1
|
Guerra M, Marini A, Pagliarini V, Pitolli C, Coratti G, Bonvissuto D, Bravetti C, Pane M, Mercuri E, Sette C, Pera MC. High Expression of SMN circ4-2b-3 in SMA I Children Treated with Nusinersen is Associated with Improved Motor Outcomes. Mol Neurobiol 2025; 62:5640-5649. [PMID: 39592557 DOI: 10.1007/s12035-024-04605-7] [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: 02/21/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024]
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder resulting in the loss of α-motor neurons. Nusinersen is an antisense oligonucleotide administered intrathecally to SMA patients that corrects the splicing defect of SMN2. Not all SMA patients respond equally to the therapy and work is in progress to identify biomarkers that may help stratify to SMA patients. In this study, we evaluated the expression of SMN circular RNAs (circRNAs) as potential biomarkers of the disease. This monocentric study was conducted at Fondazione Policlinico A. Gemelli in collaboration with Catholic University of Sacred Heart between December 2019 and March 2023. The inclusion criteria comprised having a diagnosis of SMA I and being treated with Nusinersen. The quantitative analysis of SMN circ4-2b-3 was conducted analyzing patients' serum-derived exosomes. The study included 19 type I SMA patients. Among several SMN circRNAs expressed in SMA cells, only SMN circ4-2b-3 was also detected in exosomes isolated from both type I SMA cell lines and patient-derived serum. High copy number of SMN circ4-2b-3 occurred in a small subgroup of type I SMA patients who were defined as super-responders, based on their response to the Nusinersen therapy. The levels of this circRNA remained high over time. Our results suggest that SMN circ4-2b-3 is a potential biomarker to predict the therapeutic response of type I SMA patients to Nusinersen. However, since other super-responders had a lower number of SMN circ4-2b-3 copies, these findings should be confirmed in larger cohorts.
Collapse
Affiliation(s)
- Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
| | - Alberto Marini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Vittoria Pagliarini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Consuelo Pitolli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
| | - Giorgia Coratti
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Policlinico Gemelli, Largo Gemelli, 00168, Rome, Italy
| | - Davide Bonvissuto
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
| | - Chiara Bravetti
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Policlinico Gemelli, Largo Gemelli, 00168, Rome, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Policlinico Gemelli, Largo Gemelli, 00168, Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Policlinico Gemelli, Largo Gemelli, 00168, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy.
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.
| | - Maria Carmela Pera
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy.
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Policlinico Gemelli, Largo Gemelli, 00168, Rome, Italy.
| |
Collapse
|
2
|
Zhang Y, Gao L, Shi Z, Wu Q, Miao X. Simultaneous sensitive detection and imaging of dual microRNAs through DNA tetrahedral scaffold-corbelled autonomous-motion molecular machine. Talanta 2025; 286:127556. [PMID: 39805203 DOI: 10.1016/j.talanta.2025.127556] [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: 09/26/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Sensitive and accurate detection and imaging of different microRNAs (miRNAs) in cancer cells hold great promise for early disease diagnosis. Herein, a DNA tetrahedral scaffold (DTS)-corbelled autonomous-motion (AM) molecular machine based fluorescent sensing platform was designed for simultaneous detection of two types of miRNAs (miRNA-21 and miRNA-155) in HeLa cells. Locking-strand-silenced DNAzymes (P:L duplex) were firstly grafted at the loop of target-analogue-embedded double-stem hairpin substrates (TDHS) of DTS, making the sensor in a "signal off" state due to the closely distance between modified fluorophores (FAM and Cy5) with the corresponding quenchers (BHQ1 and BHQ2). The detection of miRNA-21 and miRNA-155 was mainly based on the activation of locking-strand-silenced DNAzymes, cleaving hairpin DNA into single-strand DNA segments, accompanying with the release of modified fluorophores and the signal recovery (signal on). Upon the cyclical stimulation of miRNA targets in such AM molecular machine, sensitive detection of miRNA-21 and miRNA-155 was realized in this self-feedback circuit (SFC) with the detection limit down to 38.8 aM and 27.1 aM, respectively. Moreover, the analytical performance was greatly improved for miRNAs imaging in cancer cells with enhanced tumor cell recognition ability, excellent stability in virtue of DTS, indicating a potential analytical tool in early cancer diseases diagnosis.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Pharmacy, Changzhi Medical College, Shanxi, 046012, China.
| | - Liang Gao
- Heji Hospital Affiliated to Changzhi Medical College, Shanxi, 046012, China
| | - Zhe Shi
- Department of Pharmacy, Changzhi Medical College, Shanxi, 046012, China
| | - Qiong Wu
- Department of Pharmacy, Changzhi Medical College, Shanxi, 046012, China
| | - Xiangmin Miao
- School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| |
Collapse
|
3
|
Zhang Y, Yue NN, Chen LY, Tian CM, Yao J, Wang LS, Liang YJ, Wei DR, Ma HL, Li DF. Exosomal biomarkers: A novel frontier in the diagnosis of gastrointestinal cancers. World J Gastrointest Oncol 2025; 17:103591. [DOI: 10.4251/wjgo.v17.i4.103591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Gastrointestinal (GI) cancers, which predominantly manifest in the stomach, colorectum, liver, esophagus, and pancreas, accounting for approximately 35% of global cancer-related mortality. The advent of liquid biopsy has introduced a pivotal diagnostic modality for the early identification of premalignant GI lesions and incipient cancers. This non-invasive technique not only facilitates prompt therapeutic intervention, but also serves as a critical adjunct in prognosticating the likelihood of tumor recurrence. The wealth of circulating exosomes present in body fluids is often enriched with proteins, lipids, microRNAs, and other RNAs derived from tumor cells. These specific cargo components are reflective of processes involved in GI tumorigenesis, tumor progression, and response to treatment. As such, they represent a group of promising biomarkers for aiding in the diagnosis of GI cancer. In this review, we delivered an exhaustive overview of the composition of exosomes and the pathways for cargo sorting within these vesicles. We laid out some of the clinical evidence that supported the utilization of exosomes as diagnostic biomarkers for GI cancers and discussed their potential for clinical application. Furthermore, we addressed the challenges encountered when harnessing exosomes as diagnostic and predictive instruments in the realm of GI cancers.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
- Department of Medical Administration, Huizhou Institute for Occupational Health, Huizhou 516000, Guangdong Province, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen 518000, Guangdong Province, China
| | - Li-Yu Chen
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (Jinan University of Second Clinical Medical Sciences), Shenzhen 518000, Guangdong Province, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen 518000, Guangdong Province, China
| | - Dao-Ru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Hua-Lin Ma
- Department of Nephrology, The Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong Province, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| |
Collapse
|
4
|
Cao H, Gao H, Li Y, Li L, Liu S, Jin T, Wang Y, Gong Y, Yuan S, Dong W. Zinc finger DHHC-type palmitoyltransferase 13-mediated S-palmitoylation of GNA13 from Sertoli cell-derived extracellular vesicles inhibits autophagy in spermatogonial stem cells. Cell Commun Signal 2025; 23:178. [PMID: 40205436 DOI: 10.1186/s12964-025-02177-0] [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: 01/15/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Extracellular vesicles (EVs) originating from testicular somatic cells act as pivotal intermediaries in cell signaling crosstalk between spermatogenic cells and the testicular microenvironment. The intricate balance between palmitoylation and depalmitoylation governs the positioning of protein cargos on the membrane, thereby influencing cellular activities by concentrating these proteins in EVs for delivery to recipient cells. Here, we reveal that GNA13 undergoes specific S-palmitoylation at Cys14 and Cys18 residues in Sertoli cells (SCs), a modification essential for its localization to the plasma membrane. We identify DHHC13, a member of the zinc finger DHHC-type palmitoyltransferase family that catalyzes protein S-palmitoylation, as the enzyme responsible for this critical post-translational modification. Additionally, GNA13 palmitoylation is indispensable for its selective enrichment in EVs emanating from SCs. Intriguingly, we discovered the presence of palmitoylated GNA13 in SC-derived EVs significantly downregulates autophagy levels in spermatogonial stem cells (SSCs), and the inhibition of GNA13 palmitoylation attenuates its interaction with ARHGEF12 which leads to diminished RhoA activity and consequent elevation of autophagy in SSCs. Our results illuminate the crucial role of DHHC13-mediated GNA13 S-palmitoylation in modulating autophagy levels in SSCs through SCs-derived EVs, suggesting that PM-GNA13-EV may serve as a potential candidate for further exploration in addressing fertility-related challenges during spermatogenesis.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Huihui Gao
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Li
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Tianqi Jin
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Ye Gong
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, No. 3 Taicheng Road, Yangling, Shaanxi, 712100, P. R. China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
5
|
Fu X, Shi Y, Wu H, Zhang Y, Liu Y, Wan X, Chen X, Zhou J, Qiu S, Zhao X, Tian Z, Li L, Zang H, Lin G. Inhalable liposomal delivery of osimertinib and DNA for treating primary and metastasis lung cancer. Nat Commun 2025; 16:3336. [PMID: 40199846 DOI: 10.1038/s41467-025-58312-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
Lung cancer remains one of the most common malignancies, and its brain metastases significantly worsen the prognosis for patients. Current treatments for lung cancer face many challenges, including poor drug accumulation and the inability to simultaneously control primary and metastatic tumors. Here, we show that the mRNA-binding protein insulin-like growth factor 3 is crucial for non-small cell lung cancer progression and metastasis. We construct an inhalable nanoliposome system to co-deliver osimertinib and DNA plasmid for gene knockdown. Upon inhalation, these nanoparticles efficiently penetrate pulmonary barriers and accumulate in lungs by mimicking natural lung surfactants. Within tumor cells, released osimertinib inhibits tumor growth, while the DNA triggers the production of engineered exosomes that can travel to the brain to suppress tumors. This strategy effectively inhibits both primary and metastatic tumors while enhancing antitumor immune responses. This work suggests that this inhalable nanomedicine offers a safe and versatile strategy for cancer therapy.
Collapse
Affiliation(s)
- Xianglei Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Yanbin Shi
- School of Mechanical and Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, PR China
| | - Hang Wu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yankun Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Yingying Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Xiaoyu Wan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Xiangqin Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Jiamin Zhou
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Shengnan Qiu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, PR China
| | - Zhongxian Tian
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong, PR China
| | - Lian Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Hengchang Zang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China
| | - Guimei Lin
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
6
|
Li W, He S, Lin C, Yang S, Zhang W. Mesenchymal stem cell-derived exosomes carry miR-125a-5p to improve diabetic keratopathy by regulating endoplasmic reticulum stress. Tissue Cell 2025; 93:102669. [PMID: 39674096 DOI: 10.1016/j.tice.2024.102669] [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: 08/18/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Diabetic keratopathy is a prevalent but sometimes ignored visual condition in diabetic patients, which significantly affects patients with diabetes mellitus (DM) in terms of their visual acuity. Exosomes regulate diabetes-related conditions like diabetic keratopathy (DK) by secreting their components into the body. OBJECTIVE Aim to investigate the effect and mechanism of mesenchymal stem cell (MSC)-derived exosome miR-125a-5p on DK. METHODS Transmission electron microscopy, along with nanoparticle tracking analysis, was used to determine the morphology and size of exosomes. To evaluate cell viability, proliferation, and migration, Western blotting and RT-qPCR methods were used. CCK-8, cell cloning, and scratch assays were used to measure protein levels and mRNA expression. RESULTS High glucose treatment of corneal epithelial cells weakened cell viability, proliferation and migration, and the level of miR-125a-5p was significantly reduced. It has been proposed that elevated levels of miR-125a-5p could enhance cell viability, proliferation, and migration, can inhibit endoplasmic reticulum stress induced by high glucose, which is the same as the effect of endoplasmic reticulum stress inhibitors. CONCLUSION Mouse bone marrow MSC-derived exosome miR-125a-5p repairs corneal epithelial cell viability and proliferation as well as migration ability to improve DK by inhibiting high glucose-induced endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Weina Li
- Ophthalmology Department, Nanxishan Hospital of Guangxi Zhuang Autonomous Region (The second Hospital of Guangxi Zhuang Autonomous Region), Guilin, Guangxi 541000, China; Ophthalmology Department, The Second Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, China.
| | - Shiping He
- Glaucoma and Cataract Department, Liuzhou Aier Ophthalmology Hospital, Liuzhou, Guangxi 545005, China
| | - Chaoqun Lin
- Neurosurgery Department, University of Chinese Academy of Sciences-Shenzhen Hospital (Guangming District), Shenzhen, Guangdong 518106, China
| | - Sheng Yang
- Glaucoma and Cataract Department, Liuzhou Aier Ophthalmology Hospital, Liuzhou, Guangxi 545005, China
| | - Wenbin Zhang
- Ophthalmology Department, Nanxishan Hospital of Guangxi Zhuang Autonomous Region (The second Hospital of Guangxi Zhuang Autonomous Region), Guilin, Guangxi 541000, China
| |
Collapse
|
7
|
Wen Y, Li Y, Cheng S, Crow J, Samuel G, Vishwakarma V, Turaga SM, Bantis L, Godwin AK, Zeng Y. Partition-Less Digital Immunoassay Using Configurable Topographic Nanoarrays for Extracellular Vesicle Diagnosis of Ewing Sarcoma. ACS NANO 2025; 19:11973-11986. [PMID: 40115997 DOI: 10.1021/acsnano.4c16904] [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: 03/23/2025]
Abstract
Emerging digital bioassays provide opportunities for bioanalysis and clinical diagnostics due to their improved analytical performance. Prevailing digitization strategies rely on partitioning single molecules into discrete compartments for which bead-based capture may be used. Herein we report a partition-free digital enzyme-linked immunosorbent assay (dELISA) named microfluidic Topographically Intensified, Partition-less dELISA (μTIP-dELISA). Our method builds on a single-molecule signal amplification technique that employs a simple micropost device to generate a topographic nanogap array to significantly enhance surface-bound enzymatic reactions. Compared to existing dELISA methods, our approach features appreciable simplicity and enhanced adaptability as it obviates the needs for sophisticated device fabrication, complicated workflow for off-line immunomagnetic capture, and ultralow-volume compartmentalization. Moreover, μTIP-dELISA integrates the inherent advantages of microfluidics in improving the assay performance and throughput and enhancing the scalability and automation. As a proof-of-concept for potential biomedical applications, we adapted μTIP-dELISA to extracellular vesicle (EV)-based liquid biopsy diagnosis of a pediatric cancer, Ewing sarcoma (EWS). Our technology confers >300-fold improvement over the conventional ELISA in detecting four EWS protein biomarkers. We demonstrated highly sensitive and specific detection of EWS cases with a machine-learning-defined four-marker EV signature. The four EV markers were further tested to assess age as a prognostic factor, resulting in an overall accuracy of 97% for classifying the control, pediatric and adult subjects. Overall, we envision that our μTIP single-molecule signal amplification strategy could promote the development and adaptation of digital bioassays and that the μTIP-dELISA could provide a useful tool for clinical diagnostics for many malignancies.
Collapse
Affiliation(s)
- Yunjie Wen
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yutao Li
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Shibo Cheng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Jennifer Crow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66103, United States
| | - Glenson Samuel
- Children's Mercy─Kansas City, Kansas City, Missouri 64108, United States
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66103, United States
| | - Soumya M Turaga
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66103, United States
| | - Leonidas Bantis
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66103, United States
- University of Kansas Cancer Center, Kansas City, Kansas 66160, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Yong Zeng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- University of Florida Health Cancer Center, Gainesville, Florida 32611, United States
| |
Collapse
|
8
|
Zhang X, Zhao J, Ge R, Zhang X, Sun H, Guo Y, Wang Y, Chen L, Li S, Yang J, Sun D. Arg-Gly-Asp engineered mesenchymal stem cells as targeted nanotherapeutics against kidney fibrosis by modulating m6A. Acta Biomater 2025:S1742-7061(25)00217-X. [PMID: 40158765 DOI: 10.1016/j.actbio.2025.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/17/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Background The recent surge in research on extracellular vesicles has generated considerable interest in their clinical applications. Extracellular vesicles derived from mesenchymal stem cells (MSC-EV) have emerged as a promising cell-free therapy for chronic kidney disease (CKD), offering an alternative to traditional Mesenchymal stem/stromal cells (MSCs) in extracellular vesicle-based nanotherapeutics. However, challenges such as in vivo off-target effects and limited bioavailability have impeded the wider adoption of MSC-EV in clinical settings. Methods Arginyl-glycyl-aspartic acid peptide-modified MSC-EV (RGD-MSC-EV) were developed using a donor cell-assisted membrane modification strategy. The targeting capability and therapeutic efficacy of RGD-MSC-EV were thoroughly evaluated both in vitro and in vivo. Additionally, the mechanisms of RNA N6-methyladenosine (m6A) methylation-mediated angiogenesis were extensively investigated to elucidate how RGD-MSC-EV mitigates renal fibrosis. Results RGD-MSC-EV demonstrated exceptional targeted delivery efficiency, exhibiting optimal biodistribution and retention within the target tissue. This breakthrough positions them as significantly enhanced anti-fibrotic therapeutics. Notably, RGD-MSC-EV sustains the viability of renal peritubular capillary (PTCs) endothelial cells by transporting microRNA-126-5p (miR-126-5p) and modulating alkB homolog 5 (ALKBH5)-mediated m6A modification of SIRT1(Sirtuin 1), a crucial regulator in angiogenesis. By revitalizing endothelial cells and promoting microcirculation, this approach restored oxygen metabolism homeostasis, ultimately delaying fibrogenesis associated with CKD. Conclusions RGD-MSC-EV offers a feasible and effective strategy to alleviate renal interstitial fibrosis by restoring m6A and mitigating the loss of renal PTCs. STATEMENT OF SIGNIFICANCE: Chronic kidney disease (CKD) often leads to renal fibrosis, which worsens disease progression. This study introduces a novel strategy using engineered extracellular vesicles (EVs) derived from mesenchymal stem cells (MSC-EV). By modifying these EVs with RGD peptides, we significantly enhance their targeting ability to hypoxic kidney tissues. The research reveals how these EVs deliver microRNA (miR-126-5p) to restore key molecular mechanisms, stabilizing SIRT1 expression through m6A RNA modifications. This approach promotes blood vessel health and delays fibrosis. Compared to current treatments, RGD-MSC-EV offers a safe, effective, and cell-free therapeutic alternative. These findings advance the understanding of EV-based therapies and their clinical potential, bridging basic research and real-world CKD treatment applications.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Jiaqi Zhao
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Rui Ge
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Xiangyu Zhang
- Department of Nephrology, Ningbo City first Hospital, Ningbo, China
| | - Haihan Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China
| | - Yuhan Guo
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Yanping Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Lu Chen
- Department of Rheumatology, Ningbo Medical Treatment Center Li Huili Hospital, Ningbo, China
| | - Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| | - Jing Yang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Clinical Research Center For Kidney Disease, Xuzhou Medical University, Xuzhou, China; Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
9
|
Wang Y, Huang D, Li M, Yang M. MicroRNA-99 family in cancer: molecular mechanisms for clinical applications. PeerJ 2025; 13:e19188. [PMID: 40161350 PMCID: PMC11955196 DOI: 10.7717/peerj.19188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of non-coding RNA sequences that regulate gene expression post-transcriptionally. The miR-99 family, which is highly evolutionarily conserved, comprises three homologs: miR-99a, miR-99b, and miR-100. Its members are under-expressed in most cancerous tissues, suggesting their cancer-repressing properties in multiple cancers; however, in some contexts, they also promote malignant lesion progression. MiR-99 family members target numerous genes involved in various tumor-related processes such as tumorigenesis, proliferation, cell-cycle regulation, apoptosis, invasion, and metastasis. We review the recent research on this family, summarize its implications in cancer, and explore its potential as a biomarker and cancer therapeutic target. This review contributes to the clinical translation of the miR-99 family members.
Collapse
Affiliation(s)
- Yueyuan Wang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Dan Huang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| |
Collapse
|
10
|
Pei Z, Sun Y, Zhang S, Gong C, Mao G, Zhang X, Meng W, Cen J, Li S, Sun M, Xu Q, Xiao K. Extracellular vesicles derived from mesenchymal stem cells ameliorate sulfur mustard-induced lung injury by regulating apoptosis via miR-146a-5p. Int Immunopharmacol 2025; 150:114285. [PMID: 39955917 DOI: 10.1016/j.intimp.2025.114285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
Sulfur mustard (SM) is an extremely toxic chemical warfare agent. Although SM-induced toxicity has long been studied, due to its complexity, the characterization of the precise molecular pathway it targets has been remaining an ongoing area of research. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells (hucMSC-EVs) are natural substances that participate in intercellular communication by delivering microRNA to target cells. Importantly, the microRNA content in EVs can be modified. MiR-146a-5p delivered by EVs were utilized and hucMSCs were further modified with miR-146a-5p mimics or inhibitors to collect EVs that over-(miR-146a-5p+-EVs) or underexpress (miR-146a-5p--EVs) miR-146a-5p. Transcriptome sequencing was used to identify potential mediators of the effects of miR-146a-5p delivered by hucMSC-EVs. Our results showed that hucMSC-EVs reduced SM-induced lung injury by mitigating apoptosis. These effects were enhanced by miR-146a-5p+-EVs and weakened by miR-146a-5p--EVs. Meanwhile, the relationship between apoptosis enhancing nuclease (AEN) and miR-146a-5p was discovered, a novel target of miR-146a-5p. Our study showed that hucMSC-EVs ameliorating sulfur mustard induced lung injury through miR-146a-5p, and AEN was one of the functional molecules in this process.
Collapse
Affiliation(s)
- Zhipeng Pei
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Yunrui Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; Southern Theater Air Force Hospital, Guangzhou 510050, China
| | - Shanshan Zhang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Chuchu Gong
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Guanchao Mao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Xinkang Zhang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Wenqi Meng
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Jinfeng Cen
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Songling Li
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Mingxue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China.
| | - Qingqiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China.
| | - Kai Xiao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; Marine Biomedical Science and Technology Innovation Platform of Lingang Special Area, Shanghai 201306, China.
| |
Collapse
|
11
|
Bian Z, Zhai Y, Zhang Y, Wang T, Li H, Ouyang J, Liu C, Wang S, Hu Z, Chang X, Zhang C, Liu M, Li C. Senescent cartilage endplate stem cells-derived exosomes induce oxidative stress injury in nucleus pulposus cells and aggravate intervertebral disc degeneration by regulating FOXO3. Free Radic Biol Med 2025; 233:39-54. [PMID: 40118349 DOI: 10.1016/j.freeradbiomed.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Intervertebral disc degeneration (IVDD) is the leading cause of low back pain and associated disability worldwide. The cartilage endplate (CEP) is a critical structure in maintaining the homeostasis of the intervertebral disc, by exosomes (Exos)-mediated intracellular communication between cartilage endplate stem cells (CESCs) and nucleus pulposus cells (NPCs). However, whether the senescence of CESCs influences the functionality of CESCs-derived Exos (CESCs-Exos) and participates in the progress of IVDD remains unclear. In this study, we explored the role and mechanism of the Exos-based intracellular communication between senescent CESCs and NPCs in IVDD. CESCs isolated from aged individuals (S-CESCs) exhibited high levels of senescence compared with CESCs isolated from young individuals (Y-CESCs). Exos from Y-CESCs (Y-Exos) and from S-CESCs (S-Exos) were extracted and identified. Surprisingly, we found that S-Exos lost the therapeutic effects as the Y-Exos exhibited in mitigating IVDD, and even aggravated IVDD by inducing oxidative stress injury in NPCs. MicroRNA-sequencing revealed significant upregulation of miR-29b-3p expression in S-Exos. Through microRNA target prediction, dual luciferase assays, RNA-sequencing, lentivirus-mediated overexpression and suppression, we demonstrated that miR-29b-3p regulates the expression of FOXO3 and downstream antioxidant enzymes to induce oxidative stress injury in NPCs. In vivo experiments further verified that countering miR-29b-3p by antagomir reversed the detrimental effects of S-Exos in exacerbating IVDD. This work elucidates the role and mechanism of senescent CESCs in disrupting redox homeostasis in the nucleus pulposus and exacerbating IVDD by Exos-mediated intracellular communication and offers an experimental foundation for the selection of proper CESCs-Exos to obtain better therapeutic effects in IVDD.
Collapse
Affiliation(s)
- Zhiqun Bian
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Yu Zhai
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China.
| | - Yuyao Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Tianling Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Hao Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Jian Ouyang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Chao Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Siya Wang
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zhilei Hu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China.
| |
Collapse
|
12
|
Zhang Z, Liu X, Peng C, Du R, Hong X, Xu J, Chen J, Li X, Tang Y, Li Y, Liu Y, Xu C, Liu D. Machine Learning-Aided Identification of Fecal Extracellular Vesicle microRNA Signatures for Noninvasive Detection of Colorectal Cancer. ACS NANO 2025; 19:10013-10025. [PMID: 40033785 DOI: 10.1021/acsnano.4c16698] [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: 03/05/2025]
Abstract
Colorectal cancer (CRC) remains a formidable threat to human health, with considerable challenges persisting in its diagnosis, particularly during the early stages of the malignancy. In this study, we elucidated that fecal extracellular vesicle microRNA signatures (FEVOR) could serve as potent noninvasive CRC biomarkers. FEVOR was first revealed by miRNA sequencing, followed by the construction of a CRISPR/Cas13a-based detection platform to interrogate FEVOR expression across a diverse spectrum of clinical cohorts. Machine learning-driven models were subsequently developed within the realms of CRC diagnostics, prognostics, and early warning systems. In a cohort of 38 CRC patients, our diagnostic model achieved an outstanding accuracy of 97.4% (37/38), successfully identifying 37 of 38 CRC cases. This performance significantly outpaced the diagnostic efficacy of two clinically established biomarkers, CEA and CA19-9, which showed accuracies of mere 26.3% (10/38) and 7.9% (3/38), respectively. We also examined the expression levels of FEVOR in several CRC patients both before and after surgery, as well as in patients with colorectal adenomas (CA). Impressively, the results showed that FEVOR could serve as a robust prognostic indicator for CRC and a potential predictor for CA. This endeavor aimed to harness the predictive power of FEVOR for enhancing the precision and efficacy of CRC management paradigms. We envision that these findings will propel both foundational and preclinical research on CRC, as well as clinical studies.
Collapse
Affiliation(s)
- Zhaowei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xuyang Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chuanyue Peng
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Rui Du
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqin Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jia Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jiaming Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaomin Li
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yujing Tang
- Medical and Hygienic Materials Research Institute, SINOPEC (Beijing) Research Institute of Chemical Industry Co., Ltd., Beijing 100013, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yang Liu
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Hepatobiliary and Pancreatic Oncology, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Riazanski V, Purvina L, Cavinato L, Sui Z, Sun L, Nelson DJ. Functional interaction of hybrid extracellular vesicle-liposome nanoparticles with target cells: absence of toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642711. [PMID: 40161690 PMCID: PMC11952422 DOI: 10.1101/2025.03.11.642711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Drug delivery platforms, complex lipid nanoparticles (LNPs) and extracellular vesicles (EVs) have both faced a number of key challenges ranging from organ specificity to loading capacity and stability. A key challenge in EV biology as well as LNP design remains vesicle to cell interaction and the creation of a polar permeability pathway necessary for cargo exchange. Membrane to membrane recognition and intercalation are tantamount to delivery and integral to function of both EVs and LNPs, both complex and single component. We reasoned that the overlapping advantages of both nanoparticles centered on compositional lipids. EVs are encapsulations using biological membrane lipids and expressed proteins and have a larger carrier capacity. LNPs are composed of synthetic lipids differing in charge and amount mimicking those present in biological membranes and include a synthetic lipid of choice that carries a charge, designed to enhance biological membrane disruption and subsequent cargo off-loading. Our goal was to design hybrid EVs (HEVs) that combined both elements. We manufactured positively charged liposomes (Lip) carrying mRNA coding for fluorescent proteins to load isolated EVs in order to create a combinatorial delivery platform. Using knowledge from LNP-based mRNA vaccine delivery, we have formulated and characterized HEVs. Future therapeutic strategies could involve isolating EVs from patients, hybridizing them with synthetic lipids loaded with desired payloads, and reintroducing them to the patient. This approach is particularly relevant for enhancing the function of pulmonary innate immunity in diseases like cystic fibrosis, chronic granulomatous disease, and pulmonary fibrosis. By conducting both in-vitro and in-vivo assays, we demonstrate that HEVs exhibit comparable transfection efficacy to LNPs composed of complex synthetic lipids, while significantly reducing cytotoxicity. This highlights their potential as safer and more efficient delivery vehicles.
Collapse
|
14
|
Huang Y, Liu P, Xu Y, Qian C, Wu T, Li T. Plasma Exosomes Derived from Patients with Primary Immune Thrombocytopenia Attenuate TBX21 + Regulatory T Cell-Mediated Immune Suppression via MiR-363-3p. Inflammation 2025:10.1007/s10753-025-02275-8. [PMID: 40032779 DOI: 10.1007/s10753-025-02275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Primary Immune Thrombocytopenia (ITP) is characterized by reduced immunosuppressive function of regulatory T cells (Tregs), contributing to immune imbalance and decreased platelet counts. However, the mechanisms behind this reduced efficacy of Tregs remain unclear. Our study used a variety of methods, including Treg function assays, cytokine analysis, and single-cell sequencing, to explore these mechanisms. We found that exosomes from ITP patients inhibited TBX21 expression in Tregs, and impaired their ability to suppress Th1 cells. At the single-cell level, Tregs with high TBX21 expression were identified, and the activity of the TBX21 regulon was found to be enhanced in early-stage Treg subpopulations. We also discovered that ARID3A interacted with SPI1 and TBX21 gene regions, indicating a regulatory relationship between ARID3A, SPI1, and TBX21. Additionally, exosomes in ITP patients' plasma contained elevated levels of miR-363-3p, which negatively correlated with platelet count. These exosomes transferred miR-363-3p to Tregs, downregulating ARID3A, SPI1, and TBX21 expression, thereby weakening Tregs' ability to suppress conventional CD4 + T cells. In conclusion, exosomes from ITP patients reduced Treg function through the ARID3A/SPI1/TBX21 axis by miR-363-3p, diminishing their ability to regulate Th1 cells and contributing to the immune dysfunction observed in ITP.
Collapse
Affiliation(s)
- Yuanlan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai, 200000, China
| | - Peng Liu
- Department of Blood Transfusion, No.971 Hospital of the PLA Navy, Qingdao, China
| | - Ying Xu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Cheng Qian
- Department of Laboratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Tianqin Wu
- Suzhou100 Hospital, Suzhou, 215006, China
| | - Tengda Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
15
|
Sui Z, Chen B, Zhao J, Yang H, Guo L, Xu J. Dual-Accelerated Signal Amplification in Biosensing via Spatial Confining Catalytic Hairpin Assembly-Activated Spherical CRISPR/Cas12a System for Trans-Cleavage of Hairpin DNA Reporters. Anal Chem 2025; 97:4668-4677. [PMID: 39982896 DOI: 10.1021/acs.analchem.4c07111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression and are implicated in various diseases, including cancer. Due to their critical role in diagnostics, there is a growing need for sensitive, specific, and rapid detection methods for miRNAs. In this study, we present a dual-accelerated signal amplification platform for miRNA biosensing, which integrates spatial confining catalytic hairpin assembly (SC-CHA) with spherical CRISPR/Cas12a (S-CRISPR/Cas12a) system for (SC-CHA@S-CRISPR/Cas12a) trans-cleavage of hairpin DNA reporters. The method employs a biotinylated palindrome-rich assembly sequence (PAS) to form DNA nanoballs, which serve as a scaffold for the operation of SC-CHA upon miRNA binding. The SC-CHA products bind with crRNA and Cas 12a protein, activating S-CRISPR/Cas12a system to cleave the hairpin DNA reporter and generate a detectable fluorescence signal. The uniqueness of this system lies in the combined use of DNA nanoballs and hairpin DNA reporters, both of which significantly accelerate reaction kinetics, resulting in rapid signal generation. Additionally, the spherical DNA nanostructure, integrated with the S-CRISPR/Cas12a system, greatly enhances biostability and accelerating reaction kinetics. These features enable the platform to exhibit high sensitivity, with a limit of detection (LOD) as low as 13.75 fM, and excellent specificity, successfully distinguishing miRNA-21 from other miRNAs. The assay is also biostable, demonstrating reliable performance in complex biological samples such as human serum. This dual-acceleration approach offers a promising solution for sensitive, rapid, and specific miRNA biosensing, with potential applications in early cancer diagnosis and clinical monitoring.
Collapse
Affiliation(s)
- Zhuqi Sui
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
| | - Baoqiang Chen
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
| | - Jia Zhao
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Haidong Yang
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
| | - Longhua Guo
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
| | - Jianguo Xu
- Jiaxing Key Laboratory of Molecular Recognition and Sensing, College of Biological and Chemical Engineering, Jiaxing University, Jiaxing 314001, China
| |
Collapse
|
16
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [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: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
17
|
Kostyusheva A, Romano E, Yan N, Lopus M, Zamyatnin AA, Parodi A. Breaking barriers in targeted Therapy: Advancing exosome Isolation, Engineering, and imaging. Adv Drug Deliv Rev 2025; 218:115522. [PMID: 39855273 DOI: 10.1016/j.addr.2025.115522] [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: 07/30/2024] [Revised: 12/23/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
Exosomes have emerged as promising tools for targeted drug delivery in biomedical applications and medicine. This review delves into the scientific advancements, challenges, and future prospects specifically associated with these technologies. In this work, we trace the research milestones that led to the discovery and characterization of exosomes and extracellular vesicles, and discuss strategies for optimizing the synthetic yield and the loading of these particles with various therapeutics. In addition, we report the current major issues affecting the field and hampering the clinical translation of these technologies. Highlighting the pivotal role of imaging techniques, we explore how they drive exosome therapy and development by offering insights into biodistribution and cellular trafficking dynamics. Methodologies for vesicle isolation, characterization, loading, and delivery mechanisms are thoroughly examined, alongside strategies aimed at enhancing their therapeutic efficacy. Special emphasis was dedicated to their therapeutic properties, particularly to their ability to deliver biologics into the cytoplasm. Furthermore, we delve into the intricate balance between surface modifications and targeting properties including also transgenic methods aimed at their functionalization and visualization within biological systems. This review underscores the transformative potential of these carriers in targeted drug delivery and identifies crucial areas for further research and clinical translation.
Collapse
Affiliation(s)
- Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia
| | | | - Neng Yan
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Kalina Campus, Vidyanagari, Mumbai 400098, India
| | - Andrey A Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Department of Biological Chemistry, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Alessandro Parodi
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia.
| |
Collapse
|
18
|
Qu M, Su S, Jiang L, Yu X, Zhang J, Zhu H, Han K, Zhang X. Exosomal miR-27a-5p attenuates inflammation through Toll-like receptor 7 in foodborne Salmonella infections. Vet Microbiol 2025; 302:110394. [PMID: 39823714 DOI: 10.1016/j.vetmic.2025.110394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Salmonella is a common food-borne pathogen that is highly pathogenic and infectious, causing serious harm to livestock breeding and food safety. Uncovering the mechanisms of Salmonella infection and immune evasion can effectively prevent Salmonella contamination of livestock and poultry food. Here, small RNA sequencing results showed that exosomes produced by naïve murine macrophages RAW 264.7 cells contained a unique enrichment of a set of microRNAs (miRNAs) after Salmonella infection. Quantitative real-time polymerase chain reaction (qPCR) analysis verified that the tested miRNA (i.e. miR-27a-5p, miR-92a-1-5p and miR-1249-5p) showed similar expression patterns, consistent with small RNA sequencing data. TargetScan database predicted that the most promising targets for the differentially expressed miRNAs were abundant in the immune system, infectious diseases, and signal transduction pathways. Dual-luciferase reporter assays confirmed that Toll-like receptor 7 (TLR7) was the target of miR-27a-5p. Western blotting and enzyme-linked immunosorbent assay (ELISA) results revealed that overexpression of miR-27a-5p suppressed inflammation by targeting TLR7/nuclear factor kappa-B (NF-κB) signaling pathway and leading interleukin-6 (IL-6) and IL-1β cytokines slightly reduction in recipient macrophages, suggesting that exosomal miR-27a-5p uptake by naïve macrophages may inhibit pro-inflammatory macrophage differentiation. Therefore, these results contribute to our systematic understanding of the mechanism of exosomal miRNA in Salmonella infection, providing a potential target for preventing immune escape from Salmonella.
Collapse
Affiliation(s)
- Mingjuan Qu
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China; Shandong Engineering Research Center for Aquaculture Environment Control, Yantai 264025, China
| | - Shengfa Su
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China
| | - Linlin Jiang
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China; Shandong Engineering Research Center for Aquaculture Environment Control, Yantai 264025, China
| | - Xin Yu
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China; Shandong Engineering Research Center for Aquaculture Environment Control, Yantai 264025, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China
| | - Kexue Han
- Jinan Baiming Biopharmaceutical Co., Ltd, Ji'nan, Shandong 250101, China
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China; Collaborative Innovation Center for the Pet Infectious Diseases and Public Health in the Middle and Lower Stream Regions of the Yellow River, Yantai 264025, China; Shandong Engineering Research Center for Aquaculture Environment Control, Yantai 264025, China.
| |
Collapse
|
19
|
Corsi J, Semnani PS, Peroni D, Belli R, Morelli A, Lassandro M, Sidarovich V, Adami V, Valentini C, Cavallerio P, Grosskreutz J, Fabbiano F, Grossmann D, Hermann A, Tell G, Basso M, D'Agostino VG. Small molecule inhibitors of hnRNPA2B1-RNA interactions reveal a predictable sorting of RNA subsets into extracellular vesicles. Nucleic Acids Res 2025; 53:gkaf176. [PMID: 40103230 PMCID: PMC11915509 DOI: 10.1093/nar/gkaf176] [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: 02/21/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
Extracellular vesicles (EVs) are cell-secreted membranous particles contributing to intercellular communication. Coding and noncoding RNAs can be detected as EV cargo, and RNA-binding proteins (RBPs), such as hnRNPA2B1, have been circumstantially implicated in EV-RNA sorting mechanisms. However, the contribution of competitive RBP-RNA interactions responsible for RNA-sorting outcomes is still unclear, especially for predicting the EV-RNA content. We designed a reverse proteomic analysis exploiting the EV-RNA to identify intracellular protein binders in vitro. Using cells expressing a recombinant hnRNPA2B1 to normalize competitive interactions, we prioritized a network of heterogeneous nuclear ribonucleoproteins and purine-rich RNA sequences subsequently validated in secreted EV-RNA through short fluorescent RNA oligos. Then, we designed a GGGAG-enriched RNA probe that efficiently interacted with a full-length human hnRNPA2B1 protein. We exploited the interaction to conduct a pharmacological screening and identify inhibitors of the protein-RNA binding. Small molecules were orthogonally validated through biochemical and cell-based approaches. Selected drugs remarkably impacted secreted EV-RNAs and reduced an RNA-dependent, EV-mediated paracrine activation of NF-kB in recipient cells. These results demonstrate the relevance of post-transcriptional mechanisms for EV-RNA sorting and the possibility of predicting the EV-RNA quality for developing innovative strategies targeting discrete paracrine functions.
Collapse
Affiliation(s)
- Jessica Corsi
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Pouriya Sharbatian Semnani
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Daniele Peroni
- MS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Romina Belli
- MS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Alessia Morelli
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Michelangelo Lassandro
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Viktoryia Sidarovich
- HTS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Valentina Adami
- HTS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Chiara Valentini
- NGS Core Facility, Department of Cellular, Computational and Integrative Biomedicine CIBIO, University of Trento, 38122 Trento, Italy
| | - Paolo Cavallerio
- NGS Core Facility, Department of Cellular, Computational and Integrative Biomedicine CIBIO, University of Trento, 38122 Trento, Italy
| | - Julian Grosskreutz
- Excellence Cluster Precision Medicine in Inflammation, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Fabrizio Fabbiano
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Dajana Grossmann
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DMED), University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy
| | - Manuela Basso
- Laboratory of Transcriptional Neurobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Vito G D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
20
|
Liu W, Yang H, Liu X, Cai H, Bao Y, Jiang Y, Zhou W, Yuan J, Zhang Z, Fang X. Ultrasensitive Quantification of microRNA Copy Number in Individual Extracellular Vesicles Using DNA Tetrahedron-Based Single-Molecule Imaging. Anal Chem 2025; 97:4233-4240. [PMID: 39936597 DOI: 10.1021/acs.analchem.4c07068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
The ultrasensitive detection of microRNAs (miRNAs) in extracellular vesicles (EVs) can accurately reflect the progress and metastasis of miRNA-mediated intercellular communication, providing an unprecedented opportunity for liquid biopsy. However, due to the low abundance and high heterogeneity of miRNAs in EVs, the ultrasensitive quantification and establishment of a distribution model for miRNA within native EVs remain challenging. Here, we have developed a DNA tetrahedron-based single-molecule fluorescence imaging strategy to overcome this challenge. The internalization efficiency of the probe was as high as 70% without disrupting the native structure of EVs, and combined with single-molecule fluorescence imaging, we achieved in situ imaging analysis of single-copy miRNA in individual EVs without amplification for the first time. A new distribution model for miRNAs has been revealed by statistical analysis of the copy number of miRNAs in EVs across multiple cell lines, characterized by low occupancy and a heterogeneous distribution. More importantly, we found that drug resistance cancer cells promote an increase in the number of drug resistance-related miRNAs within EVs without a corresponding increase in the number of EVs secreted, providing new insights into the EV miRNA sorting mechanisms. We anticipate that this technology will rapidly advance miRNA-mediated intercellular communication based on EVs.
Collapse
Affiliation(s)
- Weifeng Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongwei Yang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaolong Liu
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heqi Cai
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Bao
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yifei Jiang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wei Zhou
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinghe Yuan
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhen Zhang
- Huairou Research Center, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohong Fang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
21
|
Li X, Liu X, Zhou J, Zhang P, Chen S, Bai D. Human dental follicle stem cell-derived exosomes reduce root resorption by inhibiting periodontal ligament cell pyroptosis. Stem Cell Res Ther 2025; 16:79. [PMID: 39985080 PMCID: PMC11846241 DOI: 10.1186/s13287-025-04216-6] [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: 11/17/2024] [Accepted: 02/10/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND To explore the therapeutic effects and mechanisms of the exosomes derived from dental follicle stem cells (DFSC-Exos) in reducing osteoclastogenesis and root resorption (RR) by inhibiting periodontal ligament cell (PDLC) pyroptosis. METHODS DFSC-Exos, with force stimulation (Force-Exos) or without (Ctrl-Exos), were co-cultured with human PDLCs in vitro and injected into the periodontal ligament (PDL) of rats following the establishment of RR models in vivo. Subsequently, resorption volume, PDLC pyroptotic ratio, and NLRP3-mediated pyroptosis pathway activation were performed to investigate the therapeutic effects of DFSC-Exos on PDLC pyroptosis during RR. Furthermore, the number of M1/M2 macrophages, osteoclast formation, and transwell polarization elucidated the role of Force-Exo treatment in macrophage polarization and osteoclastogenesis by inhibiting pyroptosis. Exosomal miRNA sequencing and bioinformatic analysis were used to identify differentially abundant exosome-derived miRNAs, as well as the dominant biological processes and pathways modulated by miRNA. The administration of miRNA inhibitors further verified the regulation of exosomal miRNA on RR via modulating pyroptosis. Moreover, the potential mechanisms involving candidate miRNAs and relevant pathways were explored. RESULTS Exosomes released by force-stimulated DFSCs (Force-Exos) inhibited NOD-like receptor 3 (NLRP3)-mediated PDLC pyroptosis, which impacted M1 macrophage activation and osteoclast formation. Based on exosomal miRNA sequencing, miR-140-3p in Force-Exos were transferred to PDLCs, and the administration of miR-140-3p inhibitors significantly reversed the reduction in PDLC pyroptosis, M1 macrophage polarization, osteoclast number, and resorption volume caused by Force-Exos. More importantly, mechanistic studies demonstrated that miR-140-3p mediated the function of Force-Exos by targeting DNA methyltransferase 1 (DNMT1) to alter the DNA methylation of suppressor of cytokine signaling (SOCS1) and the downstream nuclear factor κB (NF-κB) signaling pathway in PDLCs. Blocking the DNMT1/SOCS1/NFκB axis with DFSC-derived exosomal miR-140-3p downregulated NLRP3-mediated PDLC pyroptosis to impact M1 polarization and osteoclast formation, thereby alleviating RR. CONCLUSION DFSC-Exos downregulated NLRP3-mediated PDLC pyroptosis via miR-140-3p to block DNMT1/SOCS1/NFκB axis, which impacted M1 polarization and osteoclast formation, thereby alleviating RR.
Collapse
Affiliation(s)
- Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinyang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jianing Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ping Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Liao Y, Liu Y, Li D, Luo S, Huang Y, Wu J, Su J, Yang Y, Wu J, Zhu Z, Yanglan M, Deng H, Wu X, Xu J, Cao F, Cai C, Li Z, Yang R, Deng X, Wei J, Wang L. COVID-19 patient serum-derived extracellular vesicles deliver miR-20b-5p induces neutrophil extracellular traps. Cell Commun Signal 2025; 23:93. [PMID: 39962581 PMCID: PMC11834185 DOI: 10.1186/s12964-025-02095-1] [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: 06/07/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Severe cases of COVID-19 are characterized by an excessive presence of neutrophils. Neutrophil extracellular traps (NETs), released by activated neutrophils due to SARS-CoV-2 infection, contribute to lung epithelial cell death and are key drivers in COVID-19-associated immunothrombosis. However, the mechanism underlying NET formation in COVID-19 remain unclear. METHODS Extracellular vesicles (EVs) were isolated from the serum of COVID-19 patients and healthy volunteers, while neutrophils were isolated from blood samples of healthy volunteers. Neutrophils were treated with EVs, and the formation of NETs was observed. To identify the components responsible for the COVID-19-EVs-induced NET formation, we analyzed the expression profiles of microRNA (miRNAs) in COVID-19-EVs. We identified eight highly expressed miRNAs in COVID-19-EVs and explored their potential roles in COVID-19-EVs-mediated NET formation. Additionally, we explored the role of miR-20b-5p in COVID-19-EVs-induced NET formation. RESULTS In this study, we demonstrate that patients with COVID-19 have a higher concentration of serum EVs (COVID-19-EVs) than healthy controls (Normal-EVs). We also found that COVID-19-EVs are internalized by neutrophils to induced NET formation. Through comprehensive miRNA profiling of COVID-19-EVs versus Normal-EVs, we identified 78 differentially expressed miRNAs, with 27 of these being upregulated and 51 being downregulated. Subsequently, we discovered that COVID-19-EVs that were highly abundant with certain miRNAs promote NET formation. Specifically, miR-20b-5p was found to be the strongest inducer of NET formation of the identified miRNAs. Inhibition of miR-20b-5p resulted in a significant decrease in COVID-19-EVs-mediated induction of NET formation. CONCLUSION Herein, we reveal a previously unknown role of COVID-19-EVs in NET formation, which contributes to COVID-19 progression. This study suggests that miR-20b-5p may serve as a potential therapeutic target for COVID-19 treatment.
Collapse
Affiliation(s)
- Yao Liao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuheng Liu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Dinghao Li
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shiqi Luo
- Institute of Virology, Helmholtz Centre Munich - German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Chair for Preventions of Infectious Microbial Diseases, School of Life Sciences, Central Institute of Disease Prevention, Technical University of Munich, 85354, Freising, Germany
| | - Yun Huang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Junwei Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jin Su
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ji Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zifeng Zhu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxi Yanglan
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyi Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xinyi Wu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junhao Xu
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Feiyang Cao
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunmei Cai
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhen Li
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruibing Yang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoyan Deng
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jie Wei
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Lifu Wang
- KingMed School of Laboratory Medicine, The Second Affiliated Hospital, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
23
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
24
|
Gong S, Li Y, Yan K, Shi Z, Leng J, Bao Y, Ning K. The Crosstalk Between Endothelial Cells, Smooth Muscle Cells, and Macrophages in Atherosclerosis. Int J Mol Sci 2025; 26:1457. [PMID: 40003923 PMCID: PMC11855868 DOI: 10.3390/ijms26041457] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease closely tied to cellular metabolism. Recent genome-wide association study data have suggested the significant roles of endothelial cells, smooth muscle cells, and macrophages in the regression and exacerbation of AS. However, the impact of cellular crosstalk and cellular metabolic derangements on disease progression in AS is vaguely understood. In this review, we analyze the roles of the three cell types in AS. We also summarize the crosstalk between the two of them, and the associated molecules and consequences involved. In addition, we emphasize potential therapeutic targets and highlight the importance of the three-cell co-culture model and extracellular vesicles in AS-related research, providing ideas for future studies.
Collapse
Affiliation(s)
- Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jing Leng
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China;
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
25
|
Wang Z, Huang Y, He S, Zhou Y, Zhao L, Wang F. Dynamic and functional analyses of exosomal miRNAs regulating cellular microenvironment of ovarian cancer cells. J Ovarian Res 2025; 18:25. [PMID: 39930447 PMCID: PMC11808964 DOI: 10.1186/s13048-025-01608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Exosomes, extracellular vesicles with an average diameter of 30 ~ 150 nm, are pivotal in mediating the cellular microenvironment (CM) through their cargo-carrying capability. Despite extensive studies, the dynamic and regulatory mechanisms of exosomal cargoes, including lipids, proteins, nucleic acids, and metabolites, remain poorly understood. METHODS In this study, we collected culture medium of ovarian cancer cells at four different time points (12, 24, 36, 48 h). Exosomes were isolated using ultracentrifugation, and miRNA sequencing was performed for exosomes from each group (T12, T24, T36, and T48). RESULTS A total of 131 miRNAs were identified in all groups. Specifically, 41, 115, 63, and 24 miRNAs were detected in the T12, T24, T36, and T48 groups, respectively. Among these, 15 miRNAs were common to the all groups, while 3, 57, 10, and 3 miRNAs were unique to the T12, T24, T36, and T48 groups, respectively. Functional analyses of the target genes for both common and specific miRNAs indicated that numerous target genes were involved in signaling pathways and cancer-related processes. CONCLUSION It suggested that exosomal miRNAs might be critical in intercellular communication and in dynamically remodeling the tumor microenvironment. These insights could enhance our understanding of the role of exosomal miRNAs in cancer biology and inform the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhaoxia Wang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
- First Hospital of Shanxi Medical University, 85 South Jiefang Road, Taiyuan, Shanxi, 030001, P.R. China.
| | - Yanan Huang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Simin He
- Department of Health Statistics and Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Ying Zhou
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Le Zhao
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Fuyuan Wang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
26
|
Tao X, Zhai SN, Liu CX, Huang Y, Wei J, Guo YL, Liu XQ, Li X, Yang L, Chen LL. Degradation of circular RNA by the ribonuclease DIS3. Mol Cell 2025:S1097-2765(25)00046-2. [PMID: 39965568 DOI: 10.1016/j.molcel.2025.01.012] [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: 03/01/2024] [Revised: 07/30/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Features of circular RNAs (circRNAs) produced by back-splicing of eukaryotic exon(s) make them resistant to degradation by linear RNA decay machineries. Thus, a general circRNA degradation pathway under normal conditions has remained largely elusive. Here, we report that the endonucleolytic enzyme DIS3 is responsible for the degradation of circRNAs. Depletion of DIS3 leads to the upregulation of more than 60% of circRNAs with little effect on their linear cognates. Such DIS3-mediated circRNA degradation is conserved, occurs in the cytoplasm, and relies on DIS3's endonucleolytic activity but is independent of the RNA exosome complex. Sequence enrichment analyses suggest that DIS3 prefers to degrade circRNAs containing U-rich motifs. Correspondingly, synthesized RNA circles with or without U-rich motifs exhibit decreased or increased stabilities, respectively. Together, these findings suggest a general regulation of circRNA turnover by DIS3.
Collapse
Affiliation(s)
- Xiao Tao
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Si-Nan Zhai
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China; Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chu-Xiao Liu
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Youkui Huang
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Wei
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi-Lin Guo
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Qi Liu
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Li
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Li Yang
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Ling-Ling Chen
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; New Cornerstone Science Laboratory, Shenzhen, China; School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| |
Collapse
|
27
|
Malaguarnera M, Cauli O, Cabrera-Pastor A. Obesity and Adipose-Derived Extracellular Vesicles: Implications for Metabolic Regulation and Disease. Biomolecules 2025; 15:231. [PMID: 40001534 PMCID: PMC11853251 DOI: 10.3390/biom15020231] [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: 12/29/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity, a global epidemic, is a major risk factor for chronic diseases such as type 2 diabetes, cardiovascular disorders, and metabolic syndrome. Adipose tissue, once viewed as a passive fat storage site, is now recognized as an active endocrine organ involved in metabolic regulation and inflammation. In obesity, adipose tissue dysfunction disrupts metabolic balance, leading to insulin resistance and increased production of adipose-derived extracellular vesicles (AdEVs). These vesicles play a key role in intercellular communication and contribute to metabolic dysregulation, affecting organs such as the heart, liver, and brain. AdEVs carry bioactive molecules, including microRNAs, which influence inflammation, insulin sensitivity, and tissue remodeling. In the cardiovascular system, AdEVs can promote atherosclerosis and vascular dysfunction, while those derived from brown adipose tissue offer cardioprotective effects. In type 2 diabetes, AdEVs exacerbate insulin resistance and contribute to complications such as diabetic cardiomyopathy and cognitive decline. Additionally, AdEVs are implicated in metabolic liver diseases, including fatty liver disease, by transferring inflammatory molecules and lipotoxic microRNAs to hepatocytes. These findings highlight the role of AdEVs in obesity-related metabolic disorders and their promise as therapeutic targets for related diseases.
Collapse
Affiliation(s)
- Michele Malaguarnera
- Psychobiology Department, University of Valencia, 46010 Valencia, Spain;
- Nursing Department, University of Valencia, 46010 Valencia, Spain
| | - Omar Cauli
- Nursing Department, University of Valencia, 46010 Valencia, Spain
- Frailty Research Organized Group (FROG), University of Valencia, 46010 Valencia, Spain
| | - Andrea Cabrera-Pastor
- Pharmacology Department, University of Valencia, 46010 Valencia, Spain;
- Fundación de Investigación del Hospital Clínico Universitario de Valencia (INCLIVA), 46010 Valencia, Spain
| |
Collapse
|
28
|
Li T, Liu Y, Duan T, Guo C, Liu B, Fu X, Wang L, Wang X, Dong X, Wang C, Lu Y, Wang Y, Shi L, Tian H, Yang X. Nondigestible stachyose binds membranous HSP90β on small intestinal epithelium to regulate the exosomal miRNAs: A new function and mechanism. Cell Metab 2025; 37:345-360.e6. [PMID: 39561765 DOI: 10.1016/j.cmet.2024.10.012] [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: 11/18/2022] [Revised: 07/16/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024]
Abstract
Oligosaccharides are conventionally recognized as "passersby" in the small intestine. However, our research has reframed this understanding by uncovering a new function of oligosaccharide stachyose, which binds hydrophobic residues of membranous HSP90β on small intestinal epithelial cells, thus reprograming the exosomal miRNA profile. CRISPR-Cas9-mediated HSP90β knockout abolished the accumulation of stachyose on cell membrane and its regulatory effects on these miRNAs. Notably, stachyose's regulation on these miRNAs is independent of its prebiotic role, as evidenced by the observation of stachyose-altered fecal miRNAs in pseudo-germ-free mice. These stachyose-altered miRNAs further shaped colonic microbiome, especially harboring Lactobacillus in mice. Thereinto, miR-30a-5p that was downregulated (Log2FC < -2) in both mice and human feces following stachyose treatment could specifically suppress the growth of Lactobacillus reuteri. These findings build a new regulatory axis of stachyose-intestinal miRNAs-gut microbiota and unveil a previously unknown mechanism underlying the direct "talk" of oligosaccharides to intestine epithelium via membranous HSP90β.
Collapse
Affiliation(s)
- Ting Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| | - Yueyue Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Tianchi Duan
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Xiuqiong Fu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Lu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xiaoyuan Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xinyue Dong
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Chennan Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yalong Lu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Lin Shi
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
29
|
Hossain M, Liu Y. Extracellular Vesicles and Glaucoma: Opportunities and Challenges. Curr Eye Res 2025:1-10. [PMID: 39898581 DOI: 10.1080/02713683.2025.2459888] [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: 10/29/2024] [Revised: 01/06/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Glaucoma is one of the leading causes of irreversible blindness, characterized by progressive visual field loss. Several risk factors are associated with developing the disease. However, the exact mechanisms or pathological pathways involved are still unknown. There is an urgent need to find the mechanisms and biomarkers for early detection and therapy to halt progression or cure the disease. Extracellular vesicles (EVs), specifically exosomes, have emerged as a crucial player in all aspects of glaucoma, including pathogenesis to therapeutic application with their cell-cell communication properties. METHODS We performed a literature search on PubMed, Google Scholar, and Web of Science using different keywords. Next, we reviewed the literature with studies focusing on the role of EVs as a causative factor in disease progression, biomarker discovery based on their contents, and protection from glaucoma. RESULTS Studies summarized here provide reports of differential EV miRNA and protein expression alterations when communicating with aqueous humor drainage tissues. We described how EV contents are involved in various pathways, including extracellular matrix remodeling and miRNA-mediated oxidative stress transmission between outflow tissues, thereby contributing to glaucoma. Extracellular vesicles, mainly derived from mesenchymal stem cells protecting the optic nerve from degeneration, have also been discussed as potential therapies for glaucoma. CONCLUSIONS Overall, this review provides a comprehensive discussion of the role of extracellular vesicles in glaucoma. We identified the challenges in finding major signaling molecules of glaucoma etiology. Lastly, we highlighted future directions to improve the treatment of glaucoma by extracellular vesicles.
Collapse
Affiliation(s)
- Mofazzal Hossain
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
30
|
Chen X, Liu S, Wang H, Liu Y, Xiao Y, Li K, Ni F, Wu W, Lin H, Qing X, Pu F, Wang B, Shao Z, Peng Y. Extracellular vesicles deliver thioredoxin to rescue stem cells from senescence and intervertebral disc degeneration via a feed-forward circuit of the NRF2/AP-1 composite pathway. Acta Pharm Sin B 2025; 15:1007-1022. [PMID: 40177564 PMCID: PMC11959923 DOI: 10.1016/j.apsb.2024.12.013] [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: 04/26/2024] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 04/05/2025] Open
Abstract
Intervertebral disc degeneration (IDD) is largely attributed to impaired endogenous repair. Nucleus pulposus-derived stem cells (NPSCs) senescence leads to endogenous repair failure. Small extracellular vesicles/exosomes derived from mesenchymal stem cells (mExo) have shown great therapeutic potential in IDD, while whether mExo could alleviate NPSCs senescence and its mechanisms remained unknown. We established a compression-induced NPSCs senescence model and rat IDD models to evaluate the therapeutic efficiency of mExo and investigate the mechanisms. We found that mExo significantly alleviated NPSCs senescence and promoted disc regeneration while knocking down thioredoxin (TXN) impaired the protective effects of mExo. TXN was bound to various endosomal sorting complex required for transport (ESCRT) proteins. Autocrine motility factor receptor (AMFR) mediated TXN K63 ubiquitination to promote the binding of TXN on ESCRT proteins and sorting of TXN into mExo. Knocking down exosomal TXN inhibited the transcriptional activity of nuclear factor erythroid 2-related factor 2 (NRF2) and activator protein 1 (AP-1). NRF2 and AP-1 inhibition reduced endogenous TXN production that was promoted by exosomal TXN. Inhibition of NRF2 in vivo diminished the anti-senescence and regenerative effects of mExo. Conclusively, AMFR-mediated TXN ubiquitination promoted the sorting of TXN into mExo, allowing exosomal TXN to promote endogenous TXN production in NPSCs via TXN/NRF2/AP-1 feed-forward circuit to alleviate NPSCs senescence and disc degeneration.
Collapse
Affiliation(s)
- Xuanzuo Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huiwen Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiran Liu
- The First School of Clinical Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Xiao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kanglu Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feifei Ni
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feifei Pu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
31
|
Liu XM, Halushka MK. Beyond the Bubble: A Debate on microRNA Sorting Into Extracellular Vesicles. J Transl Med 2025; 105:102206. [PMID: 39647608 PMCID: PMC11842217 DOI: 10.1016/j.labinv.2024.102206] [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: 10/01/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024] Open
Abstract
Over the past decade, a scientific field has been developed demonstrating microRNAs (miRNAs) to be actively sorted into extracellular vesicles via specific nucleotide motifs that interact with discrete RNA-binding proteins. These miRNAs are proposed to be transported into recipient cells in which they can regulate specific cellular pathways. This mechanism could have enormous potential in explaining how cells signal and regulate other cells nearby or at a distance. Tens of studies have built this theme of a regulated transport of miRNAs. However, some concerns exist about this field. Taken together, there are concerns of a lack of a consistent motif, RNA-binding protein, or preferential miRNA involved in this process. In this study, we provide an expert and extensive analysis of the field that makes the cases for and against an active sorting mechanism. We provide potential explanations on why there is a lack of agreement. Most importantly, we provide ideas on how to move this field forward with more rigor and reproducibility. It is hoped that by engaging in a scientific debate of the pros and cons of this field, more rigorous experiments can be performed to conclusively demonstrate this biological activity.
Collapse
Affiliation(s)
- Xiao-Man Liu
- The Stanley Center for Psychiatric Research, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Marc K Halushka
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
32
|
Ning W, Yang J, Ni R, Yin Q, Zhang M, Zhang F, Yang Y, Zhang Y, Cao M, Jin L, Pan Y. Hypoxia induced cellular and exosomal RPPH1 promotes breast cancer angiogenesis and metastasis through stabilizing the IGF2BP2/FGFR2 axis. Oncogene 2025; 44:147-164. [PMID: 39496940 DOI: 10.1038/s41388-024-03213-y] [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: 08/20/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024]
Abstract
Metastasis is the major cause of breast cancer mortality, with angiogenesis and tumor-released exosomes playing key roles. However, the communication between breast cancer cells and endothelial cells and its role in tumor metastasis remains unclear. Here, we characterize a long noncoding RNA, RPPH1, which is upregulated in breast cancer tissues and positively associated with poor prognosis. Hypoxia microenvironment upregulates the expression of RPPH1 in breast cancer cells, and promotes its packaging into exosomes through hnRNPA1, leading to the maintenance of stemness and aggressive traits in cancer cells and angiogenesis in endothelial cells. The function of cellular and exosomal RPPH1 was confirmed in the MMTV-PyMT mouse model, in which ASO-RPPH1 therapy effectively inhibited tumor progression and metastasis. Mechanistically, RPPH1 protects IGF2BP2 from ubiquitination-induced degradation, stabilizes N6-methyladenosine (m6A)-modified FGFR2 mRNA, and activates the PI3K/AKT pathway. Our research unveils the role of RPPH1 in breast cancer metastasis and highlights its potential as a therapeutic target.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Female
- Animals
- Exosomes/metabolism
- Exosomes/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Mice
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Neoplasm Metastasis
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Signal Transduction
- Tumor Microenvironment/genetics
- Angiogenesis
Collapse
Affiliation(s)
- Wentao Ning
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jingyan Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ruiqi Ni
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qianqian Yin
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Manqi Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Meng Cao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
33
|
Qiao SA, Roth R. Messenger and message: Uncovering the roles, rhythm and regulation of extracellular vesicles in plant biotic interactions. CURRENT OPINION IN PLANT BIOLOGY 2025; 83:102672. [PMID: 39733495 DOI: 10.1016/j.pbi.2024.102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/31/2024]
Abstract
Extracellular vesicles (EVs) are membrane-delimited nanoparticles found in every kingdom of life and are known to mediate cell-cell communication in animal systems through the trafficking of proteins and nucleic acids. Research into plant and microbial EVs suggests that these have similar transport capacity, and moreover are able to mediate signalling not only within an organism but also between organisms, acting between plants and their microbial partners in cross-kingdom signalling. Here, we review recent research exploring the roles of these EVs, both plant and microbial, highlighting emerging trends of functional conservation between species and across kingdoms, complemented by the heterogeneity of EV subpopulations at the organism level that places EVs as powerful regulatory mechanisms in plant biotic interactions.
Collapse
Affiliation(s)
- Serena Agnes Qiao
- Department of Biology, South Parks Road, University of Oxford, Oxford, OX1 3RB, United Kingdom
| | - Ronelle Roth
- Department of Biology, South Parks Road, University of Oxford, Oxford, OX1 3RB, United Kingdom.
| |
Collapse
|
34
|
Rohm TV, Cunha E Rocha K, Olefsky JM. Metabolic Messengers: small extracellular vesicles. Nat Metab 2025; 7:253-262. [PMID: 39920357 DOI: 10.1038/s42255-024-01214-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025]
Abstract
Small extracellular vesicles (sEVs) are signalling molecules and biomarkers of cell status that govern a complex intraorgan and interorgan communication system through their cargo. Initially recognized as a waste disposal mechanism, they have emerged as important metabolic regulators. They transfer biological signals to recipient cells through their cargo content, and microRNAs (miRNAs) often mediate their metabolic effects. This review provides a concise overview of sEVs, specifically in the context of obesity-associated chronic inflammation and related metabolic disorders, describing their role as metabolic messengers, identifying their key sites of action and elucidating their mechanisms. We highlight studies that have shaped our understanding of sEV metabolism, address critical questions for future exploration, discuss the use of miRNAs as disease biomarkers and provide insights into the therapeutic potential of sEVs or specific miRNAs for treating metabolic diseases and related disorders in the future.
Collapse
Affiliation(s)
- Theresa V Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Sadeghi M, Tavakol Afshari J, Fadaee A, Dashti M, Kheradmand F, Dehnavi S, Mohammadi M. Exosomal miRNAs involvement in pathogenesis, diagnosis, and treatment of rheumatoid arthritis. Heliyon 2025; 11:e41983. [PMID: 39897907 PMCID: PMC11786886 DOI: 10.1016/j.heliyon.2025.e41983] [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: 03/21/2024] [Revised: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune arthropathy worldwide. The initiation, and progression of RA involves multiple cellular and molecular pathways, and biological interactions. Micro RNAs (miRNAs) are characterized as a class of small non-coding RNAs that influence gene expression at the post-transcriptional level. Exosomes are biological nano-vesicles that are secreted by different types of cells. They facilitate communication and signalling between cells by transferring a variety of biological substances, such as proteins, lipids, and nucleic acids like mRNA and miRNA. Exosomal miRNAs were shown to be involved in normal and pathological conditions. In RA, deregulated exosomal miRNA expression was observed to be involved in the intercellular communication between synovial cells, and inflammatory or regulatory immune cells. Furthermore, circulating exosomal miRNAs were introduced as available diagnostic and prognostic biomarkers for RA pathology. The current review categorized and summarized dysregulated pathologically involved and circulating exosomal miRNAs in the context of RA. It highlighted present situation and future perspective of using exosomal miRNAs as biomarkers and a specific gene therapy approach for RA treatment.
Collapse
Affiliation(s)
- Mahvash Sadeghi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Afsane Fadaee
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Dashti
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Kheradmand
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Dehnavi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
Huang C, Xiao Y, Qing L, Tang J, Wu P. Exosomal non-coding RNAs in the regulation of bone metabolism homeostasis: Molecular mechanism and therapeutic potential. Heliyon 2025; 11:e41632. [PMID: 39911437 PMCID: PMC11795052 DOI: 10.1016/j.heliyon.2025.e41632] [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: 07/04/2024] [Revised: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Bone metabolism is a dynamic balance between bone formation and absorption regulated by osteoblasts/osteoclasts. Bone metabolic disorders can lead to metabolic bone disease. Osteoporosis (OP), osteoarthritis (OA) and femoral head necrosis (ONFH) are common metabolic bone diseases. At present, the treatment of metabolic bone disease is still mainly to relieve pain and improve joint function. However, surgical treatment does not apply to the vast majority of high-risk groups, including postmenopausal women, patients with diabetes, cirrhosis, etc. Exosomes (Exos) are nanoscale membrane vesicles that are released by almost all cells. Exos are rich in a variety of bioactive substances, such as non-coding RNAs, nucleic acids, proteins and lipids. In view of the structure of Exos, it can protect the biologically active molecules can be smoothly delivered to the target cells and involved in the regulation of cell function. In this review, we focus on the regulation mechanism and function of bone homeostasis mediated by exosomal ncRNAs (Exos-ncRNAs), including macrophage polarization, autophagy, angiogenesis, signal transduction and competing endogenous RNA (ceRNA). We summarized the therapeutic strategies and potential drugs of Exos-ncRNAs in metabolic bone disease. Moreover, we discussed the shortcomings and potential research directions of Exos as carrier to deliver ncRNAs to play a role.
Collapse
Affiliation(s)
- Chengxiong Huang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yu Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Liming Qing
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Juyu Tang
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Panfeng Wu
- Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| |
Collapse
|
37
|
Orefice NS, Petrillo G, Pignataro C, Mascolo M, De Luca G, Verde S, Pentimalli F, Condorelli G, Quintavalle C. Extracellular vesicles and microRNAs in cancer progression. Adv Clin Chem 2025; 125:23-54. [PMID: 39988407 DOI: 10.1016/bs.acc.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Extracellular vesicles (EVs) have emerged as critical mediators of intercellular communication in cancer. These membranous structures, secreted by normal and cancerous cells, carry a cargo of bioactive molecules including microRNAs (miRNAs) that modulate various cellular processes. miRNAs are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation and have been implicated in cancer initiation, progression, and metastasis. In cancer, tumor-derived EVs transport specific miRNAs to recipient cells, modulating tumorigenesis, growth, angiogenesis, and metastasis. Dysregulation of miRNA expression profiles within EVs contributes to the acquisition of cancer hallmarks that include increased proliferation, survival, and migration. EV miRNAs influence the tumor microenvironment, promoting immune evasion, remodeling the extracellular matrix, and establishing pre-metastatic niches. Understanding the complex interplay between EVs, miRNAs, and cancer holds significant promise for developing novel diagnostic and therapeutic strategies. This chapter provides insights into the role of EV-mediated miRNA signaling in cancer pathogenesis, highlighting its potential as a biomarker for cancer detection, prognosis, and treatment response assessment.
Collapse
Affiliation(s)
- Nicola S Orefice
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Gianluca Petrillo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Martina Mascolo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Giada De Luca
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| | - Sara Verde
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; Aka biotech S.r.l., Napoli, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe DeGennaro", Bari, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy; Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy.
| | - Cristina Quintavalle
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| |
Collapse
|
38
|
Shen H, Zhang M, Liu D, Liang X, Chang Y, Hu X, Gao W. Solanum lycopersicum derived exosome-like nanovesicles alleviate restenosis after vascular injury through the Keap1/Nrf2 pathway. Food Funct 2025; 16:539-553. [PMID: 39688297 DOI: 10.1039/d4fo03993a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Despite the significant alleviation of clinical cardiovascular diseases through appropriate interventional treatments, the recurrence of vascular restenosis necessitating reoperation remains a substantial challenge impacting patient prognosis. Plant-derived exosome-like nanovesicles (PELNs) are integral to interspecies cellular communication, with their functions and potential applications garnering significant attention from the research community. This study extracted Solanum lycopersicum-derived exosome-like nanovesicles (SL-ELNs) and demonstrated their inhibition of PDGF-BB-induced proliferation, migration, and phenotypic transformation of vascular smooth muscle cells (VSMCs). Mechanistically, miRNA164a/b-5p within the SL-ELNs reduced the expression of Keap1 mRNA, thereby increasing nuclear translocation of Nrf2 and enhancing the expression of antioxidant genes to alleviate oxidative stress. In a mouse carotid artery injury model, it was further confirmed that miRNA164a/b-5p within the SL-ELNs could inhibit neointimal hyperplasia. These results suggest that SL-ELNs inhibit VSMCs proliferation, migration, and phenotypic transformation, and they might be potential therapeutic agents for the prevention or treatment of restenosis.
Collapse
Affiliation(s)
- Hechen Shen
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Meng Zhang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Dachang Liu
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
- School of Medicine, Nankai University, Tianjin 300170, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin 300170, China
| | - Xiaoyu Liang
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Yun Chang
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Xiaomin Hu
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
- Medical College, Tianjin University, Tianjin, 300072, China
- School of Medicine, Nankai University, Tianjin 300170, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin 300170, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin ECMO Treatment and Training Base, Tianjin 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
- Medical College, Tianjin University, Tianjin, 300072, China
- School of Medicine, Nankai University, Tianjin 300170, China
- Nankai University Affiliated Third Center Hospital, Nankai University, Tianjin 300170, China
| |
Collapse
|
39
|
Hu H, Wang X, Yu H, Wang Z. Extracellular vesicular microRNAs and cardiac hypertrophy. Front Endocrinol (Lausanne) 2025; 15:1444940. [PMID: 39850481 PMCID: PMC11753959 DOI: 10.3389/fendo.2024.1444940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Cardiac hypertrophy is an adaptive response to pressure or volume overload such as hypertension and ischemic heart diseases. Sustained cardiac hypertrophy eventually leads to heart failure. The pathophysiological alterations of hypertrophy are complex, involving both cellular and molecular systems. Understanding the molecular events that inhibit or repress cardiac hypertrophy may help identify novel therapeutic strategies. Increasing evidence has indicated that extracellular vesicle (EV)-derived microRNAs (miRNAs) play a significant role in the development and progression of cardiac hypertrophy. In this review, we briefly review recent advancements in EV research, especially on biogenesis, cargoes and its role in cardiac hypertrophy. We then describe the latest findings regarding EV-derived miRNAs, highlighting their functions and regulatory mechanisms in cardiac hypertrophy. Finally, the potential role of EV-derived miRNAs as targets in the diagnosis and treatment of cardiac hypertrophy will be discussed.
Collapse
Affiliation(s)
- Hai Hu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xiulian Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
40
|
Lou S, Hu W, Wei P, He D, Fu P, Ding K, Chen Z, Dong Z, Zheng J, Wang K. Artificial Nanovesicles Derived from Cells: A Promising Alternative to Extracellular Vesicles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22-41. [PMID: 39692623 DOI: 10.1021/acsami.4c12567] [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: 12/19/2024]
Abstract
As naturally secreted vesicles by cells, extracellular vesicles (EVs) play essential roles in modulating cell-cell communication and have significant potential in tissue regeneration, immune regulation, and drug delivery. However, the low yield and uncontrollable heterogeneity of EVs have been obstacles to their widespread translation into clinical practice. Recently, it has been discovered that artificial nanovesicles (NVs) produced by cell processing can inherit the components and functions of the parent cells and possess similar structures and functions to EVs, with significantly higher yields and more flexible functionalization, making them a powerful complement to natural EVs. This review focuses on recent advances in the research of artificial NVs as replacements for natural EVs. We provide an overview comparing natural EVs and artificial NVs and summarize the top-down preparation strategies of NVs. The applications of NVs prepared from stem cells, differentiated cells, and engineered cells are presented, as well as the latest advances in NV engineering. Finally, the main challenges of artificial NVs are discussed.
Collapse
Affiliation(s)
- Saiyun Lou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Wei Hu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Pengyao Wei
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Dongdong He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kejian Ding
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhenyi Chen
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Jianping Zheng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
Clarke A, Høye E, Hembrom A, Paynter V, Vinther J, Wyrożemski Ł, Biryukova I, Formaggioni A, Ovchinnikov V, Herlyn H, Pierce A, Wu C, Aslanzadeh M, Cheneby J, Martinez P, Friedländer M, Hovig E, Hackenberg M, Umu SU, Johansen M, Peterson K, Fromm B. MirGeneDB 3.0: improved taxonomic sampling, uniform nomenclature of novel conserved microRNA families and updated covariance models. Nucleic Acids Res 2025; 53:D116-D128. [PMID: 39673268 PMCID: PMC11701709 DOI: 10.1093/nar/gkae1094] [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: 09/24/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 12/16/2024] Open
Abstract
We present a major update of MirGeneDB (3.0), the manually curated animal microRNA gene database. Beyond moving to a new server and the creation of a computational mirror, we have expanded the database with the addition of 33 invertebrate species, including representatives of 5 previously unsampled phyla, and 6 mammal species. MirGeneDB now contains entries for 21 822 microRNA genes (5160 of these from the new species) belonging to 1743 microRNA families. The inclusion of these new species allowed us to refine both the evolutionary node of appearance of a number of microRNA genes/families, as well as MirGeneDB's phylogenetically informed nomenclature system. Updated covariance models of all microRNA families, along with all smallRNA read data are now downloadable. These enhanced annotations will allow researchers to analyze microRNA properties such as secondary structure and features of their biogenesis within a robust phylogenetic context and without the database plagued with numerous false positives and false negatives. In light of these improvements, MirGeneDB 3.0 will assume the responsibility for naming conserved novel metazoan microRNAs. MirGeneDB is part of RNAcentral and Elixir Norway and is publicly and freely available at mirgenedb.org.
Collapse
Affiliation(s)
- Alexander W Clarke
- Department of Biological Sciences, 321 Life Sciences Center, Dartmouth College, 78 College St., Hanover, NH 03755, USA
- The Arctic University Museum of Norway, UiT - The Arctic University of Norway, Lars Thørings veg 10, 9006 Tromsø, Norway
| | - Eirik Høye
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Anju Angelina Hembrom
- The Arctic University Museum of Norway, UiT - The Arctic University of Norway, Lars Thørings veg 10, 9006 Tromsø, Norway
| | - Vanessa Molin Paynter
- The Arctic University Museum of Norway, UiT - The Arctic University of Norway, Lars Thørings veg 10, 9006 Tromsø, Norway
| | - Jakob Vinther
- School of Earth Sciences & School of Biological Sciences, University of Bristol, Life Sciences Building, 24 Tyndall Avenue, BS5 8EH, Bristol, UK
| | - Łukasz Wyrożemski
- The Arctic University Museum of Norway, UiT - The Arctic University of Norway, Lars Thørings veg 10, 9006 Tromsø, Norway
| | - Inna Biryukova
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691,Stockholm, Sweden
| | - Alessandro Formaggioni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Vladimir Ovchinnikov
- Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Holger Herlyn
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Ackermannweg 4, 55128 Mainz, Germany
| | - Alexandra Pierce
- Department of Biological Sciences, 321 Life Sciences Center, Dartmouth College, 78 College St., Hanover, NH 03755, USA
| | - Charles Wu
- Valley Stream North High School, 750 Herman Ave, Franklin Square, NY 11010, USA
| | - Morteza Aslanzadeh
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691,Stockholm, Sweden
| | - Jeanne Cheneby
- Center for Bioinformatics, Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| | - Pedro Martinez
- Department de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Avinguda Diagonal, 643; 08028-Barcelona, Spain
- ICREA (Institut Català de Recerca i Estudis Avancats), Passeig Lluis Companys 23; 08010-Barcelona, Spain
| | - Marc R Friedländer
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691,Stockholm, Sweden
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Ullernchausseen 70, 0379 Oslo, Norway
| | - Michael Hackenberg
- Department of Genetics, Faculty of Sciences, University of Granada, Avenida de la Fuente Nueva S/N, C.P. 18071 Granada, Spain
- Bioinformatics Laboratory, Biotechnology Institute & Biomedical Research Centre (CIBM), Avenida del Conocimiento 19 Granada, 18100, Spain
| | - Sinan Uğur Umu
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Morten Johansen
- Center for Bioinformatics, Department of Informatics, University of Oslo, Gaustadalléen 23B, 0373 Oslo, Norway
| | - Kevin J Peterson
- Department of Biological Sciences, 321 Life Sciences Center, Dartmouth College, 78 College St., Hanover, NH 03755, USA
| | - Bastian Fromm
- The Arctic University Museum of Norway, UiT - The Arctic University of Norway, Lars Thørings veg 10, 9006 Tromsø, Norway
| |
Collapse
|
42
|
Liu ZZ, Ji FH, Piao Y. Non-coding RNAs participate in interactions between senescence and gastrointestinal cancers. Front Genet 2025; 15:1461404. [PMID: 39831201 PMCID: PMC11739115 DOI: 10.3389/fgene.2024.1461404] [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: 07/08/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Relationships between cellular senescence and gastrointestinal cancers have gained prominence in recent years. The currently accepted theory suggests that cellular senescence and cancer occurrence exhibit "double-edged sword" effects. Cellular senescence is related to cancer via four "meta-hallmarks" i.e., genomic instability, epigenetic alterations, chronic inflammation, and dysbiosis, along with two "antagonistic hallmarks" i.e., telomere attrition and stem cell exhaustion. These relationships are characterized by both agonistic and antagonistic elements, but the existence of an intricate dynamic balance remains unknown. Non-coding RNAs (ncRNAs) have vital roles in post-transcriptional regulation, but how they participate in agonistic and antagonistic relationships between cellular senescence and gastrointestinal cancers remains to be fully investigated. In this article, we systematically review how ncRNAs (including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circularRNAs (circRNAs)) participate in interactions between cellular senescence and gastrointestinal cancers. Our aim is to elucidate a triangular relationship between "ncRNAs-senescence-gastrointestinal cancers" which considered these three elements as an equal important standing. We are keen to identify prognostic or therapeutic targets for gastrointestinal cancers from, i.e., aging-related ncRNAs, or discover novel strategies to treat and manage in the elderly. We seek to clarify complex relationships where ncRNAs participate in "senescence-gastrointestinal cancers" interactions.
Collapse
Affiliation(s)
| | | | - Ying Piao
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
43
|
Li YZ, Tian Y, Yang C, Liu YF, Qu SL, Huang L, Zhang C. Adipose tissue macrophages-derived exosomal MiR-500a-5p under high glucose promotes adipocytes inflammation by suppressing Nrf2 expression. Int J Biochem Cell Biol 2025; 178:106713. [PMID: 39617207 DOI: 10.1016/j.biocel.2024.106713] [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: 10/11/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Type 2 diabetes (T2DM) is a chronic metabolic disorder characterized by insulin resistance and chronic inflammation. Adipose tissue macrophages (ATMs), central players in mediating pro-inflammatory responses within adipose tissue, have been shown to influence insulin sensitivity through exosome secretion. While the role of macrophages-derived exosomal miRNA has been studied in various diseases, their pathogenic roles in T2DM, particularly ATMs-derived exosomal miRNA in adipose tissue inflammation, remain underexplored. OBJECTIVES This study focuses specifically on T2DM, investigating the role of ATM-derived exosomal miRNAs in adipose tissue inflammation, a critical factor in the pathogenesis of T2DM. METHODS ATM were isolated from visceral adipose tissues in patients with or without diabetes. Differentially expressed miRNAs in ATM-derived exosomes were predicted by high-throughput RNA sequencing. The RAW264.7 macrophages and 3T3-L1 preadipocytes was selected as a model system. Quantitative RT-PCR was used to assess miR-500a-5p expression. The direct binding of miR-500a-5p to Nrf2 mRNA 3' UTR was verified by dual luciferase assay. RESULTS MiR-500a-5p was also enriched in the exosomes of high-glucose-treated macrophages. Furthermore, these exosomes induced high expression of miR-500a-5p and activation of the NLRP3 inflammasome in adipocytes when co-cultured with them. Additionally, the reduction of miR-500a-5p expression in macrophages by using a miR-500a-5p inhibitor ameliorated the pro-inflammatory properties of the exosomes, and co-culturing these exosomes with adipocytes resulted in decreased expression of NLRP3 inflammasome-associated proteins in adipocytes. In contrast, induction of miR-500a-5p expression led to the opposite results. Moreover, the dual-luciferase assay confirmed that miR-500a-5p directly targeted the 3' UTR of Nrf2 mRNA. Unlike miR-500a-5p, Nrf2 exhibited an anti-inflammatory response. CONCLUSION The results indicate that ATM-derived exosomal miR-500a-5p promotes NLRP3 inflammasome activation and adipose tissue inflammation through down-regulation of Nrf2 in adipocytes.
Collapse
Affiliation(s)
- Yong-Zhen Li
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Department of Pathology, The First People's Hospital of Zigong, Zigong 643099, PR China
| | - Yuan Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Chen Yang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; Department of Pathology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441021, PR China
| | - Yi-Fan Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
44
|
Padilla JCA, Barutcu S, Deschamps-Francoeur G, Lécuyer E. Exploring Extracellular Vesicle Transcriptomic Diversity Through Long-Read Nanopore Sequencing. Methods Mol Biol 2025; 2880:227-241. [PMID: 39900762 DOI: 10.1007/978-1-0716-4276-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Nanopore long-read RNA sequencing is reshaping extracellular vesicle (EV) research by providing the capacity to analyze full-length RNA molecules. EVs are crucial for intercellular communication, carrying a diverse range of RNA cargo that can regulate recipient cell behavior. However, traditional short-read sequencing methods involve transcript fragmentation, limiting our understanding of the EV transcriptomic landscape. Furthermore, it has been generally assumed that EV RNAs are likely to be fragmentation products of cellular RNAs, and the extent to which full length RNAs are present within EVs remains to be clarified. Recent advancements in sequencing technology, particularly long-read sequencing by Oxford Nanopore Technologies (ONT), offer a solution to this limitation. Hence, long-read sequencing allows for the analysis of full-length EV RNA molecules, providing deeper insights into their integrity and isoform diversity. Here, we present a comprehensive protocol for EV RNA purification, cDNA library preparation, and sequencing using ONT's MinION platform.
Collapse
Affiliation(s)
- Juan-Carlos A Padilla
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Seda Barutcu
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada
| | | | - Eric Lécuyer
- Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada.
- Département de Biochimie et de Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
45
|
Yim KHW, Al Hrout A, Chahwan R. Intercellular Epigenomic Signalling via Extracellular Vesicles During B Cell Maturation. J Extracell Vesicles 2025; 14:e70040. [PMID: 39868437 PMCID: PMC11770373 DOI: 10.1002/jev2.70040] [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: 03/20/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/28/2025] Open
Abstract
B cell maturation is crucial for effective adaptive immunity. It requires a complex signalling network to mediate antibody diversification through mutagenesis. B cells also rely on queues from other cells within the germinal centre. Recently, a novel class of intercellular signals mediated by extracellular vesicles (EVs) has emerged. Studies have shown that B cell EV-mediated signalling is involved in immune response regulation and tumorigenesis. However, the mechanistic role of B cell EVs is not yet established. We herein study the biological properties and physiological function of B cell EVs during B cell maturation. We use emerging technologies to profile B cell EV surface marker signatures at the single particle level, molecular cargo and physiological roles in B cell maturation. EV ncRNA cargo, characterised by RNA-seq, identified an EV-mediated novel non-coding RNA (ncRNA) regulatory network for B cell maturation. We show that a previously uncharacterised micro-RNA (miR-5099) in combination with a set of long ncRNA are carried within B cell EVs and could contribute to antibody diversification. The physiological role of EVs in B cell maturation is investigated using EV blockade assays and complementation studies using diverse EV sources further confirmed the physiological role and mode of action of EVs in B cell maturation.
Collapse
Affiliation(s)
- Kevin Ho Wai Yim
- Institute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
- EVIIVE AGZurichSwitzerland
| | - Ala'a Al Hrout
- Institute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
| | - Richard Chahwan
- Institute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
- EVIIVE AGZurichSwitzerland
| |
Collapse
|
46
|
Venu E, Ramya A, Babu PL, Srinivas B, Kumar S, Reddy NK, Babu YM, Majumdar A, Manik S. Exogenous dsRNA-Mediated RNAi: Mechanisms, Applications, Delivery Methods and Challenges in the Induction of Viral Disease Resistance in Plants. Viruses 2024; 17:49. [PMID: 39861836 PMCID: PMC11769437 DOI: 10.3390/v17010049] [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: 11/30/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The increasing challenges posed by plant viral diseases demand innovative and sustainable management strategies to minimize agricultural losses. Exogenous double-stranded RNA (dsRNA)-mediated RNA interference (RNAi) represents a transformative approach to combat plant viral pathogens without the need for genetic transformation. This review explores the mechanisms underlying dsRNA-induced RNAi, highlighting its ability to silence specific viral genes through small interfering RNAs (siRNAs). Key advancements in dsRNA production, including cost-effective microbial synthesis and in vitro methods, are examined alongside delivery techniques such as spray-induced gene silencing (SIGS) and nanocarrier-based systems. Strategies for enhancing dsRNA stability, including the use of nanomaterials like layered double hydroxide nanosheets and carbon dots, are discussed to address environmental degradation challenges. Practical applications of this technology against various plant viruses and its potential to ensure food security are emphasized. The review also delves into regulatory considerations, risk assessments, and the challenges associated with off-target effects and pathogen resistance. By evaluating both opportunities and limitations, this review underscores the role of exogenous dsRNA as a sustainable solution for achieving viral disease resistance in plants.
Collapse
Affiliation(s)
- Emmadi Venu
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| | - Akurathi Ramya
- Department of Plant Pathology, Junagadh Agricultural University, Junagadh 362001, India
| | - Pedapudi Lokesh Babu
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| | - Bhukya Srinivas
- Department of Plant Pathology, Professor Jayashankar Telangana State Agricultural University, Rajendranagar, Hyderabad 500030, India;
| | - Sathiyaseelan Kumar
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| | - Namburi Karunakar Reddy
- Department of Plant Pathology, University of Agricultural Sciences, GKVK, Bengaluru 560065, India;
| | - Yeluru Mohan Babu
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| | - Anik Majumdar
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| | - Suryakant Manik
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi 110012, India; (P.L.B.); (S.K.); (Y.M.B.); (A.M.); (S.M.)
| |
Collapse
|
47
|
Agrawal P, Olgun G, Singh A, Gopalan V, Hannenhalli S. Characterizing the pan-cancer role of exosomal miRNAs in metastasis across cancers. Comput Struct Biotechnol J 2024; 27:252-264. [PMID: 39866667 PMCID: PMC11763893 DOI: 10.1016/j.csbj.2024.12.025] [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: 11/13/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/28/2025] Open
Abstract
Exosomal microRNAs (exomiRs) play a critical role in intercellular communication, especially in cancer, where they regulate key cellular processes like proliferation, angiogenesis, and metastasis, highlighting their significance as potential diagnostic and therapeutic targets. Here, we aimed to characterize the role of exomiRs, derived from seven cancer types (four cell lines and three tumors), in influencing the pre-metastatic niche (PMN). In each cancer type we extracted high confidence exomiRs (LogFC >= 2 in exosomes relative to control), their experimentally validated targets, and the enriched pathways among those targets. We then selected the top100 high-confidence targets based on their frequency of appearance in the enriched pathways. We observed significantly higher GC content in exomiRs relative to genomic background. Gene Ontology analysis revealed both general cancer processes, such as wound healing and epithelial cell proliferation, as well as cancer-specific processes, such as "angiogenesis" in the kidney and "ossification" in the lung. ExomiR targets were enriched for cancer-specific tumor suppressor genes and downregulated in PMN formed in lungs compared to normal. Motif analysis showed high inter-cancer similarity among motifs enriched in exomiRs. Our analysis recapitulated exomiRs associated with M2 macrophage differentiation and chemoresistance, such as miR-21 and miR-222-3p, regulating signaling pathways like PTEN/PI3/Akt, NF-kB, etc. Additionally, Cox regression analysis in TCGA indicated that exomiR targets are significantly associated with better overall survival of patients. Lastly, support vector machine model using exomiR targets gene expression classified responders and non-responders to therapy with an AUROC ranging from 0.72 to 0.96, higher than previously reported gene signatures.
Collapse
Affiliation(s)
- Piyush Agrawal
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Gulden Olgun
- Department of Computer Engineering, Hacettepe University, Ankara 06800, Turkey
| | - Arashdeep Singh
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
48
|
Wang L, Liu H, Chen G, Wu Q, Xu S, Zhou Q, Zhao Y, Wang Q, Yan T, Cheng X. Bubble Ticket Trip: Exploring the Mechanism of miRNA Sorting into Exosomes and Maintaining the Stability of Tumor Microenvironment. Int J Nanomedicine 2024; 19:13671-13685. [PMID: 39723172 PMCID: PMC11669276 DOI: 10.2147/ijn.s498599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Exosomes are vesicles ranging from 30 to 100 nanometers in size that show great potential as carriers for therapeutic uses and drug delivery. Enriching a specific set of miRNAs in exosomes emphasizes the existence of particular sorting mechanisms that manage the targeted cargo packaging. The molecular mechanism for miRNA sorting has not been understood. It is crucial to understand the mechanism of exosome encapsulation to develop its therapeutic potential. In this review, we will explore the particular processes through which exosomes naturally encapsulate miRNA, as well as discuss the effect on tumors after encapsulation of miRNAs. We also summarize the effects of targeted drug delivery using genetic engineering and chemical methods to modify exosome-encapsulated miRNA. Finally, gaining insight into how exosome cargo is sorted could be applied in clinical settings for precise drug delivery and to hinder the progression of diseases.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Guohui Chen
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qinglu Wu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Songrui Xu
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qichao Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Yadong Zhao
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Qiaorong Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Ting Yan
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, & Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, People's Republic of China
| |
Collapse
|
49
|
Acosta Montaño P, Olvera Félix E, Castro Flores V, Hernández García A, Cadena-Nava RD, Galindo Hernández O, Juárez P, Fournier PGJ. Development of Liver-Targeting α Vβ 5+ Exosomes as Anti-TGF-β Nanocarriers for the Treatment of the Pre-Metastatic Niche. BIOLOGY 2024; 13:1066. [PMID: 39765733 PMCID: PMC11673512 DOI: 10.3390/biology13121066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
Liver metastases frequently occur in pancreatic and colorectal cancer. Their development is promoted by tumor-derived exosomes with the integrin αVβ5 on their membrane. This integrin directs exosomes to the liver, where they promote a TGF-β-dependent pre-metastatic niche. We proposed the development of αVβ5+ exosomes to deliver anti-TGF-β therapy to the liver. This study demonstrates that the overexpression of αVβ5 in 293T cells allows its transfer to the secreted exosomes. αVβ5 overexpression increases exosome delivery to the liver, and αVβ5+ exosomes accumulate more in the liver compared to the lungs, kidneys, and brain in mice. We then sought 293T cells to directly produce and load an anti-TGF-β agent in their exosomes. First, we transduced 293T cells to express shRNAs against Tgfb1; however, the exosomes isolated from these cells did not knock down Tgfb1 in treated macrophages in vitro. However, when 293T expressed an mRNA coding a soluble form of betaglycan (sBG), a TGF-β inhibitor, this mRNA was detected in the isolated exosomes and the protein in the conditioned media of macrophages treated in vitro. In turn, this conditioned media decreased the TGF-β-induced phosphorylation of SMAD2/3 in hepatic cells in vitro. Our findings suggest that αVβ5+ exosomes could serve as nanocarriers for liver-targeted anti-TGF-β therapies.
Collapse
Affiliation(s)
- Paloma Acosta Montaño
- Posgrado en Ciencias de la Vida, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico; (P.A.M.); (E.O.F.); (V.C.F.); (A.H.G.)
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| | - Eréndira Olvera Félix
- Posgrado en Ciencias de la Vida, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico; (P.A.M.); (E.O.F.); (V.C.F.); (A.H.G.)
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| | - Veronica Castro Flores
- Posgrado en Ciencias de la Vida, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico; (P.A.M.); (E.O.F.); (V.C.F.); (A.H.G.)
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| | - Arturo Hernández García
- Posgrado en Ciencias de la Vida, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico; (P.A.M.); (E.O.F.); (V.C.F.); (A.H.G.)
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| | - Ruben D. Cadena-Nava
- Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada 22860, BC, Mexico;
| | - Octavio Galindo Hernández
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California (UABC), Mexicali 21000, BC, Mexico;
| | - Patricia Juárez
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| | - Pierrick G. J. Fournier
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, BC, Mexico;
| |
Collapse
|
50
|
Ji J, He Q, Xia Y, Sha X, Liang Q, Xu Y, Chen P, Dong C, Zhao R, Yang J, Guo H, Wang Y, Cao H, Li J, Yang M, Gu Z. Circulating plasma derived exosomes from systemic lupus erythematosus aggravate lupus nephritis through miR-122-5p/FOXO3-mediated macrophage activation. J Nanobiotechnology 2024; 22:779. [PMID: 39702207 DOI: 10.1186/s12951-024-03063-6] [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: 02/20/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic and systemic autoimmune disease characterized by dysregulation in both innate and adaptive immunity. Polarization of macrophages into M1/M2 macrophages affects the development of lupus. Exosomes-miRNA plays a crucial role in disease progression. This study aims to explore the mechanism of circulating exosomes participating in the pathogenesis of SLE and seek new therapeutic targets. Plasma derived-exosomes from SLE patients accelerated the disease progression and polarization of macrophages of the kidney in MRL/lpr mice. Exosomes were taken up by macrophages and stimulated macrophage polarization in vitro. MiRNA-sequence analysis revealed that plasma-derived exosomal miR-151a-5p, miR-1180a-5p, miR-1246 and miR-122-5p were abnormal. Of them, the expression of miR-122-5p was significantly upregulated in SLE exosomes, and positively correlated with systemic lupus erythematosus disease activity index (SLEDAI) and the dsDNA levels. Compared with SLE exosomes, inhibition of circulating exosomal miR-122-5p from SLE patients relieved lupus clinical aspects and polarization of macrophage. SLE exosomal miR-122-5p motivated M1 macrophage polarization by targeting FOXO3/NF-κB signaling pathway. Based on these findings, we conclude that SLE exosomal miR-122-5p can promote M1 macrophage polarization via targeting FOXO3/NF-κB signaling pathway and participate in pathogenesis of SLE. Collectively, plasma-derived exosomal miR-122-5p is a promising and effective target for treating SLE.
Collapse
Affiliation(s)
- Juan Ji
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian He
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Xiaoqi Sha
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian Liang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongxin Xu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Pengyu Chen
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chen Dong
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Rui Zhao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Junling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Hua Guo
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yunan Wang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Haixia Cao
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Jing Li
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Mei Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| |
Collapse
|