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are strongly associated with autism spectrum disorders (ASDs). Indeed, the reduction of CHD8 causes transcriptional, epigenetic, and cellular phenotypic changes correlated to disease, which can be monitored in assessing new therapeutic approaches. SINEUPs are a functional class of natural and synthetic antisense long non-coding RNAs able to stimulate the translation of sense target mRNA, with no effect on transcription. Here, we employed synthetic SINEUP-CHD8 targeting the first and third AUG of the CHD8 coding sequence to efficiently stimulate endogenous CHD8 protein production. SINEUP-CHD8 were effective in cells with reduced levels of the target protein and in patient-derived fibroblasts with CHD8 mutations. Functionally, SINEUP-CHD8 were able to revert molecular phenotypes associated with CHD8 suppression, i.e., genome-wide transcriptional dysregulation, and the reduction of H3K36me3 levels. Strikingly, in chd8-morpholino-treated and ENU mutant zebrafish embryos, SINEUP-chd8 injection confirmed the ability of SINEUP RNA to rescue the chd8-suppression-induced macrocephaly phenotype and neuronal hyperproliferation. Thus, SINEUP-CHD8 molecule(s) represent a proof-of-concept toward the development of an RNA-based therapy for neurodevelopmental syndromes with implications for, and beyond ASD, and relevant to genetic disorders caused by protein haploinsufficiency.
Collapse
Affiliation(s)
- Francesca Di Leva
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Michele Arnoldi
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Stefania Santarelli
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Mathieu Massonot
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104-UMR-S 1258, Department of Translational Medicine and Neurogenetics, 67404 Illkirch, France
| | - Marianne Victoria Lemée
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104-UMR-S 1258, Department of Translational Medicine and Neurogenetics, 67404 Illkirch, France
| | - Carlotta Bon
- Center for Human Technologies, Non-coding RNAs and RNA-based Therapeutics, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Miguel Pellegrini
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Maria Elena Castellini
- Neural Development and Regeneration Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Giulia Zarantonello
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Andrea Messina
- Neural Development and Regeneration Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Rovereto, 38060 Trento, Italy
| | - Raphael Bernier
- Department of Psychiatry and Behavioral Science, University of Washington School of Medicine, Seattle, WA 98195-6560, USA
| | - Silvia Zucchelli
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Simona Casarosa
- Neural Development and Regeneration Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Giuseppe Ronzitti
- Genethon, 91000 Evry, France; Université Paris-Saclay, University Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Christelle Golzio
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104-UMR-S 1258, Department of Translational Medicine and Neurogenetics, 67404 Illkirch, France
| | - Jasmin Morandell
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy.
| | - Stefano Gustincich
- Center for Human Technologies, Non-coding RNAs and RNA-based Therapeutics, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Stefano Espinoza
- Center for Human Technologies, Non-coding RNAs and RNA-based Therapeutics, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy; Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy.
| | - Marta Biagioli
- NeuroEpigenetics Laboratory, Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy.
| |
Collapse
|
16
|
Li S, Zhao Y, Lyu X, Chen Y, Zhang T, Lin S, Liu Z, Cai X, Tian T, Lin Y. Enzyme-Responsive Nanoparachute for Targeted miRNA Delivery: A Protective Strategy Against Acute Liver and Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411210. [PMID: 39717886 PMCID: PMC11905073 DOI: 10.1002/advs.202411210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/10/2024] [Indexed: 12/25/2024]
Abstract
MicroRNA (miRNA)-based therapy holds significant potential; however, its structural limitations pose a challenge to the full exploitation of its biomedical functionality. Framework nucleic acids are promising owing to their transportability, biocompatibility, and functional editability. MiRNA-125 is embedded into a nucleic acid framework to create an enzyme-responsive nanoparachute (NP), enhancing the miRNA loading capacity while preserving the attributes of small-scale framework nucleic acids and circumventing the uncertainty related to RNA exposure in conventional loading methods. An enzyme-sensitive sequence is designed in NP as a bioswitchable apparatus for cargo miRNAs release. NP is compared with conventional delivery modes and delivery vehicles, confirming its excellent transportability and sustained release properties. Moreover, NP confers good enzyme and serum resistance to the cargo miRNAs. Simultaneously, it can easily deliver miRNA-125 to liver and kidney lesions owing to its passive targeting properties. This allows for Keap1/Nrf2 pathway regulation and p53 protein targeting in the affected tissues. Additionally, NP negatively regulates the expression of Bax and Caspase-3. These combined actions help to inhibit oxidation, prevent cell cycle arrest, and reduce the apoptosis of liver and kidney cells. Consequently, this strategy offers a potential treatment for acute liver and kidney injury.
Collapse
Affiliation(s)
- Songhang Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yuxuan Zhao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiaoying Lyu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Ye Chen
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Shiyu Lin
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Taoran Tian
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Biomedical EngineeringSichuan UniversityChengdu610041P. R. China
| |
Collapse
|
17
|
Wu S, Lu J. Liposome-Enabled Nanomaterials for Muscle Regeneration. SMALL METHODS 2025:e2402154. [PMID: 39967365 DOI: 10.1002/smtd.202402154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/08/2025] [Indexed: 02/20/2025]
Abstract
Muscle regeneration is a vital biological process that is crucial for maintaining muscle function and integrity, particularly for the treatment of muscle diseases such as sarcopenia and muscular dystrophy. Generally, muscular tissues can self-repair and regenerate under various conditions, including acute or chronic injuries, aging, and genetic mutation. However, regeneration becomes challenging beyond a certain threshold, particularly in severe muscle injuries or progressive diseases. In recent years, liposome-based nanotechnologies have shown potential as promising therapeutic strategies for muscle regeneration. Liposomes offer an adaptable platform for targeted drug delivery due to their cell membrane-like structure and excellent biocompatibility. They can enhance drug solubility, stability, and targeted delivery while minimizing systemic side effects by different mechanisms. This review summarizes recent advancements, discusses current applications and mechanisms, and highlights challenges and future directions for possible clinical translation of liposome-based nanomaterials in the treatment of muscle diseases. It is hoped this review offers new insights into the development of liposome-enabled nanomedicine to address current limitations.
Collapse
Affiliation(s)
- Shuang Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
18
|
Wei X, Xiong X, Chen Z, Chen B, Zhang C, Zhang W. MicroRNA155 in non-small cell lung cancer: a potential therapeutic target. Front Oncol 2025; 15:1517995. [PMID: 39963112 PMCID: PMC11830606 DOI: 10.3389/fonc.2025.1517995] [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: 10/27/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Lung cancer (LC) is the second most commonly diagnosed cancer among both men and women, and it stands as the leading cause of cancer-related mortality, characterized by high rates of morbidity and mortality. Among its subtypes, non-small cell lung cancer (NSCLC) is the most prevalent and one of the most challenging malignant tumors to treat. To date, various therapeutic approaches, including surgery, radiotherapy, and chemotherapy, have been employed in the management of lung cancer; however, due to its aggressive nature, the survival rates remain low. Consequently, exploring novel treatment strategies is of paramount importance. MicroRNAs (miRNAs), a large family of non-coding RNAs, play crucial roles in regulating several key biological processes, including cell proliferation, differentiation, inflammation, and apoptosis. Among these, microRNA155(miR-155) is one of the most conserved and versatile miRNAs, predominantly overexpressed in various diseases, including malignant tumors. This review elucidates the biological functions and roles of miR-155 in NSCLC and discusses its potential significance as a therapeutic target for future research directions and clinical applications.
Collapse
Affiliation(s)
- Xiangju Wei
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xianmin Xiong
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ze Chen
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Bi Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cantang Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenhui Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
20
|
Huang B, Guo F, Chen J, Lu L, Gao S, Yang C, Wu H, Luo W, Pan Q. Regulation of B-cell function by miRNAs impacting Systemic lupus erythematosus progression. Gene 2025; 933:149011. [PMID: 39427831 DOI: 10.1016/j.gene.2024.149011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease marked by abnormal B-cell proliferation and increased autoantibodies. miRNAs play a crucial role in regulating B-cell dysfunction and SLE pathology. miRNAs influence DNA methylation, B-cell activation, and gene expression, contributing to SLE pathogenesis. miRNAs impact B cells through key processes like proliferation, differentiation, tolerance, and apoptosis. miRNAs also exacerbate inflammation and immune responses by modulating Interleukin 4 (IL-4), IL-6, and interferon cytokines. Autophagy, a key degradation mechanism, is also regulated by specific miRNAs that impact SLE pathology. This article explores the role of multiple miRNAs in regulating B-cell development, proliferation, survival, and immune responses, influencing SLE pathogenesis. miRNAs like miR-23a, the miR-17 ∼ 92 family, and miR-125b/miR-221 affect B-cell development by regulating transcription factors, signaling pathways, and cell cycle genes. miRNAs such as miR-181a-5p and miR-23a-5p are differentially regulated across developmental stages, emphasizing their complex regulatory roles in B-cell biology. This article synthesizes miRNA-B cell interactions to offer new strategies and directions for SLE diagnosis and treatment.
Collapse
Affiliation(s)
- Bitang Huang
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Fengbiao Guo
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jiaxuan Chen
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lu Lu
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Shenglan Gao
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Chunlong Yang
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Han Wu
- Clinical Laboratory, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Wenying Luo
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| | - Qingjun Pan
- Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
21
|
Kimura T, Sakata KI, Ohga N, Sato J, Itagaki T, Munekata T, Yanagawa-Matsuda A, Maeda T, Hojo M, Hatanaka KC, Hatanaka Y, Iizasa H. Salivary miRNAs as a novel therapeutic marker in a patient with advanced oral squamous cell carcinoma: A case report. Oncol Lett 2025; 29:52. [PMID: 39564374 PMCID: PMC11574702 DOI: 10.3892/ol.2024.14798] [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: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024] Open
Abstract
The global prevalence of oral squamous cell carcinoma (OSCC) has been increasing. OSCC at the advanced stage tends to resist conventional treatment and causes local recurrence and distant metastasis, resulting in poor prognosis. Therefore, detecting this cancer at an early stage and performing early intervention are important. Promising biomarkers to detect OSCC have yet to be established; however, microRNAs (miRNAs/miRs) serve a crucial role in OSCC tumorigenesis and may be potential biomarkers. In the present case report, the availability of salivary miRNAs as a therapeutic and prognostic marker for patients with OSCC was assessed. The patient was a 33-year-old woman who was diagnosed with advanced OSCC of the tongue, and their miRNA profile isolated from a saliva sample at each clinical course was evaluated. Microarray analysis of the salivary samples revealed changes in the levels of four miRNAs (hsa-miR-6798-5p, miR-6803-5p, miR-6805-5p and miR-6845-5p) in accordance with the clinical course. Neoadjuvant chemotherapy and surgical procedure decreased the levels, whereas the levels increased when the patient was diagnosed with lung metastasis. Furthermore, tongue and lung metastatic lesion specimens exhibited expression of the vascular endothelial growth factor receptor-2, which is regulated by the four miRNAs. Accordingly, the present report proposed that salivary miRNAs could be a therapeutic and prognostic biomarker for OSCC.
Collapse
Affiliation(s)
- Taku Kimura
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Ken-Ichiro Sakata
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Noritaka Ohga
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Jun Sato
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Tatsuki Itagaki
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Takeshi Munekata
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Aya Yanagawa-Matsuda
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido 060-8586, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8648, Japan
| | - Masahiro Hojo
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8648, Japan
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Hokkaido 060-8648, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Hokkaido 060-8648, Japan
| | - Hisashi Iizasa
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501, Japan
| |
Collapse
|
22
|
Brown SD, Klimi E, Bakker WAM, Beqqali A, Baker AH. Non-coding RNAs to treat vascular smooth muscle cell dysfunction. Br J Pharmacol 2025; 182:246-280. [PMID: 38773733 DOI: 10.1111/bph.16409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
Vascular smooth muscle cell (vSMC) dysfunction is a critical contributor to cardiovascular diseases, including atherosclerosis, restenosis and vein graft failure. Recent advances have unveiled a fascinating range of non-coding RNAs (ncRNAs) that play a pivotal role in regulating vSMC function. This review aims to provide an in-depth analysis of the mechanisms underlying vSMC dysfunction and the therapeutic potential of various ncRNAs in mitigating this dysfunction, either preventing or reversing it. We explore the intricate interplay of microRNAs, long-non-coding RNAs and circular RNAs, shedding light on their roles in regulating key signalling pathways associated with vSMC dysfunction. We also discuss the prospects and challenges associated with developing ncRNA-based therapies for this prevalent type of cardiovascular pathology. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
MESH Headings
- Animals
- Humans
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- RNA, Untranslated/pharmacology
- RNA, Untranslated/therapeutic use
Collapse
Affiliation(s)
- Simon D Brown
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Eftychia Klimi
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Abdelaziz Beqqali
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
23
|
Shao S, Du W, Liu S, Hu C, Zhang C, Li L, Yang F, Liu Q, Tan W. Reconfigurable Amphiphilic DNA Nanotweezer for Targeted Delivery of Therapeutic Oligonucleotides. ACS CENTRAL SCIENCE 2024; 10:2338-2345. [PMID: 39735310 PMCID: PMC11672532 DOI: 10.1021/acscentsci.4c01152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024]
Abstract
Amphiphilic lipid oligonucleotide conjugates are powerful molecular-engineering materials that have been used for delivery of therapeutic oligonucleotides. However, conventional lipid oligonucleotide conjugates suffer from poor selectivity to target cells due to the nonspecific interaction between lipid tails and cell membranes. Herein, a reconfigurable DNA nanotweezer consisting of a c-Met aptamer and bischolesterol-modified antisense oligonucleotide was designed for c-Met-targeted delivery of therapeutic antisense oligonucleotides. The c-Met aptamer is used to keep the DNA nanotweezer in a "closed" state, which enables the hydrophobic interaction within bischolesterol moieties. As a result, the amphiphilic DNA nanotweezer shows only a weak interaction with the cell membrane. Upon the release of the c-Met aptamer, the DNA nanotweezer converts to an "open" state, which facilitates the insertion of a cholesterol moiety into the cell membrane. Thus, the reconfigurable DNA nanotweezer enables the selective membrane anchoring of the DNA nanotweezer in cancerous cells that highly expressed c-Met protein. Moreover, this amphiphilic DNA nanotweezer shows enhanced accumulation at the tumor site and the inhibition of tumor growth. Taking advantage of the stimuli-responsive membrane anchoring capability, this reconfigurable DNA nanotweezer could be further explored as a smart multifunctional platform for cancer therapy.
Collapse
Affiliation(s)
- Shuxuan Shao
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wei Du
- Department
of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s
Hospital of Changde City), Changde, Hunan 415000, China
| | - Shuang Liu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Canqiong Hu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Cao Zhang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Lexun Li
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Fan Yang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Qiaoling Liu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, FuRong Laboratory, College of Biology, Hunan University, Changsha, Hunan 410082, China
- The
Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang
Cancer Hospital), Institute of Basic Medicine
and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- School
of Materials Science and Engineering, Institute of Molecular Medicine
(IMM), Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
24
|
Yu Y, He N, Song Z, Wang C, Xu J. LIMD1-AS1 promotes the progression of prostate cancer and affects the function of prostate cancer cells by down-regulating miR-29c-3p. J Cancer Res Clin Oncol 2024; 151:5. [PMID: 39636414 PMCID: PMC11621152 DOI: 10.1007/s00432-024-06046-0] [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: 10/24/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Prostate cancer (PCa) is a prevalent and lethal malignancy affecting males, with a considerable proportion of patients experiencing poor survival outcomes. The regulatory role of LIMD1-AS1 in the initiation and progression of PCa is emerging as a significant factor, however, the precise mechanisms governing its influence are yet to be fully elucidated. METHODS qRT-PCR was employed to assess the expression of LIMD1-AS1 and miR-29c-3p. The Cell Counting Kit-8 (CCK-8) was used to assess cell proliferation in PCa cells. Apoptosis rates were determined using flow cytometry. Cell migration and invasion were evaluated using the transwell assay. The targeted relationship of LIMD1-AS1 and miR-29c-3p was confirmed through dual-luciferase reporter gene analysis. RESULTS Increased expression of LIMD1-AS1 and decreased expression of miR-29c-3p were observed in both tumor tissues and serum from PCa patients. LIMD1-AS1 exhibited diagnostic and prognostic significance in PCa patients. Functionally, LIMD1-AS1 modulated the expression of miR-29c-3p to potentiate the proliferative, migratory, and invasive capabilities of PCa cells while concurrently inhibiting apoptosis. CONCLUSION LncRNA LIMD1-AS1 promotes the advancement of PCa by regulating miR-29c-3p, indicating that LIMD1-AS1/miR-29c-3p axis could serve as potential therapeutic targets for the therapeutic intervention of PCa.
Collapse
Affiliation(s)
- Yongsheng Yu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangdong, 511300, China
| | - Nan He
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Hubei, 430071, China
| | - Zhaolu Song
- Department of Urology Surgery, Jiaozhou Central Hospital of Qingdao, Shandong, 266300, China
| | - Chang Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou City, Guangdong Province, 510080, China.
| | - Jinhuang Xu
- Department of Trauma and Joint Surgery, The Fourth Affiliated Hospital of Guangzhou Medical University, No. 1, Guangming East Road, Zengcheng District, Guangzhou City, Guangdong Province, 511300, China.
| |
Collapse
|
25
|
Zhao J, Zhang X, Li F, Lei X, Ge L, Li H, Zhao N, Ming J. The Effects of Interventions with Glucosinolates and Their Metabolites in Cruciferous Vegetables on Inflammatory Bowel Disease: A Review. Foods 2024; 13:3507. [PMID: 39517291 PMCID: PMC11544840 DOI: 10.3390/foods13213507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract which affects millions of individuals worldwide. Despite advancements in treatment options, there is increasing interest in exploring natural interventions with minimal side effects. Cruciferous vegetables, such as broccoli, cabbage, and radishes, contain bioactive compounds known as glucosinolates (GLSs), which have shown promising effects in alleviating IBD symptoms. This review aims to provide a comprehensive overview of the physiological functions and mechanisms of cruciferous GLSs and their metabolites in the context of IBD. Reviewed studies demonstrated that GLSs attenuated all aspects of IBD, including regulating the intestinal microbiota composition, exerting antioxidant and anti-inflammatory effects, restoring intestinal barrier function, and regulating epigenetic mechanisms. In addition, a few interventions with GLS supplementation in clinical studies were also discussed. However, there are still several challenges and remaining knowledge gaps, including variations in animals' experimental outcomes, the bioavailability of certain compounds, and few clinical trials to validate their effectiveness in human subjects. Addressing these issues will contribute to a better understanding of the therapeutic potential of cruciferous GLSs and their metabolites in the management of IBD.
Collapse
Affiliation(s)
- Jichun Zhao
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
| | - Xiaoqin Zhang
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
| | - Fuhua Li
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
| | - Xiaojuan Lei
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
| | - Lihong Ge
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Honghai Li
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
| | - Nan Zhao
- Institute of Agro-products Processing Science and Technology, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China
| | - Jian Ming
- College of Food Science, Southwest University, Chongqing 400715, China; (J.Z.)
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Research Center for Fruits and Vegetables Logistics Preservation and Nutritional Quality Control, Southwest University, Chongqing 400715, China
| |
Collapse
|
26
|
Theotoki EI, Kakoulidis P, Velentzas AD, Nikolakopoulos KS, Angelis NV, Tsitsilonis OE, Anastasiadou E, Stravopodis DJ. TRBP2, a Major Component of the RNAi Machinery, Is Subjected to Cell Cycle-Dependent Regulation in Human Cancer Cells of Diverse Tissue Origin. Cancers (Basel) 2024; 16:3701. [PMID: 39518139 PMCID: PMC11545598 DOI: 10.3390/cancers16213701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Transactivation Response Element RNA-binding Protein (TRBP2) is a double-stranded RNA-binding protein widely known for its critical contribution to RNA interference (RNAi), a conserved mechanism of gene-expression regulation mediated through small non-coding RNA moieties (ncRNAs). Nevertheless, TRBP2 has also proved to be involved in other molecular pathways and biological processes, such as cell growth, organism development, spermatogenesis, and stress response. Mutations or aberrant expression of TRBP2 have been previously associated with diverse human pathologies, including Alzheimer's disease, cardiomyopathy, and cancer, with TRBP2 playing an essential role(s) in proliferation, invasion, and metastasis of tumor cells. METHODS Hence, the present study aims to investigate, via employment of advanced flow cytometry, immunofluorescence, cell transgenesis and bioinformatics technologies, new, still elusive, functions and properties of TRBP2, particularly regarding its cell cycle-specific control during cancer cell division. RESULTS We have identified a novel, mitosis-dependent regulation of TRBP2 protein expression, as clearly evidenced by the lack of its immunofluorescence-facilitated detection during mitotic phases, in several human cancer cell lines of different tissue origin. Notably, the obtained TRBP2-downregulation patterns seem to derive from molecular mechanisms that act independently of oncogenic activities (e.g., malignancy grade), metastatic capacities (e.g., low versus high), and mutational signatures (e.g., p53-/- or p53ΔΥ126) of cancer cells. CONCLUSIONS Taken together, we herein propose that TRBP2 serves as a novel cell cycle-dependent regulator, likely exerting mitosis-suppression functions, and, thus, its mitosis-specific downregulation can hold strong promise to be exploited for the efficient and successful prognosis, diagnosis, and (radio-/chemo-)therapy of diverse human malignancies, in the clinic.
Collapse
Affiliation(s)
- Eleni I. Theotoki
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (E.I.T.); (K.-S.N.)
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 115 27 Athens, Greece;
| | - Panos Kakoulidis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 115 27 Athens, Greece;
- Department of Informatics and Telecommunications, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece
| | - Athanassios D. Velentzas
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (E.I.T.); (K.-S.N.)
| | - Konstantinos-Stylianos Nikolakopoulos
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (E.I.T.); (K.-S.N.)
| | - Nikolaos V. Angelis
- Section of Animal and Human Physiology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (N.V.A.); (O.E.T.)
| | - Ourania E. Tsitsilonis
- Section of Animal and Human Physiology, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (N.V.A.); (O.E.T.)
| | - Ema Anastasiadou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 115 27 Athens, Greece;
- Department of Health Science, Higher Colleges of Technology (HCT), Academic City Campus, Dubai 17155, United Arab Emirates
| | - Dimitrios J. Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), 157 01 Athens, Greece; (E.I.T.); (K.-S.N.)
| |
Collapse
|
27
|
Rahdan F, Saberi A, Saraygord-Afshari N, Hadizadeh M, Fayeghi T, Ghanbari E, Dianat-Moghadam H, Alizadeh E. Deciphering the multifaceted role of microRNAs in hepatocellular carcinoma: Integrating literature review and bioinformatics analysis for therapeutic insights. Heliyon 2024; 10:e39489. [PMID: 39498055 PMCID: PMC11532857 DOI: 10.1016/j.heliyon.2024.e39489] [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: 05/15/2024] [Revised: 08/06/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Hepatocellular carcinoma (HCC) poses a significant global health challenge, necessitating innovative therapeutic strategies. MicroRNAs (miRNAs) have emerged as pivotal regulators of HCC pathogenesis, influencing key processes such as self-renewal, angiogenesis, glycolysis, autophagy, and metastasis. This article integrates findings from a comprehensive literature review and bioinformatics analysis to elucidate the role of miRNAs in HCC. We discuss how dysregulation of miRNAs can drive HCC initiation, progression, and metastasis by modulating various signaling pathways and target genes. Moreover, leveraging high-throughput technology and bioinformatics tools, we identify key miRNAs involved in multiple cancer hallmarks, offering insights into potential combinatorial therapeutic strategies. Through our analysis considering p-values and signaling pathways associated with key features, we unveil miRNAs with simultaneous roles across critical cancer characteristics, providing a basis for the development of high-performance biomarkers. The microRNAs, miR-34a-5p, miR-373-3p, miR-21-5p, miR-214-5p, miR-195-5p, miR-139-5p were identified to be shared microRNAs in stemness, angiogenesis, glycolysis, autophagy, EMT, and metastasis of HCC. However, challenges such as miRNA stability and delivery hinder the translation of miRNA-based therapeutics into clinical practice. This review underscores the importance of further research to overcome existing barriers and realize the full potential of miRNA-based interventions for HCC management.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alihossein Saberi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahura Fayeghi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ghanbari
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Zhan T, Tong L, Wang L, Dong J. CRISPR-based molecule-regulatory expression platform for specific immunotherapy of cancer. Front Oncol 2024; 14:1469319. [PMID: 39507755 PMCID: PMC11537849 DOI: 10.3389/fonc.2024.1469319] [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: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Cancer is still a major challenge of human health. The abnormality of intracellular cancer-related signal pathways is an important mechanism for the occurrence of cancer. Methods We used a molecular-senor to act on the endogenous signal molecules within the cell to redirect the abnormal signal flows in the cell to treat cancer. Based on CRISPR-dCas12f procedures, we combined aptamers and ribozymes to construct riboswitches, which served as molecular switches to reprogram sgRNAs, so that CRISPR-dCas12f redirected the intracellular anti-cancer signal flows after sensing specific input signal molecules. In addition, the activated molecular sensors and the inhibitory molecular sensors were constructed by combining transcription factors (VP64) and transcription inhibitors (KRAB) to specifically activate and inhibit target genes of interest. Results Our experimental results showed that the molecular sensors that we designed and constructed specifically sensed the endogenous signal molecules and then redirect the cancer related signal networks of cancer cells. In addition, corresponding logic gates were constructed to distinguish cancer cells from normal cells and redirect anticancer signal flows to trigger specific cancer immunotherapy. Conclusion The constructed molecular sensors constructed specifically recognized the signal molecules within the cell and redirected the endogenous signal pathway to reprogram the fate of cancer cells.
Collapse
|
29
|
Shaheen N, Shaheen A, Osama M, Nashwan AJ, Bharmauria V, Flouty O. MicroRNAs regulation in Parkinson's disease, and their potential role as diagnostic and therapeutic targets. NPJ Parkinsons Dis 2024; 10:186. [PMID: 39369002 PMCID: PMC11455891 DOI: 10.1038/s41531-024-00791-2] [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: 06/03/2023] [Accepted: 09/15/2024] [Indexed: 10/07/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression by binding to target messenger RNA (mRNA) molecules and promoting their degradation or blocking their translation. Parkinson's disease (PD) is a neurodegenerative disorder caused by the loss of dopaminergic neurons in the substantia nigra. There is increasing evidence to suggest that miRNAs play a role in the pathogenesis of PD. Studies have identified several miRNAs that are dysregulated in the brains of PD patients, and animal models of the disease. MiRNA expression dysregulation contributes to the onset and progression of PD by modulating neuroinflammation, oxidative stress, and protein aggregation genes. Moreover, miRNAs have emerged as potential therapeutic targets for PD. This review elucidates the changes in miRNA expression profiles associated with PD, emphasising their potential as diagnostic biomarkers and therapeutic targets, and detailing specific miRNAs implicated in PD and their downstream targets. Integrated Insights into miRNA Function, Microglial Activation, Diagnostic, and Treatment Prospects in PD Note: This figure is an original figure created by the authors.
Collapse
Affiliation(s)
- Nour Shaheen
- Alexandria University, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Ahmed Shaheen
- Alexandria University, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Mahmoud Osama
- Department of Neurosurgery, Nasser Institute for Research and Treatment, Cairo, Egypt
| | | | - Vishal Bharmauria
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- Center for Vision Research and Center for Integrative and Applied Neuroscience, York University, Toronto, ON, Canada
- Tampa Human Neurophysiology Lab, Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, USA
| | - Oliver Flouty
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA.
- Tampa Human Neurophysiology Lab, Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
30
|
Moustafa HAM, Elsakka EGE, Abulsoud AI, Elshaer SS, Rashad AA, El-Dakroury WA, Sallam AAM, Rizk NI, Zaki MB, Gomaa RM, Elesawy AE, Mohammed OA, Abdel Mageed SS, Eleragi AMS, ElBoghdady JA, El-Fayoumi SH, Abdel-Reheim MA, Doghish AS. The miRNA Landscape in Crohn's disease: Implications for novel therapeutic approaches and interactions with Existing therapies. Exp Cell Res 2024; 442:114234. [PMID: 39233267 DOI: 10.1016/j.yexcr.2024.114234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
MicroRNAs (miRNAs), which are non-coding RNAs consisting of 18-24 nucleotides, play a crucial role in the regulatory pathways of inflammatory diseases. Several recent investigations have examined the potential role of miRNAs in forming Crohn's disease (CD). It has been suggested that miRNAs serve as diagnostics for both fibrosis and inflammation in CD due to their involvement in the mechanisms of CD aggravation and fibrogenesis. More information on CD pathophysiology could be obtained by identifying the miRNAs concerned with CD and their target genes. These findings have prompted several in vitro and in vivo investigations into the putative function of miRNAs in CD treatment. Although there are still many unanswered questions, the growing body of evidence has brought miRNA-based therapy one step closer to clinical practice. This extensive narrative study offers a concise summary of the most current advancements in CD. We go over what is known about the diagnostic and therapeutic benefits of miRNA mimicry and inhibition so far, and we see what additional miRNA family targets could be useful for treating CD-related inflammation and fibrosis.
Collapse
Affiliation(s)
- Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
| | - Al-Aliaa M Sallam
- epartment of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Nehal I Rizk
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Rania M Gomaa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, Cairo P.O. Box 11829, Egypt
| | - Ahmed E Elesawy
- epartment of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Ali M S Eleragi
- Department of Microorganisms and Clinical Parasitology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Jasmine A ElBoghdady
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Shaimaa H El-Fayoumi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | | | - Ahmed S Doghish
- epartment of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
31
|
Jafarzadeh A, Naseri B, Khorramdelazad H, Jafarzadeh S, Ghorbaninezhad F, Asgari Z, Masoumi J, Nemati M. Reciprocal Interactions Between Apelin and Noncoding RNAs in Cancer Progression. Cell Biochem Funct 2024; 42:e4116. [PMID: 39233464 DOI: 10.1002/cbf.4116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Apelin, a bioactive peptide that serves as an endogenous ligand for the apelin receptor (APJ), is overexpressed in various types of cancers and contributes to cancer cell proliferation, viability, migration, angiogenesis, and metastasis, as well as immune deviation. Noncoding RNAs (ncRNAs) regulate gene expression, and there is growing evidence suggesting a bidirectional crosstalk between ncRNAs (including long noncoding RNAs [lncRNAs], circular RNAs [circRNAs], and microRNAs [miRNAs]) and apelin in cancers. Certain miRNAs can directly target the apelin and inhibit its expression, thereby suppressing tumor growth. It has been indicated that miR-224, miR-195/miR-195-5p, miR-204-5p, miR-631, miR-4286, miR-637, miR-4493, and miR-214-3p target apelin mRNA and influence its expression in prostate cancer, lung cancer, esophageal cancer, chondrosarcoma, melanoma, gastric cancer, glioma, and hepatocellular carcinoma (HCC), respectively. Moreover, circ-NOTCH1, circ-ZNF264, and lncRNA BACE1-AS upregulate apelin expression in gastric cancer, glioma, and HCC, respectively. On the other hand, apelin has been shown to regulate the expression of certain ncRNAs to affect tumorigenesis. It was revealed that apelin affects the expression of circ_0000004/miR-1303, miR-15a-5p, and miR-106a-5p in osteosarcoma, lung cancer, and prostate cancer, respectively. This review explains a bidirectional interplay between ncRNAs and apelin in cancers to provide insights concerning the molecular mechanisms underlying this crosstalk and potential implications for cancer therapy.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sara Jafarzadeh
- Student Research Committee, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Farid Ghorbaninezhad
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Asgari
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Javad Masoumi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
32
|
Zhou S, Shan F, Cao Y, Huang P, Yang H, Liu S. In Situ Intracellular Autocatalytic Hairpin Assembly of the Y-Shaped DNA Nanostructure for miR-155 Sensing and Gene Silencing. Anal Chem 2024. [PMID: 39150789 DOI: 10.1021/acs.analchem.4c02490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024]
Abstract
miR-155 is a class of cancer markers closely related to cancer metastasis and invasion. Combining in situ detection with gene silencing not only helps to analyze the information on the abundance and spatial location of microRNA expression in the cell but also synergizes the therapy. In this work, we prepared HD@CM vesicles with three hairpin DNAs by using MCF-7 cell membranes. The hairpin DNAs can be triggered by endogenous miR-155, which opens the autocatalytic molecular circuit (ACHA) and obtains Y-shaped DNA nanostructures. This nanostructure not only detects endogenous miR-155 with high sensitivity for in situ imaging but also enables gene regulation of intracellular survivin mRNA. The levels of miR-155 in MDA-MB-231, MCF-7, Hela, and HEK-293T cells are found to be 7703, 3978, 1696, and 1229 copies/cell, respectively, as detected by HD@CMs. The fluorescence produced by HD@CM after coincubation with different cells is found to be proportional to the intracellular miR-155 content by confocal imaging. In addition, the gene regulatory function of the Y-shaped DNA structure resulted in significant inhibition of survivin protein expression and apoptosis rates of up to 83%. We look forward to the future application of our HD@CM platform for the precise diagnosis and programmable treatment of clinical cancers.
Collapse
Affiliation(s)
- Sisi Zhou
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Fanshu Shan
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yu Cao
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Puzhen Huang
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Haitang Yang
- Food and Pharmacy College, Xuchang University, Xuchang 461000, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
33
|
Micheva ID, Atanasova SA. MicroRNA dysregulation in myelodysplastic syndromes: implications for diagnosis, prognosis, and therapeutic response. Front Oncol 2024; 14:1410656. [PMID: 39156702 PMCID: PMC11327013 DOI: 10.3389/fonc.2024.1410656] [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: 04/01/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Myelodysplastic syndromes (MDS) are a group of malignant clonal hematological disorders with heterogeneous clinical course and risk of transformation to acute myeloid leukemia. Genetic and epigenetic dysregulation, including alterations in microRNA (miRNA) expression, plays a pivotal role in MDS pathogenesis influencing disease development and progression. MiRNAs, known for their regulatory roles in gene expression, have emerged as promising biomarkers in various malignant diseases. This review aims to explore the diagnostic and prognostic roles of miRNAs in MDS. We discuss research efforts aimed at understanding the clinical utility of miRNAs in MDS management. MiRNA dysregulation is linked to specific chromosomal abnormalities in MDS, providing insights into the molecular landscape of the disease. Circulating miRNAs in plasma offer a less invasive avenue for diagnostic and prognostic assessment, with distinct miRNA profiles identified in MDS patients. Additionally, we discuss investigations concerning the role of miRNAs as markers for treatment response to hypomethylating and immunomodulating agents, which could lead to improved treatment decision-making and monitoring. Despite significant progress, further research in larger patient cohorts is needed to fully elucidate the role of miRNAs in MDS pathogenesis and refine personalized approaches to patient care.
Collapse
Affiliation(s)
- Ilina Dimitrova Micheva
- Hematology Department, University Hospital St. Marina, Varna, Bulgaria
- Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| | - Svilena Angelova Atanasova
- Hematology Department, University Hospital St. Marina, Varna, Bulgaria
- Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| |
Collapse
|
34
|
Li C, Yang K, Song H, Xia C, Wu Q, Zhu J, Liu W, Gao T, Guo R, Liu Z, Yuan F, Tian Y, Zhou D. Porcine circovirus type 2 ORF5 induces an inflammatory response by up-regulating miR-21 levels through targeting nuclear ssc-miR-30d. Virus Res 2024; 346:199396. [PMID: 38763299 PMCID: PMC11144814 DOI: 10.1016/j.virusres.2024.199396] [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: 02/17/2024] [Revised: 05/07/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Porcine circovirus type 2 (PCV2) infection leads to multi-system inflammation in pigs, and this effect can be achieved by upregulating host miR-21. The underlying mechanism of miR-21 regulates PCV2-induced inflammation is already known, however, how PCV2 regulates miR-21 levels and function using both autonomic and host factors remains to be further revealed. Here we present the first evidence that PCV2 ORF5 induces an inflammatory response by up-regulating miR-21 level through targeting nuclear miR-30d. In this study, we found that overexpression of ORF5 significantly increased miR-21 level and promoted the expression of inflammatory cytokines and activation of the NF-κB pathway, while ORF5 mutation had the opposite effect. Moreover, the differential expression of miR-21 could significantly change the pro-inflammatory effect of ORF5, indicating that ORF5 promotes inflammatory response by up-regulating miR-21. Bioinformatics analysis and clinical detection found that nuclear miR-30d was significantly down-regulated after ORF5 overexpression and PCV2 infection, and targeted pri-miR-21 and PCV2 ORF5. Functionally, we found that miR-30d inhibited the levels of miR-21 and inflammatory cytokines in cells. Mechanistically, we demonstrated that ORF5 inhibits miR-30d expression levels through direct binding but not via the circRNA pathway, and miR-30d inhibits miR-21 levels by targeting pri-miR-21. In summary, the present study revealed the molecular mechanism of ORF5 upregulation of miR-21, further refined the molecular chain of PCV2-induced inflammatory response and elucidated the role of miRNAs in it.
Collapse
Affiliation(s)
- Chang Li
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Keli Yang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Haofei Song
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Chuqiao Xia
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Qiong Wu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Jiajia Zhu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Wei Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Ting Gao
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Rui Guo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Zewen Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Fangyan Yuan
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China
| | - Yongxiang Tian
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China.
| | - Danna Zhou
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064 China.
| |
Collapse
|
35
|
Li J, Zhang Y, Fu T, Xing G, Tong Y. Advancing cancer therapy: The role of MicroRNA in clinical applications. Pharmacol Res 2024; 206:107299. [PMID: 38972495 DOI: 10.1016/j.phrs.2024.107299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/09/2024]
Affiliation(s)
- Jianing Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | | | - Tong Fu
- Brandeis University, Waltham, MA, United States
| | - Guoli Xing
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Tong
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
36
|
Alkhazaali-Ali Z, Sahab-Negah S, Boroumand AR, Tavakol-Afshari J. MicroRNA (miRNA) as a biomarker for diagnosis, prognosis, and therapeutics molecules in neurodegenerative disease. Biomed Pharmacother 2024; 177:116899. [PMID: 38889636 DOI: 10.1016/j.biopha.2024.116899] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Neurodegenerative diseases that include Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), Huntington's disease (HD), and multiple sclerosis (MS) that arise due to numerous causes like protein accumulation and autoimmunity characterized by neurologic depletion which lead to incapacity in normal physiological function such as thinking and movement in these patients. Glial cells perform an important role in protective neuronal function; in the case of neuroinflammation, glial cell dysfunction can promote the development of neurodegenerative diseases. miRNA that participates in gene regulation and plays a vital role in many biological processes in the body; in the central nervous system (CNS), it can play an essential part in neural maturation and differentiation. In neurodegenerative diseases, miRNA dysregulation occurs, enhancing the development of these diseases. In this review, we discuss neurodegenerative disease (Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS)) and how miRNA is preserved as a diagnostic biomarker or therapeutic agent in these disorders. Finally, we highlight miRNA as therapy.
Collapse
Affiliation(s)
- Zahraa Alkhazaali-Ali
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Amir Reza Boroumand
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol-Afshari
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Bi S, He H, Gao F, Zhao Y. Ultrasensitive Photoelectrochemical Biosensor for Dual-miRNAs Detection Based on Molecular Logic Gates and Methylene Blue Sensitized ZnO@CdS@Au Nanorods. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36194-36203. [PMID: 38952261 DOI: 10.1021/acsami.4c08276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The occurrence of cancer is often closely related to multiple tumor markers, so it is important to develop multitarget detection methods. By the proper design of the input signals and logical operations of DNA logic gates, detection and diagnosis of cancer at different stages can be achieved. For example, in the early stages, specific input signals can be designed to correspond to early specific tumor markers, thereby achieving early cancer detection. In the late stage, logic gates for multitarget detection can be designed to simultaneously detect multiple biomarkers to improve diagnostic accuracy and comprehensiveness. In this work, we constructed a dual-target-triggered DNA logic gate for anchoring DNA tetrahedra, where methylene blue was embedded in the DNA tetrahedra to sensitize ZnO@CdS@Au, achieving ultrasensitive detection of the target substance. We tested the response of AND and OR logic gates to the platform. For AND logic gates, the sensing platform only responds when both miRNAs are present. In the concentration range of 10 aM to 10 nM, the photoelectric signal gradually increases with an increase of the target concentration. Subsequently, we used OR logic gates for miRNA detection. Even if only one target exists, the sensing platform exhibits excellent performance. Similarly, within the concentration range of 10 aM to 10 nM, the photoelectric signal gradually increases with an increase of the target concentration. The minimum detection limit is 1.10 aM. Whether it is the need to detect multiple targets simultaneously or only one of them, we can achieve it by selecting the appropriate logic gate. This strategy holds promising application prospects in fields such as biosensing, medical diagnosis, and environmental monitoring.
Collapse
Affiliation(s)
- Shiliang Bi
- Hebei Key Laboratory of Nano-Biotechnology, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China
| | - Hanxiao He
- Hebei Key Laboratory of Nano-Biotechnology, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China
| | - Faming Gao
- Hebei Key Laboratory of Nano-Biotechnology, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China
| | - Yang Zhao
- Hebei Key Laboratory of Nano-Biotechnology, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao 066004, China
| |
Collapse
|
38
|
Stiff T, Bayraktar S, Dama P, Stebbing J, Castellano L. CRISPR screens in 3D tumourspheres identified miR-4787-3p as a transcriptional start site miRNA essential for breast tumour-initiating cell growth. Commun Biol 2024; 7:859. [PMID: 39003349 PMCID: PMC11246431 DOI: 10.1038/s42003-024-06555-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/04/2024] [Indexed: 07/15/2024] Open
Abstract
Our study employs pooled CRISPR screens, integrating 2D and 3D culture models, to identify miRNAs critical in Breast Cancer (BC) tumoursphere formation. These screens combine with RNA-seq experiments allowing identification of miRNA signatures and targets essential for tumoursphere growth. miR-4787-3p exhibits significant up-regulation in BC, particularly in basal-like BCs, suggesting its association with aggressive disease. Surprisingly, despite its location within the 5'UTR of a protein coding gene, which defines DROSHA-independent transcription start site (TSS)-miRNAs, we find it dependant on both DROSHA and DICER1 for maturation. Inhibition of miR-4787-3p hinders tumoursphere formation, highlighting its potential as a therapeutic target in BC. Our study proposes elevated miR-4787-3p expression as a potential prognostic biomarker for adverse outcomes in BC. We find that protein-coding genes positively selected in the CRISPR screens are enriched of miR-4787-3p targets. Of these targets, we select ARHGAP17, FOXO3A, and PDCD4 as known tumour suppressors in cancer and experimentally validate the interaction of miR-4787-3p with their 3'UTRs. Our work illuminates the molecular mechanisms underpinning miR-4787-3p's oncogenic role in BC. These findings advocate for clinical investigations targeting miR-4787-3p and underscore its prognostic significance, offering promising avenues for tailored therapeutic interventions and prognostic assessments in BC.
Collapse
Affiliation(s)
- Tom Stiff
- University of Sussex, School of life Sciences, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK
| | - Salih Bayraktar
- University of Sussex, School of life Sciences, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK
| | - Paola Dama
- University of Sussex, School of life Sciences, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK
| | | | - Leandro Castellano
- University of Sussex, School of life Sciences, John Maynard Smith Building, Falmer, Brighton, BN1 9QG, UK.
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, W12 0NN, UK.
| |
Collapse
|
39
|
Wei J, Liang S, Yang P, Qing B, Ma J, Jiang L, Deng Q, Zhong W, Wang M, Qin Z. The therapeutic potential of Laggera alata in alleviating inflammation and oxidative stress: insights into the miR-150-5p/TRIM8 axis. Mol Cell Toxicol 2024. [DOI: 10.1007/s13273-024-00468-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 04/02/2025]
|
40
|
Qin C, Zhang J, Ma L. EMCMDA: predicting miRNA-disease associations via efficient matrix completion. Sci Rep 2024; 14:12761. [PMID: 38834687 DOI: 10.1038/s41598-024-63582-y] [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: 03/11/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
Abundant researches have consistently illustrated the crucial role of microRNAs (miRNAs) in a wide array of essential biological processes. Furthermore, miRNAs have been validated as promising therapeutic targets for addressing complex diseases. Given the costly and time-consuming nature of traditional biological experimental validation methods, it is imperative to develop computational methods. In the work, we developed a novel approach named efficient matrix completion (EMCMDA) for predicting miRNA-disease associations. First, we calculated the similarities across multiple sources for miRNA/disease pairs and combined this information to create a holistic miRNA/disease similarity measure. Second, we utilized this biological information to create a heterogeneous network and established a target matrix derived from this network. Lastly, we framed the miRNA-disease association prediction issue as a low-rank matrix-complete issue that was addressed via minimizing matrix truncated schatten p-norm. Notably, we improved the conventional singular value contraction algorithm through using a weighted singular value contraction technique. This technique dynamically adjusts the degree of contraction based on the significance of each singular value, ensuring that the physical meaning of these singular values is fully considered. We evaluated the performance of EMCMDA by applying two distinct cross-validation experiments on two diverse databases, and the outcomes were statistically significant. In addition, we executed comprehensive case studies on two prevalent human diseases, namely lung cancer and breast cancer. Following prediction and multiple validations, it was evident that EMCMDA proficiently forecasts previously undisclosed disease-related miRNAs. These results underscore the robustness and efficacy of EMCMDA in miRNA-disease association prediction.
Collapse
Affiliation(s)
- Chao Qin
- School of Information Science and Engineering, Qilu Normal University, Jinan, 250200, China.
| | - Jiancheng Zhang
- School of Information Science and Engineering, Qilu Normal University, Jinan, 250200, China
| | - Lingyu Ma
- School of Control Science and Engineering, Harbin Institute of Technology, Weihai, 250200, China
| |
Collapse
|
41
|
Chuang YT, Yen CY, Tang JY, Chang FR, Tsai YH, Wu KC, Chien TM, Chang HW. Protein phosphatase 2A modulation and connection with miRNAs and natural products. ENVIRONMENTAL TOXICOLOGY 2024; 39:3612-3627. [PMID: 38491812 DOI: 10.1002/tox.24199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/28/2024] [Accepted: 02/10/2024] [Indexed: 03/18/2024]
Abstract
Protein phosphatase 2A (PP2A), a heterotrimeric holoenzyme (scaffolding, catalytic, and regulatory subunits), regulates dephosphorylation for more than half of serine/threonine phosphosites and exhibits diverse cellular functions. Although several studies on natural products and miRNAs have emphasized their impacts on PP2A regulation, their connections lack systemic organization. Moreover, only part of the PP2A family has been investigated. This review focuses on the PP2A-modulating effects of natural products and miRNAs' interactions with potential PP2A targets in cancer and non-cancer cells. PP2A-modulating natural products and miRNAs were retrieved through a literature search. Utilizing the miRDB database, potential PP2A targets of these PP2A-modulating miRNAs for the whole set (17 members) of the PP2A family were retrieved. Finally, PP2A-modulating natural products and miRNAs were linked via a literature search. This review provides systemic directions for assessing natural products and miRNAs relating to the PP2A-modulating functions in cancer and disease treatments.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung, Taiwan
| | - Tsu-Ming Chien
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
42
|
Singh N, Rai MK, Agarwal V, Agarwal V. In Silico Prediction of NOS2, NOS3 and Arginase 1 Genes Targeting by Micro RNAs Upregulated in Systemic Sclerosis. INDIAN JOURNAL OF RHEUMATOLOGY 2024; 19:100-109. [DOI: 10.1177/09733698241229803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025] Open
Abstract
Introduction: Systemic sclerosis (SSc) is a chronic disorder, characterised by endothelial dysfunction and fibrosis of skin and internal organs. Endothelium dysfunction leads to a decrease in nitric oxide levels, consequent to reduced Nitric Oxide synthase 3 (NOS3) activity due to decreased NOS3 gene function. Besides endothelium, macrophages too play an important role in the pathogenesis of SSc; classically activated M1 macrophages (express NOS2) and alternatively activated M2 macrophages (express Arginase1) are differentially altered in favour of M2 macrophages. Micro-RNAs (miRNAs) regulate expression of various genes and may favour disease phenotype. Aim: Our aim was to identify differentially expressed micro-RNAs in SSc, which target NOS2, NOS3 and Arginase1 genes by in-silico approach. Methods: Data on miRNAs upregulation reported in various studies was collected and their sequence ID from miRBase was identified in FASTA format from NCBI. Binding strength of these miRNAs against NOS2, NOS3 and Arginase1 genes was evaluated by RNA22. Results: After scanning 39 publications reporting miRNA expression in SSc, a total of 13 miRNAs were found to be up-regulated for NOS2 (Gibbs free energy ≤18.0Kj/mol) and 15 miRNAs up-regulated for NOS3 (Gibbs free energy ≤18.0Kj/mol) whereas for Arginase1 only 2 miRNAs were up-regulated. Conclusion: There is differential upregulation of miRNAs in SSc. Micro RNAs targeting NOS2 and NOS3 genes are highly up-regulated in comparison to Arginase 1. Role of epigenetic regulation of genes by miRNAs may play a key role in pathogenesis of SSc.
Collapse
Affiliation(s)
- Neha Singh
- Department of Clinical Immunology and Rheumatology, SGPGIMS, Lucknow, Uttar Pradesh, India
| | - Mohit Kumar Rai
- Department of Clinical Immunology and Rheumatology, SGPGIMS, Lucknow, Uttar Pradesh, India
| | - Vishwesh Agarwal
- Mahatma Gandhi Missions Medical College, Navi Mumbai, Maharashtra, India
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, SGPGIMS, Lucknow, Uttar Pradesh, India
| |
Collapse
|
43
|
Jiang H, Qin H, Yang Q, Huang L, Liang X, Wang C, Moro A, Xu S, Wei Q. Effective delivery of miR-150-5p with nucleus pulposus cell-specific nanoparticles attenuates intervertebral disc degeneration. J Nanobiotechnology 2024; 22:292. [PMID: 38802882 PMCID: PMC11129471 DOI: 10.1186/s12951-024-02561-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The use of gene therapy to deliver microRNAs (miRNAs) has gradually translated to preclinical application for the treatment of intervertebral disc degeneration (IDD). However, the effects of miRNAs are hindered by the short half-life time and the poor cellular uptake, owing to the lack of efficient delivery systems. Here, we investigated nucleus pulposus cell (NPC) specific aptamer-decorated polymeric nanoparticles that can load miR-150-5p for IDD treatment. METHODS The role of miR-150-5p during disc development and degeneration was examined by miR-150-5p knockout (KO) mice. Histological analysis was undertaken in disc specimens. The functional mechanism of miR-150-5p in IDD development was investigated by qRT-PCR assay, Western blot, coimmunoprecipitation and immunofluorescence. NPC specific aptamer-decorated nanoparticles was designed, and its penetration, stability and safety were evaluated. IDD progression was assessed by radiological analysis including X-ray and MRI, after the annulus fibrosus needle puncture surgery with miR-150-5p manipulation by intradiscal injection of nanoparticles. The investigations into the interaction between aptamer and receptor were conducted using mass spectrometry, molecular docking and molecular dynamics simulations. RESULTS We investigated NPC-specific aptamer-decorated polymeric nanoparticles that can bind to miR-150-5p for IDD treatment. Furthermore, we detected that nanoparticle-loaded miR-150-5p inhibitors alleviated NPC senescence in vitro, and the effects of the nanoparticles were sustained for more than 3 months in vivo. The microenvironment of NPCs improves the endo/lysosomal escape of miRNAs, greatly inhibiting the secretion of senescence-associated factors and the subsequent degeneration of NPCs. Importantly, nanoparticles delivering miR-150-5p inhibitors attenuated needle puncture-induced IDD in mouse models by targeting FBXW11 and inhibiting TAK1 ubiquitination, resulting in the downregulation of NF-kB signaling pathway activity. CONCLUSIONS NPC-targeting nanoparticles delivering miR-150-5p show favorable therapeutic efficacy and safety and may constitute a promising treatment for IDD.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Hongyu Qin
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qinghua Yang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Longao Huang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiao Liang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Congyang Wang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Abu Moro
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Sheng Xu
- Research Centre for Regenerative Medicine, Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qingjun Wei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
44
|
Wang Y, Chen Y, Yang F, Yu X, Chu Y, Zhou J, Yan Y, Xi J. MiR-4465-modified mesenchymal stem cell-derived small extracellular vesicles inhibit liver fibrosis development via targeting LOXL2 expression. J Zhejiang Univ Sci B 2024; 25:594-604. [PMID: 39011679 PMCID: PMC11254680 DOI: 10.1631/jzus.b2300305] [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: 05/09/2023] [Accepted: 07/29/2023] [Indexed: 05/23/2024]
Abstract
Liver fibrosis is a significant health burden, marked by the consistent deposition of collagen. Unfortunately, the currently available treatment approaches for this condition are far from optimal. Lysyl oxidase-like protein 2 (LOXL2) secreted by hepatic stellate cells (HSCs) is a crucial player in the cross-linking of matrix collagen and is a significant target for treating liver fibrosis. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) have been proposed as a potential treatment option for chronic liver disorders. Previous studies have found that MSC-sEV can be used for microRNA delivery into target cells or tissues. It is currently unclear whether microRNA-4465 (miR-4465) can target LOXL2 and inhibit HSC activation. Additionally, it is uncertain whether MSC-sEV can be utilized as a gene therapy vector to carry miR-4465 and effectively inhibit the progression of liver fibrosis. This study explored the effect of miR-4465-modified MSC-sEV (MSC-sEVmiR-4465) on LOXL2 expression and liver fibrosis development. The results showed that miR-4465 can bind specifically to the promoter of the LOXL2 gene in HSC. Moreover, MSC-sEVmiR-4465 inhibited HSC activation and collagen expression by downregulating LOXL2 expression in vitro. MSC-sEVmiR-4465 injection could reduce HSC activation and collagen deposition in the CCl4-induced mouse model. MSC-sEVmiR-4465 mediating via LOXL2 also hindered the migration and invasion of HepG2 cells. In conclusion, we found that MSC-sEV can deliver miR-4465 into HSC to alleviate liver fibrosis via altering LOXL2, which might provide a promising therapeutic strategy for liver diseases.
Collapse
Affiliation(s)
- Yanjin Wang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yifei Chen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fuji Yang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xiaolong Yu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| | - Jing Zhou
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China. ,
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China. ,
| | - Jianbo Xi
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Clinical College of Xuzhou Medical University, Changzhou 213017, China.
| |
Collapse
|
45
|
Modarresi Chahardehi A, Afrooghe A, Emtiazi N, Rafiei S, Rezaei NJ, Dahmardeh S, Farz F, Naderi Z, Arefnezhad R, Motedayyen H. MicroRNAs and angiosarcoma: are there promising reports? Front Oncol 2024; 14:1385632. [PMID: 38826780 PMCID: PMC11143796 DOI: 10.3389/fonc.2024.1385632] [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: 02/14/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
In recent years, microRNAs (miRNAs) have garnered increasing attention for their potential implications in cancer pathogenesis, functioning either as oncogenes or tumor suppressors. Notably, angiosarcoma, along with various other cardiovascular tumors such as lipomas, rhabdomyomas, hemangiomas, and myxomas, has shown variations in the expression of specific miRNA subtypes. A substantial body of evidence underscores the pivotal involvement of miRNAs in the genesis of angiosarcoma and certain cardiovascular tumors. This review aims to delve into the current literature on miRNAs and their prospective applications in cardiovascular malignancies, with a specific focus on angiosarcoma. It comprehensively covers diagnostic methods, prognostic evaluations, and potential treatments while providing a recapitulation of angiosarcoma's risk factors and molecular pathogenesis, with an emphasis on the role of miRNAs. These insights can serve as the groundwork for designing randomized control trials, ultimately facilitating the translation of these findings into clinical applications. Moving forward, it is imperative for studies to thoroughly scrutinize the advantages and disadvantages of miRNAs compared to current diagnostic and prognostic approaches in angiosarcoma and other cardiovascular tumors. Closing these knowledge gaps will be crucial for harnessing the full potential of miRNAs in the realm of angiosarcoma and cardiovascular tumor research.
Collapse
Affiliation(s)
| | - Arya Afrooghe
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikoo Emtiazi
- Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Rafiei
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | | | - Sarvin Dahmardeh
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Farz
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Naderi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Arefnezhad
- Coenzyme R Research Institute, Tehran, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
46
|
Yin Y, Yang Y, Zhang Y, Shang Y, Li Q, Yuan J. MiR-132-3p suppresses peritoneal fibrosis induced by peritoneal dialysis via targeting TGF-β1/Smad2/3 signaling pathway. PLoS One 2024; 19:e0301540. [PMID: 38603722 PMCID: PMC11008817 DOI: 10.1371/journal.pone.0301540] [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: 11/20/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Peritoneal fibrosis (PF) is the main complication of peritoneal dialysis (PD) and the most common cause of cessation from PD. There is still no effective therapeutic approach to reserve PF. We aimed to investigate the role of miR-132-3p and underlying potential mechanisms in PF. METHODS A total of 18 Sprague-Dawley (SD) rats were divided randomly into three groups (n = 6): (i)Control group (ii)PF group (iii)PF+Losartan group; Rats in the PF group and PF+Losartan group received daily intraperitoneal injections of 3 mg/kg chlorhexidine for 14 days, and rats in the PF+Losartan group simultaneously received daily intraperitoneal injections of 2 mg/kg losartan for 14 days. The control group was injected with saline in the same volume. Met-5A cells were treated for 24h with TGF-β1 dissolved in recombinant buffered saline at a concentration of 10 ng/ml, meanwhile, PBS solution as a negative control. The human peritoneal solution was collected for the detection of miR-132-3p. RESULTS In vivo, SD rats were infused with chlorhexidine to establish PF model, and we found that miR-132-3p significantly decreased and the expressions of transforming growth factor-β1 (TGF-β1), and Smad2/3 were up-regulated in PF. In vitro, miR-132-3p mimics suppressed TGF-β1/Smad2/3 activity, whereas miR-132-3p inhibition activated the pathway. In human peritoneal solution, we found that the expression of miR-132-3p decreased in a time-dependent model and its effect became more pronounced with longer PD duration. CONCLUSION MiR-132-3p ameliorated PF by suppressing TGF-β1/Smad2/3 activity, suggesting that miR-132-3p represented a potential therapeutic approach for PF.
Collapse
Affiliation(s)
- Yangyang Yin
- School of Medicine, Guizhou University, Guiyang, Guizhou, China
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yuqi Yang
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yongqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yu Shang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qian Li
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Jing Yuan
- School of Medicine, Guizhou University, Guiyang, Guizhou, China
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
47
|
Zhai W, Zhao M, Wei C, Zhang G, Qi Y, Zhao A, Sun L. Biomarker profiling to determine clinical impact of microRNAs in cognitive disorders. Sci Rep 2024; 14:8270. [PMID: 38594359 PMCID: PMC11004146 DOI: 10.1038/s41598-024-58882-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
Alzheimer's disease (AD) and post-stroke cognitive impairment (PSCI) are the leading causes of progressive dementia related to neurodegenerative and cerebrovascular injuries in elderly populations. Despite decades of research, patients with these conditions still lack minimally invasive, low-cost, and effective diagnostic and treatment methods. MicroRNAs (miRNAs) play a vital role in AD and PSCI pathology. As they are easily obtained from patients, miRNAs are promising candidates for the diagnosis and treatment of these two disorders. In this study, we performed complete sequencing analysis of miRNAs from 24 participants, split evenly into the PSCI, post-stroke non-cognitive impairment (PSNCI), AD, and normal control (NC) groups. To screen for differentially expressed miRNAs (DE-miRNAs) in patients, we predicted their target genes using bioinformatics analysis. Our analyses identified miRNAs that can distinguish between the investigated disorders; several of them were novel and never previously reported. Their target genes play key roles in multiple signaling pathways that have potential to be modified as a clinical treatment. In conclusion, our study demonstrates the potential of miRNAs and their key target genes in disease management. Further in-depth investigations with larger sample sizes will contribute to the development of precise treatments for AD and PSCI.
Collapse
Affiliation(s)
- Weijie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yiming Qi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Anguo Zhao
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215000, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Xinmin Street 1#, Changchun, 130021, China.
- Department of Neurology, Cognitive Center, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
48
|
Elnaggar MG, He Y, Yeo Y. Recent trends in the delivery of RNA drugs: Beyond the liver, more than vaccine. Eur J Pharm Biopharm 2024; 197:114203. [PMID: 38302049 PMCID: PMC10947810 DOI: 10.1016/j.ejpb.2024.114203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
RNAs are known for versatile functions and therapeutic utility. They have gained significant interest since the approval of several RNA drugs, including COVID-19 mRNA vaccines and therapeutic agents targeting liver diseases. There are increasing expectations for a new class of RNA drugs for broader applications. Successful development of RNA drugs for new applications hinges on understanding their diverse functions and structures. In this review, we explore the last five years of literature to understand current approaches to formulate a spectrum of RNA drugs, focusing on new efforts to expand their applications beyond vaccines and liver diseases.
Collapse
Affiliation(s)
- Marwa G Elnaggar
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Yanying He
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
49
|
Liang D, Li G. Pulling the trigger: Noncoding RNAs in white adipose tissue browning. Rev Endocr Metab Disord 2024; 25:399-420. [PMID: 38157150 DOI: 10.1007/s11154-023-09866-6] [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] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
White adipose tissue (WAT) serves as the primary site for energy storage and endocrine regulation in mammals, while brown adipose tissue (BAT) is specialized for thermogenesis and energy expenditure. The conversion of white adipocytes to brown-like fat cells, known as browning, has emerged as a promising therapeutic strategy for reversing obesity and its associated co-morbidities. Noncoding RNAs (ncRNAs) are a class of transcripts that do not encode proteins but exert regulatory functions on gene expression at various levels. Recent studies have shed light on the involvement of ncRNAs in adipose tissue development, differentiation, and function. In this review, we aim to summarize the current understanding of ncRNAs in adipose biology, with a focus on their role and intricate mechanisms in WAT browning. Also, we discuss the potential applications and challenges of ncRNA-based therapies for overweight and its metabolic disorders, so as to combat the obesity epidemic in the future.
Collapse
Affiliation(s)
- Dehuan Liang
- The Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Beijing Institute of Geriatrics, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, People's Republic of China
- Fifth School of Clinical Medicine (Beijing Hospital), Peking University, Beijing, 100730, People's Republic of China
| | - Guoping Li
- The Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Beijing Institute of Geriatrics, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, People's Republic of China.
| |
Collapse
|
50
|
La Sala L, Carlini V, Conte C, Macas-Granizo MB, Afzalpour E, Martin-Delgado J, D'Anzeo M, Pedretti RFE, Naselli A, Pontiroli AE, Cappato R. Metabolic disorders affecting the liver and heart: Therapeutic efficacy of miRNA-based therapies? Pharmacol Res 2024; 201:107083. [PMID: 38309383 DOI: 10.1016/j.phrs.2024.107083] [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: 10/14/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Liver and heart disease are major causes of death worldwide. It is known that metabolic alteration causing type 2 diabetes (T2D) and Nonalcoholic fatty liver (NAFLD) coupled with a derangement in lipid homeostasis, may exacerbate hepatic and cardiovascular diseases. Some pharmacological treatments can mitigate organ dysfunctions but the important side effects limit their efficacy leading often to deterioration of the tissues. It needs to develop new personalized treatment approaches and recent progresses of engineered RNA molecules are becoming increasingly viable as alternative treatments. This review outlines the current use of antisense oligonucleotides (ASOs), RNA interference (RNAi) and RNA genome editing as treatment for rare metabolic disorders. However, the potential for small non-coding RNAs to serve as therapeutic agents for liver and heart diseases is yet to be fully explored. Although miRNAs are recognized as biomarkers for many diseases, they are also capable of serving as drugs for medical intervention; several clinical trials are testing miRNAs as therapeutics for type 2 diabetes, nonalcoholic fatty liver as well as cardiac diseases. Recent advances in RNA-based therapeutics may potentially facilitate a novel application of miRNAs as agents and as druggable targets. In this work, we sought to summarize the advancement and advantages of miRNA selective therapy when compared to conventional drugs. In particular, we sought to emphasise druggable miRNAs, over ASOs or other RNA therapeutics or conventional drugs. Finally, we sought to address research questions related to efficacy, side-effects, and range of use of RNA therapeutics. Additionally, we covered hurdles and examined recent advances in the use of miRNA-based RNA therapy in metabolic disorders such as diabetes, liver, and heart diseases.
Collapse
Affiliation(s)
- Lucia La Sala
- IRCCS MultiMedica, 20138 Milan, Italy; Dept. of Biomedical Sciences for Health, University of Milan, Milan, Italy.
| | | | - Caterina Conte
- IRCCS MultiMedica, 20138 Milan, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy
| | | | - Elham Afzalpour
- Dept. of Biomedical Sciences and Clinic, University of Milan, Milan, Italy
| | - Jimmy Martin-Delgado
- Hospital Luis Vernaza, Junta de Beneficiencia de Guayaquil, 090603 Guayaquil, Ecuador; Instituto de Investigacion e Innovacion en Salud Integral, Universidad Catolica de Santiago de Guayaquil, Guayaquil 090603, Ecuador
| | - Marco D'Anzeo
- AUO delle Marche, SOD Medicina di Laboratorio, Ancona, Italy
| | | | | | | | | |
Collapse
|