1
|
Payne JL, Sabunciyan S. Liquid biopsies in psychiatric disorders: Identifying peripheral biomarkers of brain health. Neural Regen Res 2026; 21:691-692. [PMID: 39819865 DOI: 10.4103/nrr.nrr-d-24-00962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Jennifer L Payne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA (Payne JL)
| | - Sarven Sabunciyan
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA (Sabunciyan S)
| |
Collapse
|
2
|
Yang X, Yue R, Zhao L, Wang Q. Integration of transcriptome and Mendelian randomization analyses in exploring the extracellular vesicle-related biomarkers of diabetic kidney disease. Ren Fail 2025; 47:2458767. [PMID: 39957315 PMCID: PMC11834810 DOI: 10.1080/0886022x.2025.2458767] [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: 07/17/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Diabetic Kidney Disease (DKD) is a common complication in patients with diabetes, and its pathogenesis remains incompletely understood. Recent studies have suggested that extracellular vesicles (EVs) may play a significant role in the initiation and progression of DKD. This study aimed to identify biomarkers associated with EVs in DKD through bioinformatics and Mendelian randomization (MR) analysis. METHODS This study utilized two DKD-related datasets, GSE96804 and GSE30528, alongside 121 exosome-related genes (ERGs) and 200 inflammation-related genes (IRGs). Differential analysis, co-expression network construction, and MR analysis were conducted to identify candidate genes. Machine learning techniques and expression validation were then employed to determine biomarkers. Finally, the potential mechanisms of action of these biomarkers were explored through Immunohistochemistry (IHC) staining, enrichment analysis, immune infiltration analysis, and regulatory network construction. RESULTS A total of 22 candidate genes were identified as causally linked to DKD. CMAS and RGS10 were identified as biomarkers, with both showing reduced expression in DKD. IHC confirmed low RGS10 expression, providing new insights into DKD management. CMAS was involved primarily in mitochondria-related pathways, while RGS10 was enriched in the extracellular matrix and associated pathways. Significant differences were observed in neutrophils and M2 macrophages between DKD and normal groups, correlating strongly with the biomarkers. CONCLUSION This study identified two EV-associated biomarkers, CMAS and RGS10, linked to DKD and elucidated their potential roles in disease progression. These results offer valuable insights for further exploration of DKD pathogenesis and the development of new therapeutic targets.
Collapse
Affiliation(s)
- Xu Yang
- Second Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rensong Yue
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liangbin Zhao
- Second Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiyue Wang
- Department of Pediatrics, Chengdu Jinniu Hospital of TCM, Chengdu, China
| |
Collapse
|
3
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
4
|
Felici E, González-Martínez C, Griñán TV, Gato-Zambrano S, Pereira SV, Fernández-Baldo MA, Ortega-Sanchez FG. Electrochemical immunoplatform for the quantification of epithelial extracellular vesicles applied to prostate cancer diagnosis. Talanta 2025; 293:128130. [PMID: 40222093 DOI: 10.1016/j.talanta.2025.128130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/27/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed cancer in men worldwide, and its early detection is critical for improving patient outcomes through timely and effective treatment. In this work, we present the first electrochemical immunoplatform based on magnetic microbeads (MBs) for the determination of epithelial extracellular vesicles (EpEVs), which are emerging as promising biomarkers for PCa diagnosis and prognosis. The immunoplatform employs MBs functionalized with anti-EpCAM antibodies to selectively capture EpEVs, forming sandwich-type immune complexes that are detected via amperometry at disposable screen-printed carbon electrodes. The method demonstrated a detection limit of 0.4 ng μL-1 of EpEVs obtained from PC-3 cell line's culture, excellent reproducibility (coefficient of variation <5 %), and high selectivity against potential interferences. Comparative analysis with colorimetric immune-magnet ELISA test showed a strong correlation between the two methods, confirming the reliability of the proposed approach. Furthermore, the electrochemical platform provided better precision and a lower limit of detection than the immune magnet ELISA method, indicating its superior analytical performance. Clinical validation using patient samples revealed that the combination of EpEV detection with PSA levels significantly improves the sensitivity and specificity of PCa diagnosis. This novel immunoplatform represents a promising analytical tool for early detection and monitoring of PCa, with potential applications in personalized cancer management.
Collapse
Affiliation(s)
- Emiliano Felici
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Instituto de Química de San Luis, INQUISAL (UNSL - CONICET), Av. Ejército de los Andes 950, San Luis, D5700BWS, Argentina
| | - Coral González-Martínez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada, 18016, Spain; Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, Granada, 18071, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de Madrid, 15, Granada, 18012, Spain
| | - Teresa Valero Griñán
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada, 18016, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de Madrid, 15, Granada, 18012, Spain; Department of Medicinal and Organic Chemistry, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain.
| | - Sheila Gato-Zambrano
- Seliver Group, Institute of Biomedicine of Seville/Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Sirley V Pereira
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Instituto de Química de San Luis, INQUISAL (UNSL - CONICET), Av. Ejército de los Andes 950, San Luis, D5700BWS, Argentina
| | - Martín A Fernández-Baldo
- Universidad Nacional de San Luis, Facultad de Química, Bioquímica y Farmacia, Instituto de Química de San Luis, INQUISAL (UNSL - CONICET), Av. Ejército de los Andes 950, San Luis, D5700BWS, Argentina.
| | - Francisco G Ortega-Sanchez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada, 18016, Spain; Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, Granada, 18071, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de Madrid, 15, Granada, 18012, Spain.
| |
Collapse
|
5
|
Khameneh AJ, Kordzadeh A, Rastgoo A, Hadi A. Investigation of the temperature effect on the properties of biological nanomembranes with different concentrations of cholesterol using molecular dynamics simulation. J Mol Graph Model 2025; 139:109074. [PMID: 40382911 DOI: 10.1016/j.jmgm.2025.109074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025]
Abstract
The cell membrane is the outermost boundary of the cell, and its role is to protect the cell. Drugs must first pass through the membrane to enter the cell; therefore, investigating the properties of the bilayer membrane is of great importance and can provide important information regarding drug delivery processes. In this study, the effect of increasing temperature on the properties of bilayer membranes with 0 %, 10 %, 30 %, and 50 % cholesterol was investigated using molecular dynamics simulation. Moreover, the phase transition temperature of the lipid bilayer was selected based on the percentages of cholesterol. The results indicated that with increasing temperature, the membrane changed from gel phase to fluid phase. Moreover, a rise in temperature led to an increase in the area per lipid and a decrease in the order parameter, the reason for which was the increase in the kinetic energy of the molecules. The fluid phase membrane, which is at a higher temperature, had a higher diffusion coefficient than the gel phase membrane. The increase in the temperature caused the membrane with 0 % cholesterol to experience a 24 % decrease in the order parameter, while the structural properties of the membrane with 50 % cholesterol are almost constant with increasing temperature, indicating the rigidity of the membrane. With the increase in cholesterol concentration from 0 % to 50 %, the time for pore formation under the electric field increased from 0.26 ns to 8.46 ns. These results will be helpful in the development of drug delivery and therapeutic processes.
Collapse
Affiliation(s)
- Armin Jarahi Khameneh
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Azadeh Kordzadeh
- Chemical and Petroleum Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Abbas Rastgoo
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Amin Hadi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| |
Collapse
|
6
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
7
|
Zhang AY, Xie QZ, Guo SZ, Liu X, Yu YH, Tang H, Yao H, Guo L, Xu SB, Bian XT, Tao X. Platelet-rich plasma-derived exosomes have the novel ability to alleviate insertional Achilles tendinopathy by promoting tenogenesis in tendon stem/progenitor cells. BIOMATERIALS ADVANCES 2025; 173:214272. [PMID: 40081287 DOI: 10.1016/j.bioadv.2025.214272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Insertional Achilles tendinopathy (IAT) is a highly prevalent overuse injury affecting the foot and ankle in clinical settings. Currently, the primary management approach is conservative treatment. Platelet-rich plasma-derived exosomes (PRP-Exos) effectively preserve essential growth factors and other vital components inherent in PRP, thereby optimizing overall treatment outcomes. Furthermore, the standardized microinjection technique for PRP-Exos significantly enhances the treatment experience for patients. In this study, PRP-Exos were isolated from SD rats, and their effects on proliferation, migration, differentiation, apoptosis and other physiological processes in tendon-derived stem cells (TDSCs) in an IL-1β-induced inflammatory state were investigated in vitro. In this context, we conducted a thorough investigation of the impact of PRP-Exos on the tendinogenic differentiation of TDSCs under inflammatory conditions and explored the underlying mechanisms through cellular RNA sequencing. In vivo, the therapeutic effects of PRP-Exos on IAT at different times after treatment were evaluated comprehensively via histological analysis, behavioral tests and biomechanical tests. The results showed that PRP-Exos significantly increased the proliferation and migration of TDSCs in an inflammatory state in vitro and promoted their differentiation into tendon cells. Animal experiments confirmed that the histology, biomechanical performance and behavior of the animals in the PRP-Exos group were significantly normalized. This work demonstrated that the topical use of PRP-Exos at the insertion site of the Achilles tendon is an effective strategy for regulating proliferation and tendinogenic differentiation and represents a novel treatment approach for IAT.
Collapse
Affiliation(s)
- An-Yang Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Qi-Zhong Xie
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Shi-Zhen Guo
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Xiao Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Yi-Hang Yu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Hang Yao
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Lin Guo
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Shi-Bo Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China.
| | - Xu-Ting Bian
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Shigatse Branch, Xinjian Hospital, Third Military Medical University, Shigatse 857000, China.
| | - Xu Tao
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Foot, Ankle and Hand Surgery Department, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
8
|
Pérez-Isidoro R, Díaz-Salazar AJ, Pérez-Casas S, Bernal-Sánchez LJ, Saldívar-Guerra E, Guevara-Pantoja FJ. Liposome dimensions at a glance by bright-field microscopy and fractal analysis. Micron 2025; 195:103822. [PMID: 40222210 DOI: 10.1016/j.micron.2025.103822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Patterns from evaporated micro-droplets of complex fluids represent an emerging method for biological analysis. Although the driving forces behind the interactions in droplet evaporation of liquids have been vastly explored, an analytical technique derived from this knowledge needs to be well-established. In this work, we take advantage of the knowledge of the patterns found after the evaporation of microdroplets of lipid vesicles with potassium chloride to introduce a reliable method to differentiate the micro- and nano-size of liposomes composed of zwitterionic and anionic phospholipids. We use a statistical index of complexity, fractal dimension, to correlate the liposome size with the pattern index. We conclude that the values of fractal dimensions of 1.7 ( ± 0.1) that describe a monofractal can be related to the nanoscale range of unilamellar vesicles; otherwise, the vesicle sizes range the microscale. This suggests that our method could be a practical reference to reveal the nanoscale dimension of several systems in biophysics, bioengineering, and nanotechnology.
Collapse
Affiliation(s)
- R Pérez-Isidoro
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, Saltillo, Coahuila 25294, Mexico
| | | | - S Pérez-Casas
- Facultad de Química, Universidad Nacional Autónoma de México, México City 04510, Mexico
| | - L J Bernal-Sánchez
- Facultad de Química, Universidad Nacional Autónoma de México, México City 04510, Mexico
| | - E Saldívar-Guerra
- Centro de Investigación en Química Aplicada (CIQA), Enrique Reyna, 140, Saltillo, Coahuila 25294, Mexico
| | | |
Collapse
|
9
|
Cao LM, Qiu YZ, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Wu Q, Bu LL. Extracellular Vesicles: Hermes between cancers and lymph nodes. Cancer Lett 2025; 623:217735. [PMID: 40268131 DOI: 10.1016/j.canlet.2025.217735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Cancer is one of the main causes of death and a major obstacle to increasing life expectancy in all countries of the world. Lymph node metastasis (LNM) of in cancer patients indicates poor prognosis and it is an important indication to determine the therapeutic regime. Therefore, more attention should be given to the molecular mechanics of tumor lymphangiogenesis and LNM. Extracellular vesicles (EVs) are nanoscale cargo-bearing membrane vesicles that can serve as key mediators for the intercellular communication. Like Hermes, the messenger of the Greek gods, EVs can be secreted by tumor cells to regulate the LNM process. Many evidence has proved the clinical correlation between EVs and LNM in various cancer types. EVs plays an active role in the process of metastasis by expressing its connotative molecules, including proteins, nucleic acids, and metabolites. However, the clear role of EVs in the process of cancer LNM has not been thoroughly studied yet. In this review, we will summarize the clinical and mechanical findings of EVs regulating role on cancer LNM, and discuss the advanced modification of the research proposal. We propose the "PUMP" principle of EVs in LNM, including Preparation, Unleash, Migration, and Planting.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yu-Zhong Qiu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Han-Yue Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
10
|
Xu S, Zhang Z, Zhou X, Liao Y, Peng Z, Meng Z, Nüssler AK, Ma L, Xia H, Liu L, Yang W. Gouqi-derived Nanovesicles (GqDNVs) promoted MC3T3-E1 cells proliferation and improve fracture healing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156755. [PMID: 40252435 DOI: 10.1016/j.phymed.2025.156755] [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: 09/18/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Lycium barbarum L., also known as Gouqi, a traditional Chinese herbal medicine, is widely utilized in health care products and clinical therapies. Its muscle and bone strengthening efficacy has been recorded in medical classics for a long time. In addition, plant exosome-like nanovesicles (PELNVs) have attracted more and more attention owing to their biological traits. Therefore, we intended to explore the functions, regulatory role, and underlying mechanism of Gouqi-derived Nanovesicles (GqDNVs) on fracture healing. METHODS In this study, we employed the sucrose density gradient differential ultracentrifugation to isolate GqDNVs. The effects of GqDNVs on the proliferation and differentiation of MC3T3-E1 cells were evaluated using the CCK-8 assay, ALP activity measurement, and cell scratch assay. Additionally, leveraging a fracture mouse model, we utilized Micro-CT, immunological staining, and histologic analyses to comprehensively assess the impact of GqDNVs on fracture healing in mice. RESULTS GqDNVs stimulated cell viability, increased ALP activity, and promoted cellular osteogenic protein expression (OPN, ALP, and RUNX2). Subsequently, in the mouse fracture model, trabecular thickness, and bone marrow density were increased in the GqDNVs treatment group after 28 days of injection. Meanwhile, the expressions of OPN and BGP were significantly elevated after both 14 and 28 days. Additionally, the expressions of p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, p-4EBP1/4EBP1 and p-p70S6K/ p70S6K were also increased after14 days of treatment. CONCLUSIONS GqDNVs effectively promoted the proliferation and differentiation of MC3T3-E1 cells. Furthermore, GqDNVs could improve fracture healing, which is associated with PI3K/Akt/mTOR/p70S6K/4EBP1 signaling pathway.
Collapse
Affiliation(s)
- Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, Tübingen 72076, Germany
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China.
| |
Collapse
|
11
|
Seegobin N, Taub M, Vignal C, Waxin C, Chris V, Awad A, Murdan S, Basit AW. Small milk-derived extracellular vesicles: Suitable vehicles for oral drug delivery? Eur J Pharm Biopharm 2025; 212:114744. [PMID: 40355010 DOI: 10.1016/j.ejpb.2025.114744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Current treatments for inflammatory bowel disease often fail due to systemic side effects, but bovine milk-derived extracellular vesicles (EVs) show promise for targeted delivery to inflamed gut tissue via the leaky gut effect. This study assessed the stability of EVs as drug carriers in simulated gastrointestinal (GI) fluids and their efficacy in a colitis mouse model. EVs were characterised after incubation in PBS at various pH levels, and their lipid bilayer stability in biorelevant GI fluids was evaluated using the polar probe laurdan. Two small molecules, acridine orange (lipophilic) and riboflavin (hydrophilic), were loaded into EVs to test their release under GI conditions, while unloaded EVs were investigated for therapeutic effect via oral gavage or rectal enema in a colitis mouse model. Although no significant changes in EVs' physical properties were observed at different pH levels, lipid bilayer damage was evident in acidic (p ≤ 0.05) and enzyme-rich environments (p ≤ 0.01). Acridine orange release was significant (p ≤ 0.05), butriboflavin remained encapsulated, and no therapeutic effect was observed with unloaded EVs in vivo. These results suggest that physical characterisation alone does not reflect EV stability, that bovine milk EVs have limited potential for oral drug delivery and are better suited for hydrophilic drugs.
Collapse
Affiliation(s)
- Nidhi Seegobin
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Marissa Taub
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, UMR1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, UMR1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Victoria Chris
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Atheer Awad
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, United Kingdom
| | - Sudaxshina Murdan
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Abdul W Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom.
| |
Collapse
|
12
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [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: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
13
|
Avalos-de Leon CG, Thomson AW. Regulatory Immune Cell-derived Exosomes: Modes of Action and Therapeutic Potential in Transplantation. Transplantation 2025; 109:1124-1137. [PMID: 39865513 PMCID: PMC12187562 DOI: 10.1097/tp.0000000000005309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Reduced dependence on antirejection agents, improved long-term allograft survival, and induction of operational tolerance remain major unmet needs in organ transplantation due to the limitations of current immunosuppressive therapies. To address this challenge, investigators are exploring the therapeutic potential of adoptively transferred host- or donor-derived regulatory immune cells. Extracellular vesicles of endosomal origin (exosomes) secreted by these cells seem to be important contributors to their immunoregulatory properties. Twenty years ago, it was first reported that donor-derived exosomes could extend the survival of transplanted organs in rodents. Recent studies have revealed that regulatory immune cells, such as regulatory myeloid cells (dendritic cells, macrophages, or myeloid-derived suppressor cells), regulatory T cells, or mesenchymal stem/stromal cells can suppress graft rejection via exosomes that express a cargo of immunosuppressive molecules. These include cell surface molecules that interact with adaptive immune cell receptors, immunoregulatory enzymes, and micro- and long noncoding RNAs that can regulate inflammatory gene expression via posttranscriptional changes and promote tolerance through promotion of regulatory T cells. This overview analyzes the diverse molecules and mechanisms that enable regulatory immune cell-derived exosomes to modulate alloimmunity and promote experimental transplant tolerance. We also discuss the potential benefits and limitations of their application as therapeutic entities in organ transplantation.
Collapse
Affiliation(s)
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, Pittsburgh PA, USA
- Department of Immunology, University of Pittsburgh School of medicine, Pittsburgh PA, USA
| |
Collapse
|
14
|
Malik SZA, Muhilan Y, Nordin F, Ng MH, Law JX, Imran SAM, Idris IM, Tye GJ. Stem cell derived exosome trilogy: an epic comparison of human MSCs, ESCs and iPSCs. Stem Cell Res Ther 2025; 16:318. [PMID: 40551257 PMCID: PMC12186388 DOI: 10.1186/s13287-025-04440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 06/10/2025] [Indexed: 06/28/2025] Open
Abstract
Exosomes, containing molecular constituents of their cell of origin, including proteins and nucleic acids, were first discovered in immature red blood cells in 1983. Excellent intercell communication can be achieved by shuttling these various molecules between cells. Stem cell-derived exosomes (SC-Exos) contain paracrine-soluble factors that play important roles in tissue development, homeostasis, and regeneration. This paracrine activity of SC-Exos has been found to be a predominant mechanism by which stem cell-based therapies mediate their effects on degenerative, autoimmune and/or inflammatory diseases. Compared to other types of stem cells, human embryonic stem cells (hESCs), human induced pluripotent stem cells (hiPSCs), human mesenchymal stem cells (hMSCs) are the most popular because of their efficient immunomodulatory effects. The advantages and disadvantages of using exosomes isolated from the stem cell trio for therapeutic applications are further discussed in this review.
Collapse
Affiliation(s)
- Siti Zawiah Abdul Malik
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Yugashini Muhilan
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Fazlina Nordin
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Siti A M Imran
- Ming Medical Sdn. Bhd., Dana 1 Commercial Centre, Jalan PJU 1a/46, Petaling Jaya, 47301, Selangor, Malaysia
| | - Izyan Mohd Idris
- Institute for Medical Research (IMR), National Institutes of Health (NIH), Ministry of Health Malaysia, Shah Alam, Selangor, Malaysia
| | - Gee Jun Tye
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Halaman Bukit Gambir, 11700, Gelugor, Pulau Pinang, Malaysia
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| |
Collapse
|
15
|
Ahmad P, Estrin N, Farshidfar N, Zhang Y, Miron RJ. Isolation methods of exosomes derived from dental stem cells. Int J Oral Sci 2025; 17:50. [PMID: 40523888 DOI: 10.1038/s41368-025-00370-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 06/19/2025] Open
Abstract
Mesenchymal stem cells are highly regarded for their potential in tissue repair and regenerative medicine due to their multipotency and self-renewal abilities. Recently, mesenchymal stem cells have been redefined as "medical signaling cells," with their primary biological effects mediated through exosome secretion. These exosomes, which contain lipids, proteins, RNA, and metabolites, are crucial in regulating various biological processes and enhancing regenerative therapies. Exosomes replicate the effects of their parent cells while offering benefits such as reduced side effects, low immunogenicity, excellent biocompatibility, and high drug-loading capacity. Dental stem cells, including those from apical papilla, gingiva, dental pulp, and other sources, are key contributors to exosome-mediated regenerative effects, such as tumor cell apoptosis, neuroprotection, angiogenesis, osteogenesis, and immune modulation. Despite their promise, clinical application of exosomes is limited by challenges in isolation techniques. Current methods face issues of complexity, inefficiency, and insufficient purity, hindering detailed analysis. Recent advancements, such as micro-electromechanical systems, alternating current electroosmosis, and serum-free three-dimensional cell cultures, have improved exosome isolation efficacy. This review synthesizes nearly 200 studies on dental stem cell-derived exosomes, highlighting their potential in treating a wide range of conditions, including periodontal diseases, cancer, neurodegenerative disorders, diabetes, and more. Optimized isolation methods offer a path forward for overcoming current limitations and advancing the clinical use of exosome-based therapies.
Collapse
Affiliation(s)
- Paras Ahmad
- Department of Research, Advanced PRF Education, Bradenton, FL, USA
| | - Nathan Estrin
- Lake Erie College of Osteopathic Medicine School of Dental Medicine, Bradenton, FL, USA
| | - Nima Farshidfar
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Fang W, Wang G, Lin L, Ajoolabady A, Ren J. Extracellular vesicles in skin health and diseases. Life Sci 2025; 378:123813. [PMID: 40527444 DOI: 10.1016/j.lfs.2025.123813] [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: 04/09/2025] [Revised: 06/03/2025] [Accepted: 06/11/2025] [Indexed: 06/19/2025]
Abstract
The global burden of skin diseases has markedly increased in recent years, posing significant socioeconomic challenges. Although recent technological advances in dermatological care aim to address these issues, further research is needed to develop effective and innovative strategies. Intercellular communication plays a vital role in skin regeneration and repair. Extracellular vesicles (EVs), lipid bilayer-enclosed particles released by nearly all cell types, are categorized into exosomes, microvesicles, and apoptotic bodies based on their biogenesis and size. EVs carry diverse bioactive molecules, including proteins, lipids, and nucleic acids, enabling them to mediate critical cell-cell communication. Recent breakthroughs, particularly through spatially resolved multi-omics approaches, have underscored the essential roles of EVs in both physiological and pathological processes of the skin. In this review, we provide a comprehensive overview of EV biology, with a particular focus on their functions in skin homeostasis, aging, and diseases. We highlight emerging evidence of their therapeutic potential in preclinical models, with emphasis on EV interactions with various skin cell types, inflammatory and autoimmune skin conditions, skin aging, and tissue repair.
Collapse
Affiliation(s)
- Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China.
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Ling Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
| | - Amir Ajoolabady
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China.
| |
Collapse
|
17
|
Akter A, Kamal T, Akter S, Auwal A, Islam F. Exosomes: a potential tool in the diagnosis, prognosis and treatment of patients with colorectal cancer. Future Oncol 2025:1-19. [PMID: 40515703 DOI: 10.1080/14796694.2025.2520150] [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: 11/15/2024] [Accepted: 06/11/2025] [Indexed: 06/16/2025] Open
Abstract
Colorectal cancer (CRC), a commonly diagnosed malignancy, is one of the most frequent causes of cancer-related deaths worldwide. To effectively lower the death rate from this disease, it is essential to create public health methods, including developing new biomarkers that facilitate screening, diagnosis, prognosis, and therapy response prediction. CRC-derived Exosomes are a type of extracellular vesicle that transport functional molecules like proteins, lipids, nucleic acids (DNA, mRNA, miRNA, lncRNA, and noncoding RNA), and other metabolites, which act as molecular cargos to facilitate transportation. Exosomes generated and secreted from cancer cells are key biomarkers for early, noninvasive cancer diagnosis, prognosis, and treatment response, with their biogenesis in CRC offering molecular insights. Their expression varies across time, tissues, and disease stages. Thus, the development of innovative and effective techniques for isolating and detecting exosomes holds great potential for tumor diagnosis, prognosis prediction, and developing techniques (MSC-derived exosome, DC-derived exosome, engineered exosome, etc.) and their contents to improve the specificity and efficacy of therapies for patients with CRC. This review explores the features and formation of CRC-derived exosomes, highlighting their diagnostic, prognostic, and therapeutic significance through a comprehensive analysis of exosome extraction, identification, purification, and documented biological roles in existing literature.
Collapse
Affiliation(s)
- Azmin Akter
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Tasnima Kamal
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Sharmin Akter
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Abdul Auwal
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
- School of Medicine and Dentsitry, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
18
|
Zheng Y, Ruan Z, Liu S, Yang X, Chen Z. Exosome-mediated macrophage polarization: Pioneering pathways in diabetic wound healing. Int Immunopharmacol 2025; 161:115058. [PMID: 40516253 DOI: 10.1016/j.intimp.2025.115058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 06/01/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025]
Abstract
Recent studies have identified an essential role of macrophage polarization imbalance in the impaired diabetic wound healing process. Macrophages are key players in wound healing, and their transition from the pro-inflammatory M1 phenotype to the anti-inflammatory and reparative M2 phenotype is crucial for effective wound repair. However, in diabetic conditions, this balance is disrupted, leading to prolonged M1 activation and reduced M2 functionality, which hinders the healing process. Exosomes, known for their role in intercellular communication, have garnered significant interest for their diverse functions in immune modulation, angiogenesis, inflammation regulation, and tissue regeneration. Research has demonstrated the ability of exosomes to modulate macrophage polarization, facilitating the shift from M1 to M2 phenotypes, ameliorating the inflammatory milieu, and thereby promoting tissue repair and regeneration. However, the specific mechanisms underlying exosome-mediated regulation of macrophage phenotypic transitions in diabetic wounds remain inadequately elucidated. Moreover, exosomes, serving as novel drug delivery vehicles, present advantages such as enhanced targeting, reduced immunogenicity, and prolonged drug presence, offering considerable promise for diabetic wound management. This review comprehensively outlines advancements in understanding how exosomes influence macrophage polarization in diabetic wound healing, emphasizing the pathophysiological processes of diabetic wounds, the dysregulation of macrophage polarization, and their interactions with exosomes. It also assesses current research limitations and clinical challenges while proposing future research directions, thereby introducing novel theoretical perspectives and potential strategies for diabetic wound therapy.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhichao Ruan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research (Huazhong University of Science and Technology), Wuhan, Hubei 430022, China.
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Clinical Research Center for Chronic Wound and Diabetic Foot,Wuhan, Hubei 430077, China.
| |
Collapse
|
19
|
Lu X, Xu R, Dong X, Bai D, Ji W, Chen X, Chen H, Hou C, Gao J. Cell-derived exosome therapy for diabetic peripheral neuropathy: a preclinical animal studies systematic review and meta-analysis. Stem Cell Res Ther 2025; 16:297. [PMID: 40490808 DOI: 10.1186/s13287-025-04432-0] [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: 04/23/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUNDS Exosomes is a promising cell-free therapy for Diabetic peripheral neuropathy (DPN) that imposes long-term negative effects on patients' finances, mental health, and quality of life. We conducted a meta-analysis to assess the therapeutic effects of exosomes (such as SCs-derived, FCs-derived, BMSCs-derived, MSCs-derived, and Plasma-derived) on DPN. METHODS We searched nine databases from inception to February 2025, then two researchers independently screened studies, extracted data, and assessed the quality of included studies using SYRCLE's tool. The outcome indicators consisted of at least one of the three key DPN endpoints (electrophysiology, behavioural assessment, and nerve structure) based on the Neurodiab guidelines. R 4.4.2 software was used to conduct all statistical analyses. RESULTS 11 studies were identified, and the risk of bias in most studies was unclear generally. Pooled analyses demonstrated that exosome improved the nerve conduction velocity [MCV (SMD = 4.71 [2.18;7.25], P = 0.0003; I²= 91.8%), SCV (SMD = 1.07 [0.30;1.85], P = 0.0069; I²= 85.3%)], may restore IENFD [SMD = 1.46 [-0.85; 3.77], P = 0.2164; I²=88.7%], alleviated neuropathic pain [mechanical allodynia (SMD= -0.27 [-1.02;0.47], P = 0.4697; I2 = 85.0%), thermal hyperalgesia (SMD= -1.48 [-2.45;-0.50], P = 0.003; I2 = 88.4%)], ameliorated vascular function [blood flow perfusion in plantar (SMD = 2.84 [0.89; 4.80], P = 0.0043; I2 = 74.9%), blood flow perfusion in sciatic nerves (SMD = 2.62 [0.80; 4.43], P = 0.0047; I2 = 75.9%), vessel density (SMD = 2.69 [0.90; 4.49], P = 0.0032; I2 = 0%)], and restored the peripheral nerve structure [sciatic nerve fiber diameter (SMD = 3.29 [1.61; 4.96], P = 0.0066; I2 = 75.5%), axon diameter (SMD = 2.26 [1.64; 2.88], P < 0.0001; I2 = 54.3%), myelin sheath thickness (SMD = 2.56 [1.39; 3.72], P < 0.0001; I2 = 73.0%), g-ratio (SMD= -1.64 [-3.28; 0.00], P = 0.0502; I2 = 34.17)]. Furthermore, after exosome therapy, the expressions of NF-200 (SMD = 2.57 [0.39; 4.75], P = 0.0210; I2 = 33.0%), MBP (SMD = 2.27 [-1.49; 6.02], P = 0.1064; I2 = 59.0%), and S-100β (SMD = 1.90 [0.09; 3.72], P = 0.0399; I2 = 32.5%) evaluating axonal regeneration and remyelination increased significantly. Notably, high-glucose pretreatment of exosomes significantly attenuated these effects, while genetic overexpression modifications or novel dressings-mediated delivery partially counteracted this suppression. CONCLUSIONS Exosome therapy provides a novel therapeutic strategy for the benefit of neurovascular remodeling and functional recovery of DPN, especially when used in conjunction with exosome modification and novel dressings. To bridge the translational gap between preclinical and clinical studies, future research should conduct more large-scale, meticulously designed preclinical trials adhering to ARRIVE criteria before proceeding to clinical translation, to enhance translational rigor and mitigate risks associated with variability in study design.
Collapse
Affiliation(s)
- Xianying Lu
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ran Xu
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohui Dong
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dingxi Bai
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenting Ji
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Chen
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Chen
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaoming Hou
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jing Gao
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
20
|
Poudel S, He Z, Izac J, Wang L. Gene Copy Number Dictates Extracellular Vesicle Cargo. Int J Mol Sci 2025; 26:5496. [PMID: 40564958 DOI: 10.3390/ijms26125496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/31/2025] [Accepted: 06/03/2025] [Indexed: 06/28/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-surrounded vesicles that carry heterogeneous cellular components, including proteins, nucleic acids, lipids, and metabolites. EVs' intravesicular and surface contents possess many biomarkers of physiological and pathological importance. Because of the heterogeneous cargo, EVs can mediate local and distal cell-cell communication. However, the way in which the genome signature regulates EV cargo has not been well studied. This study aimed to understand how genetics impact EV cargo loading. EVs were isolated from vector copy number cells with a fluorescent reporter (GFP) with varying inserted transgene copies and from NIST SRM 2373 cells (MDA-MB-231, MDA-MB-453, SK-BR-3, and BT-474), which contain varying copies of the HER2 gene. Spectradyne nCS1 was utilized to count EVs and measure size distribution. Imaging Flow Cytometry was used to analyze the surface protein content of single EVs and for total EV counts. The RNA content of the EVs was measured using ddPCR. Our results from stable reporter cell lines and breast cancer cell lines suggest that the gene copy number dictates the protein cargo of the EVs but not the RNA content. Increasing copies of a reporter gene (GFP) or a naturally occurring gene (HER2) from breast cancer cells correlated with increasing EV counts positive for the protein cargo compared to total EV counts until a copy threshold was reached. This study has broad implications for understanding EV biology in the context of cancer biology, diagnostics, EV biology/manufacturing, and therapeutic delivery.
Collapse
Affiliation(s)
- Sumeet Poudel
- National Institute of Standards and Technology (NIST), Gaithersburg, MD 20817, USA
| | - Zhiyong He
- National Institute of Standards and Technology (NIST), Gaithersburg, MD 20817, USA
| | - Jerilyn Izac
- National Institute of Standards and Technology (NIST), Gaithersburg, MD 20817, USA
| | - Lili Wang
- National Institute of Standards and Technology (NIST), Gaithersburg, MD 20817, USA
| |
Collapse
|
21
|
Almasry Y, Mustafa F, Alfuwais M, AlNachef S, Mohamed H, Gaber NS, Khan MI, Saadeldin IM, Yaqinuddin A. Current trends in theranostic applications of extracellular vesicles in cancer. Front Oncol 2025; 15:1592006. [PMID: 40530018 PMCID: PMC12170317 DOI: 10.3389/fonc.2025.1592006] [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: 03/11/2025] [Accepted: 05/05/2025] [Indexed: 06/20/2025] Open
Abstract
Background Extracellular vesicles (EVs) play an integral role in cancer biology, influencing tumor progression, metastasis, and tumor microenvironment. Due to their distinctive molecular composition, including proteins, nucleic acids, and lipids, EVs present a promising candidate for cancer diagnostics and precision therapeutics. Methods This review was conducted by looking up recent studies obtained through PubMed, Scopus, and Web of Science databases using targeted keywords such as "Extracellular Vesicles," "Cancer Therapy," "Biomarkers," "Exosomes," "Tumor Microenvironment," and "Precision Medicine." From an initial 4,320 articles identified, 427 were screened after applying publication filters, resulting in the inclusion of 298 articles relevant to EV isolation, characterization, diagnostic sensitivity, specificity, and therapeutic efficacy. Results Biomarkers derived from EVs derived across various cancers showed high diagnostic performance. For example, four miRNA EVs showing sensitivity and specificity of 98% and 96% respectively was found in breast cancer. EV-RNA and surface antigen analyses for hepatocellular carcinoma with 93.8% sensitivity and 74.5% specificity. Additionally, EV biomarker cancers of the colorectal microRNA miR-23a and miR-301a had 89% sensitivity and >70% specificity. EVs in a therapeutic context were an effective drug delivery system for enhancing precision of chemotherapy and immunotherapy with reduced systemic toxicity. Conclusion The theranostics of EVs provide great capacity for early cancer diagnosis and personalized treatment based on their high diagnostic sensitivity and specificity. Future standardization protocols are essential to translate EV technologies into clinical oncology.
Collapse
Affiliation(s)
- Yazan Almasry
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fayrouz Mustafa
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Sara AlNachef
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hager Mohamed
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Mohammed Imran Khan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Research Center, King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
| | - Islam M. Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | |
Collapse
|
22
|
Isogai T, Hirosawa KM, Kanno M, Sho A, Kasai RS, Komura N, Ando H, Furukawa K, Ohmi Y, Furukawa K, Yokota Y, Suzuki KG. Extracellular vesicles adhere to cells primarily by interactions of integrins and GM1 with laminin. J Cell Biol 2025; 224:e202404064. [PMID: 40304687 PMCID: PMC12042775 DOI: 10.1083/jcb.202404064] [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: 04/12/2024] [Revised: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-derived extracellular vesicles (EVs) have attracted significant attention, yet the molecular mechanisms that govern their specific binding to recipient cells remain elusive. Our in vitro study utilizing single-particle tracking demonstrated that integrin heterodimers comprising α6β4 and α6β1 and ganglioside, GM1, are responsible for the binding of small EV (sEV) subtypes to laminin. EVs derived from four distinct tumor cell lines, regardless of size, exhibited high binding affinities for laminin but not for fibronectin, although fibronectin receptors are abundant in EVs and have functional roles in EV-secreting cells. Our findings revealed that integrins in EVs bind to laminin via the conventional molecular interface, facilitated by CD151 rather than by inside-out signaling of talin-1 and kindlin-2. Super-resolution movie observation revealed that sEV integrins bind only to laminin on living recipient cells. Furthermore, sEVs bound to HUVEC and induced cell branching morphogenesis in a laminin-dependent manner. Thus, we demonstrated that EVs predominantly bind to laminin on recipient cells, which is indispensable for cell responses.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | | | - Miki Kanno
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Ayano Sho
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Rinshi S. Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hiromune Ando
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yasunari Yokota
- Department of Information Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Kenichi G.N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
23
|
Fan B, Wang L, Hu T, Zheng L, Wang J. Exosomal miR-196a-5p Secreted by Bone Marrow Mesenchymal Stem Cells Inhibits Ferroptosis and Promotes Drug Resistance of Acute Myeloid Leukemia. Antioxid Redox Signal 2025; 42:933-953. [PMID: 40388337 DOI: 10.1089/ars.2024.0882] [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] [Indexed: 05/21/2025]
Abstract
Background: Ferroptosis is a nonapoptotic type of cell death characterized by an increase in lipid reactive oxygen species (ROS). Acute myeloid leukemia (AML)-derived bone marrow mesenchymal stem cells (AML-BMSCs) support the progression and drug resistance of AML by secreting various bioactive substances, including exosomes. However, the role of BMSCs in regulating lipid metabolism and ferroptosis in AML remains unexplored. Results: Exosomes secreted by AML-BMSCs increased the expression of miR-196a-5p in AML cells. MiR-196a-5p promoted the proliferation of AML cells, reduced lipid ROS and ferroptosis, and was associated with poor prognosis in AML patients. Mechanistically, miR-196a-5p inhibited the expression level of neural precursor cell expressed developmentally down-regulated 4-like (NEDD4L). Co-immunoprecipitation (CO-IP) analysis showed that NEDD4L was bound to long-chain acyl-CoA synthetase (ACSL)3 and promoted ubiquitin-mediated degradation of ACSL3 protein. In addition, we also demonstrated that AML-BMSCs highly expressed Ras-associated binding protein 7A (RAB7A), which was associated with exosomal miR-196a-5p release. Importantly, cytarabine (Ara-C) activated the expression of RAB7A and promoted the secretion of exosomal miR-196a-5p, which weakened the ubiquitination of ACSL3 by NEDD4L, leading to ferroptosis inhibition and Ara-C resistance in AML. Innovation: This is the first time that exosomes secreted by BMSCs (BMSCs-exos) have been linked to ferroptosis in AML cells, thereby expanding our understanding of the mechanism of drug resistance in AML cells. High miR-196a-5p expression reduced lipid ROS levels and ferroptosis in AML cells by inhibiting NEDD4L-mediated ubiquitination of ACSL3. Conclusion: This study identified a new network through which BMSCs-exos regulate ferroptosis in AML cells. We combined BMSCs and AML cells to provide new ideas for drug research targeting exosome secretion and ferroptosis. Antioxid. Redox Signal. 42, 933-953.
Collapse
MESH Headings
- Ferroptosis/genetics
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Mesenchymal Stem Cells/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Exosomes/metabolism
- Exosomes/genetics
- Drug Resistance, Neoplasm/genetics
- Animals
- Mice
- Reactive Oxygen Species/metabolism
- Male
- Female
- Cell Line, Tumor
- Coenzyme A Ligases/metabolism
Collapse
Affiliation(s)
- Bingjie Fan
- Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, China
| | - Li Wang
- Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhen Hu
- Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lin Zheng
- Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Key Laboratory of Hematological Disease Diagnostic and Treatment Centre, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
24
|
Tamura T, Yamamoto T, Kogure A, Yoshioka Y, Yamamoto Y, Sakamoto S, Ichikawa T, Ochiya T. Extracellular Vesicles From Prostate Cancer-Corrupted Osteoclasts Drive a Chain Reaction of Inflammatory Osteolysis and Tumour Progression at the Bone Metastatic Site. J Extracell Vesicles 2025; 14:e70091. [PMID: 40545975 PMCID: PMC12183380 DOI: 10.1002/jev2.70091] [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: 08/15/2024] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 06/28/2025] Open
Abstract
Advanced-stage prostate cancer (PCa) frequently causes bone metastases, resulting in a poor prognosis and a 5-year survival rate of 30%. PCa bone metastasis is a highly complex and fluctuating process, comprising of osteolytic (bone-degrading) and osteogenic (bone-forming) lesions. Although this system is mainly controlled by alterations in the receptor activator of NF-κB ligand (RANKL), RANKL-based treatment does not prolong the overall survival of patients with PCa bone metastasis. Therefore, it is essential to understand the other interactions between tumour cells and bone-resident cells in the metastatic niche to develop novel treatments. Extracellular vesicles (EVs) play key roles in intercellular communication and actively function in the bone microenvironment. We report that PCa cells corrupt osteoclasts (OCs) via their secretomes, inducing a pathological phenotype. EVs from pathological OCs activate bone-resorbing OCs and suppress bone-forming osteoblasts (OBs), leading to bone destruction. Pathological OCs increased IL-1β secretion and produced EVs with miR-5112 and miR-1963, targeting Parp1 in OCs and Hoxa1 in OBs. This led to OC maturation and IL-1β secretion, and inhibited OB mineralization. Injection of these miRNAs in vivo promoted PCa metastasis-disrupting bone. We report the mediation of EVs from OCs under pathological conditions that modulate the bone metastatic niche independently of RANKL.
Collapse
Affiliation(s)
- Takaaki Tamura
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
- Department of UrologyGraduate School of MedicineChiba UniversityChibaJapan
| | - Tomofumi Yamamoto
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
- Division of Biological Chemistry and BiologicalsNational Institute of Health SciencesTokyoJapan
| | - Akiko Kogure
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
| | - Yusuke Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Shinichi Sakamoto
- Department of UrologyGraduate School of MedicineChiba UniversityChibaJapan
| | - Tomohiko Ichikawa
- Department of UrologyGraduate School of MedicineChiba UniversityChibaJapan
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
| |
Collapse
|
25
|
Tabaie EZ, Gao Z, Kachour N, Ulu A, Gomez S, Figueroa ZA, Bergersen KV, Zhong W, Wilson EH. Toxoplasma gondii infection of neurons alters the production and content of extracellular vesicles directing astrocyte phenotype and contributing to the loss of GLT-1 in the infected brain. PLoS Pathog 2025; 21:e1012733. [PMID: 40523037 DOI: 10.1371/journal.ppat.1012733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 06/25/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025] Open
Abstract
Toxoplasma gondii (T. gondii), a prolific protozoan parasite, forms cysts within neurons of the central nervous system that maintain infection for the lifetime of the host. Astrocytes are fundamental to neuronal health by providing nutrients and structural support and help regulate neurotransmitters by continuous communication with neurons. It is not yet known how infection and the presence of intracellular cysts, disrupts the crucial relationship between these cells. Extracellular vesicles (EVs) function in intracellular communication and can contain proteins, lipids, DNA, miRNA, and other RNA subtypes. EVs are produced by all cells and play an important role in neuronal-astrocyte interactions, including the regulation of glutamate receptors on astrocytes. Previous work has demonstrated that Toxoplasma infection reduces astrocytic expression of the primary glutamate transporter, GLT-1. Here we tested if cyst infection of neurons alters the production and content of EVs. EVs were isolated from uninfected and infected primary murine cortical neurons and their size, concentration, and characterization were confirmed with nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), ELISA, western blot, liquid chromatography (LC)-mass spectrometry (MS)/MS, and microRNA sequencing. Analysis reveals that infection of neurons reduced neuronal production of EVs and altered their protein and miRNA content. In addition to changes in host protein content, EVs from infected neurons contained the Toxoplasma proteins GRA1, GRA2, GRA7, MAG1 and MAG2. Following incubation of neuronal EVs with primary astrocytes, GRA7 protein could be observed within intracellular EVs and the nuclei of GRA7 + EV-containing cells. EVs from infected neurons altered gene expression of astrocytes resulting in an increase in pro-inflammatory transcriptional signatures, along with a downregulation of GLT-1 protein expression with similar transcriptional changes found in astrocytes in vivo. These results demonstrate the ability of a parasitic infection in the brain to alter EV production and the fundamental communication between neurons and astrocytes.
Collapse
Affiliation(s)
- Emily Z Tabaie
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Ziting Gao
- Department of Chemistry, University of California, Riverside, California, United States of America
| | - Nala Kachour
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Stacey Gomez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Zoe A Figueroa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Kristina V Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| | - Wenwan Zhong
- Department of Chemistry, University of California, Riverside, California, United States of America
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, United States of America
| |
Collapse
|
26
|
Zhu Y, Liu Y, Yang K, Wu W, Cheng Y, Ding Y, Gu R, Liu H, Zhang X, Liu Y. Apoptotic vesicles inhibit bone marrow adiposity via wnt/β-catenin signaling. Regen Ther 2025; 29:262-270. [PMID: 40230357 PMCID: PMC11994938 DOI: 10.1016/j.reth.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/14/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background There is currently increasing focus on aging-related diseases. Osteoporosis is a common disease the incidence of which increases with age. In older patients with osteoporosis, bone marrow mesenchymal stem cells (BMMSCs) have a decreased capacity for osteogenesis and an increased capacity for adipogenesis, causing excessive accumulation of adipose tissue in the bone marrow. Therefore, means of reducing bone marrow adiposity may have therapeutic potential for osteoporosis. Apoptotic vesicles (apoVs) participate in a wide range of physiological processes and have been shown to have therapeutic effects in a variety of diseases. The principal objective of this study was to examine the special properties and regulatory mechanisms of BMMSC-derived apoVs in the treatment of bone marrow adiposity. Results The results showed that apoVs could decrease bone marrow adiposity in osteoporotic mice and prevent adipogenic differentiation of MSCs by activating the Wnt/β-catenin pathway. Conclusion New apoV-based therapies have potential for the treatment of bone marrow adiposity in patients with aging-related osteoporosis and may be further applicable to the treatment of obesity and aging-related diseases.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- Department of Stomatology, Peking University Third Hospital, Beijing 100191, China
| | - Yaoshan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Kunkun Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Weiliang Wu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Yawen Cheng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Yanan Ding
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
27
|
Soni N, Rameshwari R. "Silent messengers of chaos: unveiling the dual threat of immune infiltrates in Japanese encephalitis virus neuroinflammatory storm". Virol J 2025; 22:173. [PMID: 40450327 DOI: 10.1186/s12985-025-02805-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 05/22/2025] [Indexed: 06/03/2025] Open
Affiliation(s)
- Naina Soni
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India.
| | - Rashmi Rameshwari
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| |
Collapse
|
28
|
Gan L, Guo X, Dong S, Sun C. The biology of exosomes and exosomal non-coding RNAs in cardiovascular diseases. Front Pharmacol 2025; 16:1529375. [PMID: 40492132 PMCID: PMC12147041 DOI: 10.3389/fphar.2025.1529375] [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: 11/16/2024] [Accepted: 04/07/2025] [Indexed: 06/11/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide, both in developed and developing countries. Despite the implementation of various measures in clinical practice that have shown certain curative effects, poor prognosis and irreversible pathological cardiac remodeling continue to limit the therapeutic effect of CVDs. There are still many new mechanisms worth exploring for the regulation of CVDs. Previous studies have highlighted the potential applicability of exosomes in CVDs, and significant research has been conducted in this area. In this review, we summarize the physiological mechanisms of exosomes and the basic research achievements in regulating CVDs via exosomal non-coding RNAs. We also discuss the limitations and prospects of exosome application in CVD treatment.
Collapse
Affiliation(s)
- Lu Gan
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofei Guo
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shichao Dong
- Department of Pharmacy, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chuan Sun
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
29
|
Soltanmohammadi F, Mahmoudi Gharehbaba A, Javadzadeh Y. Synergistic strategies in tissue engineering: The role of exosomes and decellularized extracellular matrix hydrogels. Biomed Pharmacother 2025; 188:118200. [PMID: 40414001 DOI: 10.1016/j.biopha.2025.118200] [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: 03/12/2025] [Revised: 05/07/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Tissue engineering aims to mimic the natural microenvironment of biological structures by utilizing the distinctive characteristics of extracellular matrix (ECM) scaffolds. The combination of decellularized extracellular matrix hydrogels (dECMHs) with exosomes (EXs) represents an innovative therapeutic approach for tissue regeneration. These dECMHs, sourced from diverse tissues, provide biocompatible scaffolds that conform to irregular defect geometries, thereby addressing the limitations of conventional ECM scaffolds. EXs, which are nanovesicles secreted by virtually all cells, play crucial role in cell communication and tissue regeneration. However, their short half-life presents challenges for systemic administration. The incorporation of EXs into dECMHs enables localized and prolonged release, thereby enhancing their therapeutic merits. This review thoroughly explains the techniques for decellularization, the characteristics of dECM, as well as the preparation and applications of dECMHs in tissue engineering. It also explores the synergistic effects of EX-dECMH systems on cellular activities essential for tissue repair, including proliferation, differentiation, and neovascularization. The mechanisms of EX release from dECMHs and their applications in the regeneration of skin, intervertebral disc, cartilage, and nerve tissues are elucidated, highlighting the considerable potential of this integrated strategy to improve tissue engineering techniques. Furthermore, the synergistic effect of EX-dECMH systems in tissue healing is investigated. Finally, the limitations associated with the clinical application of EX, dECM, and dECMH as well as the future prospect are included.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Grossini E, Ola Pour MM, Venkatesan S. The Role of Extracellular Vesicles in the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease and Other Liver Diseases. Int J Mol Sci 2025; 26:5033. [PMID: 40507843 PMCID: PMC12154092 DOI: 10.3390/ijms26115033] [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: 03/25/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
The increasing prevalence of liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD), presents considerable medical challenges, particularly given the absence of approved pharmacological treatments, which underscores the necessity to comprehend its underlying mechanisms. Extracellular vesicles (EVs), which are tiny particles released by cells, play a crucial role in facilitating communication and can transport harmful molecules that promote inflammation and tissue damage. These EVs are involved in the progression of various types of liver disorders since they aggravate inflammation and oxidative stress. Because of their critical role, it is believed that EVs are widely involved in the initiation and progression of MASLD, as well as in viral hepatitis, alcoholic liver disease, drug-induced liver injury, and hepatocellular carcinoma. This review emphasizes recent findings regarding the functions of EVs in the above liver pathologies and underscores their potential as new therapeutic targets, paving the way for innovative approaches to address those detrimental liver conditions.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (M.M.O.P.); (S.V.)
| | | | | |
Collapse
|
31
|
Liu N, Ma Y, Gong W, Shao X, Shi T, Li L, Wu W, Chen X, Shi Y, Zhang P, Lin J, Wang C, Fang D, Yang L, Wang P, Gao W, He Y, An X, Du R, Chen Y, Liu B, Qin J, Chen D, Cai P, Jiang Q, Guo B. Osteocyte-derived extracellular vesicles mediate the bone-to-cartilage crosstalk and promote osteoarthritis progression. Nat Commun 2025; 16:4746. [PMID: 40399261 PMCID: PMC12095588 DOI: 10.1038/s41467-025-59861-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025] Open
Abstract
Osteoarthritis is a common degenerative joint disease, in which mechanical overloading disrupts subchondral bone remodeling before cartilage degeneration and the osteocytes in the subchondral bone are mainly responsible for mechanosensing. However, their functional role in the early osteoarthritis is still unclear. Here we show that mechanical stress induces osteocytes in subchondral bone to secrete extracellular vesicles that accelerate cartilage metabolic dysregulation in patients with both sexes and male mice. The miR-23b-3p in extracellular vesicles promotes cartilage catabolism and inhibits anabolism by targeting OTUD4, disrupting mitophagy in chondrocytes. Inhibiting miR-23b-3p in osteocytes or chondrocytes reduces cartilage degeneration and osteoarthritis progression in male mice. Together, our findings highlight that osteocyte-derived extracellular vesicles mediate communication with chondrocytes and suggest miR-23b-3p as a potential therapeutic target for osteoarthritis.
Collapse
Affiliation(s)
- Na Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yuze Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wang Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xiaoyan Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wenshu Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yong Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Jiaquan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Chengzhi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Depeng Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Lin Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Wentian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Yi He
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Xueying An
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Rui Du
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Xuzhou Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Ying Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Bin Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Jianghui Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Dongyang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China
| | - Pingqiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, PR China.
| |
Collapse
|
32
|
Gao L, Zou H, Xie G, Li X, Chen Z. Phosphorylation of syntenin-1 by TBK1 promotes proliferation and migration of non-small cell lung cancer cells. J Biol Chem 2025:110278. [PMID: 40412527 DOI: 10.1016/j.jbc.2025.110278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
Syntenin-1 is a promising therapeutic target for cancer, as its inhibitors have shown positive efficacy in preclinical models of various cancer types. Posttranslational modifications including phosphorylation play an important role in regulating syntenin-1 activity, but the underlying molecular mechanisms have not been completely understood. To figure out the enzymes that catalyze syntenin-1 modifications, we performed mass spectrometry proteomics analysis of immunoprecipitated syntenin-1 and identified TANK-binding kinase 1 (TBK1) as a binding partner. Using biochemical and cellular assays, we demonstrated that TBK1 directly interacted with syntenin-1 and phosphorylated it at residue S6. ULK1, the reported kinase to catalyze syntenin-1 S6 phosphorylation, was shown in our assays to indirectly trigger syntenin-1 phosphorylation by activating TBK1. We also found that syntenin-1 was upregulated in non-small cell lung cancer (NSCLC) cells and TBK1-catalyzed syntenin-1 phosphorylation promoted cell growth and metastasis of the NSCLC cell line A549. Transcriptome sequencing revealed that syntenin-1 phosphorylation by TBK1 activated the MAPK signaling pathway. Our study illuminated a new mechanism that syntenin-1 phosphorylation regulated by upstream TBK1 signaling controls NSCLC progression.
Collapse
Affiliation(s)
- Lin Gao
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Hecun Zou
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Guojiao Xie
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xinning Li
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Zan Chen
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Asgari R, Rashidi S, Soleymani B, Bakhtiari M, Mohammadi P, Yarani R, Mansouri K. The supportive role of stem cells-derived exosomes in the embryo implantation process by regulating oxidative stress. Biomed Pharmacother 2025; 188:118171. [PMID: 40412359 DOI: 10.1016/j.biopha.2025.118171] [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: 03/09/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/27/2025] Open
Abstract
Oxidative stress can affect many aspects of the reproduction process. The embryo implantation process is also one of the critical steps in establishing a successful pregnancy, and several factors, including oxidative stress, can impact the process. Oxidative stress is a state of imbalance between pro-oxidant molecules such as reactive oxygen species (ROS) and antioxidant defenses. Excessive levels of ROS cause damage to the cellular macromolecules such as nucleic acids, proteins, and lipids, resulting in cell dysfunction and pathological conditions. Recently, studies have displayed the therapeutic and antioxidant properties of exosomes derived from stem cells. Exosomes are one type of extracellular vesicles (EVs) secreted by almost all cells and contain different biomolecules. The unique properties of exosomes, like regulation of biological processes, transportation of biomolecules, stability, and biodegradability, can make exosomes a promising therapeutic option in reproductive disorders and diseases. Exosomes also can significantly improve the curative effect of oxidative stress-related pathogenesis. Accordingly, this review aims to provide a novel overview of how exosomes derived from stem cells can regulate oxidative stress and support the process of embryo implantation, hoping to pave the way to clinical applications and future research in this field.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sahar Rashidi
- Department of Obstetrics and Gynecology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Bijan Soleymani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mitra Bakhtiari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Kamran Mansouri
- Regenerative Medicine Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
34
|
Liu Q, Liu M, Lu W, Li H, Ma Z, Xiong J, Zhang P. Surface-enhanced confocal Raman microscopy to characterize esophageal cancer cell-derived extracellular vesicles and maternal cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:4167-4175. [PMID: 40353506 DOI: 10.1039/d4ay02300e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Exosomes, a subtype of extracellular vesicles, are increasingly recognized as promising biomarkers for human cancers. Rapid detection and classification of esophageal cancer-associated exosomes could significantly improve non-invasive screening for potential patients. This study aims to establish a label-free, direct surface-enhanced Raman scattering (SERS) method to capture characteristic molecular information from both esophageal cancer cells and their corresponding exosomes using confocal Raman microscopy. The results revealed distinct Raman spectra for esophageal cancer cells and their exosomes within the range of 500-1600 cm-1, with notable signal similarities observed at 506-622, 778-832, 1079-1098, and 1572-1630 cm-1. In contrast, significant differences were identified in Raman peaks related to nucleic acids (723, 654, 1354 cm-1) and proteins (998, 1028, 1354, 1560 cm-1). An orthogonal partial least squares discriminant analysis (OPLS-DA) model was utilized to discern subtle variations among these highly similar samples, achieving an accuracy rate of 100%. By comparing the spectral correlations between esophageal cancer cells and their exosomes, this study provides valuable insights into the molecular composition and cellular origins of exosomes. The findings demonstrate the potential of integrating SERS with OPLS-DA for the precise and rapid detection and monitoring of esophageal cancer through exosomal biomarkers, offering a powerful tool for diagnostic applications.
Collapse
Affiliation(s)
- Qianjin Liu
- College of Future Technology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China.
| | - Mengdong Liu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| | - Wenjing Lu
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| | - Han Li
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| | - Zixuan Ma
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| | - Jingwei Xiong
- College of Future Technology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China.
| | - Ping Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
35
|
Wang D, Wang C, Hu Z, Su Y, Jia Z. A preliminary proteomic analysis of tear fluid in patients with high myopia. BMC Ophthalmol 2025; 25:302. [PMID: 40399820 PMCID: PMC12093765 DOI: 10.1186/s12886-025-04129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Using liquid chromatography-tandem mass spectrometry (LC-MS/MS) as a protein quantification technique for the analysis of the proteomic profile of myopia patient tear fluid, to clarify the role of dysregulated proteins in high myopia (HM) in order to provide a more thorough understanding of the molecular processes involved in the development of the disease. METHODS Schirmer strips were used to acquire the tear films from 20 subjects (10 high myopia patients and 10 control subjects). LC-MS/MS was utilized to identify the proteome profile of the tears in order to assess protein interrelationships utilizing bioinformatics. RESULTS The tear preparations from the HM group and the control group included a total of 1544 proteins. The expression of 79 proteins out of the identified ones differed significantly between the two groups. 51 proteins showed overexpression and 28 proteins showed downregulation. 15 differentially expressed proteins (DEPs) were enriched in metabolic pathways, 15 DEPs were enriched in extracellular exomes, and 5 DEPs were enriched in the complement and coagulation cascades pathway. Potentially important proteins and therapeutic targets in human HM include TTR and Antithrombin-III. CONCLUSION The proteomic analysis of tear fluid in high myopia patients identifies key proteins and pathways involved in the disease, offering potential biomarkers for its pathogenesis and therapeutic targets.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Ophthalmology, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
- Department of Ophthalmology, The fourth hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Cunkai Wang
- Department of Gastroenterology, Hebei General Hospital, Shijiazhuang, Hebei Province, 050000, China
| | - Zhi Hu
- Department of Ophthalmology, Beijing Puren Hospital, Beijing, 100010, China
| | - Yudan Su
- Department of Ophthalmology, The fourth hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Zhiyang Jia
- Department of Ophthalmology, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China.
- Department of Ophthalmology, Hebei General Hospital, Shijiazhuang, Hebei Province, 050000, China.
| |
Collapse
|
36
|
Perkins DO, Jeffries CD, Clark SR, Upthegrove R, Wannan CMJ, Wray NR, Li QS, Do KQ, Walker E, Paul Amminger G, Anticevic A, Cotter D, Ellman LM, Mongan D, Phassouliotis C, Barbee J, Roth S, Billah T, Corcoran C, Calkins ME, Cerrato F, Khadimallah I, Klauser P, Winter-van Rossum I, Nunez AR, Bleggi RS, Martin AR, Bouix S, Pasternak O, Shah JL, Toben C, Wolf DH, Accelerating Medicines Partnership® Schizophrenia (AMP® SCZ), Kahn RS, Kane JM, McGorry PD, Bearden CE, Nelson B, Shenton ME, Woods SW. Body fluid biomarkers and psychosis risk in The Accelerating Medicines Partnership® Schizophrenia Program: design considerations. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2025; 11:78. [PMID: 40399418 PMCID: PMC12095529 DOI: 10.1038/s41537-025-00610-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/11/2025] [Indexed: 05/23/2025]
Abstract
Advances in proteomic assay methodologies and genomics have significantly improved our understanding of the blood proteome. Schizophrenia and psychosis risk are linked to polygenic scores for schizophrenia and other mental disorders, as well as to altered blood and saliva levels of biomarkers involved in hormonal signaling, redox balance, and chronic systemic inflammation. The Accelerating Medicines Partnership® Schizophrenia (AMP®SCZ) aims to ascertain biomarkers that both predict clinical outcomes and provide insights into the biological processes driving clinical outcomes in persons meeting CHR criteria. AMP®SCZ will follow almost 2000 CHR and 640 community study participants for two years, assessing biomarkers at baseline and two-month follow-up including the collection of blood and saliva samples. The following provides the rationale and methods for plans to utilize polygenic risk scores for schizophrenia and other disorders, salivary cortisol levels, and a discovery-based proteomic platform for plasma analyses. We also provide details about the standardized methods used to collect and store these biological samples, as well as the study participant metadata and quality control measures related to preanalytical factors that could influence the values of the biomarkers. Finally, we discuss our plans for analyzing the results of blood- and saliva-based biomarkers. Watch Dr. Perkins discuss their work and this article: https://vimeo.com/1062879582?share=copy#t=0 .
Collapse
Affiliation(s)
- Diana O Perkins
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Clark D Jeffries
- Rennaisance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott R Clark
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Woodville, SA, Australia
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Birmingham, UK
- Birmingham Womens and Childrens, NHS Foundation Trust, Birmingham, UK
| | - Cassandra M J Wannan
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Naomi R Wray
- Department of Psychiatry, University of Oxford, Oxford, UK
- Institute for Molecular Biosciences, University of Queensland, Queensland, Australia
| | - Qingqin S Li
- JRD Data Science, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Kim Q Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience King's College London, London, UK
| | - Elaine Walker
- Departments of Psychology and Psychiatry, Emory University, Atlanta, GA, United States of America
| | - G Paul Amminger
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - David Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Lauren M Ellman
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Christina Phassouliotis
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Jenna Barbee
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharin Roth
- Genomics and Biomarker Research, Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD, USA
| | - Tashrif Billah
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Cheryl Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ines Khadimallah
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience King's College London, London, UK
| | - Paul Klauser
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, Service of Child and Adolescent Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Angela R Nunez
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Connecticut Mental Health Center, New Haven, CT, USA
| | - Rachel S Bleggi
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alicia R Martin
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Sylvain Bouix
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Jai L Shah
- Douglas Research Centre, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Catherine Toben
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Daniel H Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John M Kane
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hempstead, N.Y, USA
- Institute for Behavioral Science, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Patrick D McGorry
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Carrie E Bearden
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Barnaby Nelson
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Martha E Shenton
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Scott W Woods
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
37
|
Zhang Y, Wang C, Shao H. Nanoplasmonic Sensing of Heterogeneous Extracellular Vesicles: From Bulk to Single Vesicles. SMALL METHODS 2025:e2500097. [PMID: 40391615 DOI: 10.1002/smtd.202500097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoscale membrane vesicles released by almost all cell types into the circulation. Depending on their biogenesis and cells of origin, EVs show considerable heterogeneity in their biophysical and biomolecular composition and can serve as reflective and dynamic blood biomarkers for personalized medicine. Conventional analytical technologies, however, often lack the compatibility to reveal nanoscale EV features and resolve vesicle heterogeneity. The past decade has since witnessed the development of various nanoplasmonic technologies to empower EV analysis, through bulk and single-vesicle characterization, at an unprecedented scale and resolution. These platforms achieve versatile measurements that are not only size-matched to EV dimensions but can also probe multiplexed biomolecular contents, thereby providing new insights into EV heterogeneity and enabling transformative clinical opportunities. In this review, key characteristics of EVs and their remarkable heterogeneity are introduced. The sensing principles of plasmonic platforms are also discussed, with recent technology developments highlighted to resolve EV heterogeneity, through bulk analyses of EV subpopulations as well as high-resolution single-EV measurements. An outlook is further provided on emerging opportunities, at the interface of biomarker discovery and technology innovation, to develop empowering nanoplasmonic EV platforms for personalized medicine. biosensing; bulk analysis; extracellular vesicles; nanoplasmonics; single-vesicle analysis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chao Wang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 138673, Singapore
| |
Collapse
|
38
|
Shah KA, Ali T, Hussain Y, Dormocara A, You B, Cui JH. Isolation, characterization and therapeutic potentials of exosomes in lung cancer: Opportunities and challenges. Biochem Biophys Res Commun 2025; 759:151707. [PMID: 40153996 DOI: 10.1016/j.bbrc.2025.151707] [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: 12/06/2024] [Revised: 02/08/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Lung cancer (LC) signifies the primary cause of cancer-related mortality, representing 24 % of all cancer fatalities. LC is intricate and necessitates innovative approaches for early detection, precise diagnosis, and tailored treatment. Exosomes (EXOs), a subclass of extracellular vesicles (EVs), are integral to LC advancement, intercellular communication, tumor spread, and resistance to anticancer therapies. EXOs represent a viable drug delivery strategy owing to their distinctive biological characteristics, such as natural origin, biocompatibility, stability in blood circulation, minimal immunogenicity, and potential for modification. They can function as vehicles for targeted pharmaceuticals and facilitate the advancement of targeted therapeutics. EXOs are pivotal in the metastatic cascade, facilitating communication between cancer cells and augmenting their invasive capacity. Nonetheless, obstacles such as enhancing cargo loading efficiency, addressing homogeneity concerns during preparation, and facilitating large-scale clinical translation persist. Interdisciplinary collaboration in research is crucial for enhancing the efficacy of EXOs drug delivery systems. This review explores the role of EXOs in LC, their potential as therapeutic agents, and challenges in their development, aiming to advance targeted treatments. Future research should concentrate on engineering optimization and developing innovative EXOs to improve flexibility and effectiveness in clinical applications.
Collapse
Affiliation(s)
- Kiramat Ali Shah
- College of Pharmaceutical Science, Soochow University, Renai Road 199, SIP, 215213, Suzhou, Jiangsu, China
| | - Tariq Ali
- Department of Civil and Environmental Engineering, Shantou University, Shantou, Guangdong, 515063, China
| | - Yaseen Hussain
- College of Pharmaceutical Science, Soochow University, Renai Road 199, SIP, 215213, Suzhou, Jiangsu, China
| | - Amos Dormocara
- College of Pharmaceutical Science, Soochow University, Renai Road 199, SIP, 215213, Suzhou, Jiangsu, China
| | - Bengang You
- College of Pharmaceutical Science, Soochow University, Renai Road 199, SIP, 215213, Suzhou, Jiangsu, China
| | - Jing-Hao Cui
- College of Pharmaceutical Science, Soochow University, Renai Road 199, SIP, 215213, Suzhou, Jiangsu, China.
| |
Collapse
|
39
|
Longo A, Manganelli V, Misasi R, Riitano G, Caglar TR, Fasciolo E, Recalchi S, Sorice M, Garofalo T. Extracellular Vesicles in the Crosstalk of Autophagy and Apoptosis: A Role for Lipid Rafts. Cells 2025; 14:749. [PMID: 40422252 DOI: 10.3390/cells14100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/13/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Autophagy and apoptosis are two essential mechanisms regulating cell fate. Although distinct, their signaling pathways are closely interconnected through various crosstalk mechanisms. Lipid rafts are described to act as both physical and functional platforms during the early stages of autophagic and apoptotic processes. Only recently has a role for lipid raft-associated molecules in regulating EV biogenesis and release begun to emerge. In particular, lipids of EV membranes are essential components in conferring stability to these vesicles in different extracellular environments and/or to facilitate binding or uptake into recipient cells. In this review we highlight these aspects, focusing on the role of lipid molecules during apoptosis and secretory autophagy pathways. We describe the molecular machinery that connects autophagy and apoptosis with vesicular trafficking and lipid metabolism during the release of EVs, and how their alterations contribute to the development of various diseases, including autoimmune disorders and cancer. Overall, these findings emphasize the complexity of autophagy/apoptosis crosstalk and its key role in cellular dynamics, supporting the role of lipid rafts as new therapeutic targets.
Collapse
Affiliation(s)
- Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tuba Rana Caglar
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Elena Fasciolo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Serena Recalchi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
40
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Ballal S, Pathak PK, Bareja L, Aminov Z, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomal non-coding RNAs: key regulators of inflammation-related cardiovascular disorders. Eur J Med Res 2025; 30:395. [PMID: 40390035 PMCID: PMC12087048 DOI: 10.1186/s40001-025-02649-6] [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: 02/02/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Inflammation is a complex, tightly regulated process involving biochemical and cellular reactions to harmful stimuli. Often termed "the internal fire", it is crucial for protecting the body and facilitating tissue healing. While inflammation is essential for survival, chronic inflammation can be detrimental, leading to tissue damage and reduced survival. The innate immune system triggers inflammation, closely linked to the development of heart diseases, with significant consequences for individuals. Inflammation in arterial walls or the body substantially contributes to atherosclerotic disease progression, affecting the cardiovascular system. Altered lipoproteins increase the risk of excessive blood clotting, a hallmark of atherosclerotic cardiovascular disease and its complications. Integrating inflammatory biomarkers with established risk assessment techniques can enhance our ability to identify at-risk individuals, assess their risk severity, and recommend appropriate CVD prevention strategies. Exosomes, a type of extracellular vesicle, are released by various cells and mediate cell communication locally and systemically. In the past decade, exosomes have been increasingly studied for their vital roles in health maintenance and disease processes. They can transport substances like non-coding RNAs, lipids, and proteins between cells, influencing immune responses and inflammation to elicit harmful or healing effects. This study focuses on the critical role of inflammation in heart disease progression and how non-coding RNAs in exosomes modulate the inflammatory process, either exacerbating or alleviating inflammation-related damage in the cardiovascular system.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
41
|
Tian L, Wang Z, Chen S, Guo K, Hao Y, Ma L, Ma K, Chen J, Liu X, Li L, Fu X, Zhang C. Ellagic Acid-Loaded sEVs Encapsulated in GelMA Hydrogel Accelerate Diabetic Wound Healing by Activating EGFR on Skin Repair Cells. Cell Prolif 2025:e70064. [PMID: 40384373 DOI: 10.1111/cpr.70064] [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: 03/04/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
Delayed diabetic wound healing is partially attributed to the functional disorder of skin repair cells caused by high glucose (HG). Small extracellular vehicles (sEVs) loaded with small-molecule drugs represent a highly promising therapeutic strategy. This study aims to evaluate the therapeutic efficacy of ellagic acid-encapsulated small extracellular vesicles (EA-sEVs) in diabetic wound regeneration and to unravel related mechanisms. Cytotoxicity tests of ellagic acid (EA) as liposomal small molecules (LSMs) were performed with the CCK8 assay. EA was incorporated into sEVs obtained from chorionic plate-mesenchymal stem cells (CP-MSCs) to construct EA-engineered sEVs. The protective effects of EA-sEVs on human dermal fibroblasts (HDFs) and human epidermal keratinocytes (HEKs) induced by high glucose (HG) were assessed through the evaluation of their proliferative, migrative and differentiative capabilities. Furthermore, to illustrate the underlying mechanism, the specific biological targets of EA were predicted and confirmed. Finally, EA-sEVs were encapsulated in GelMA hydrogel for investigating the pro-healing effects on diabetic wounds. EA was harmless to cell viability, increasing the possibility and safety of drug development. EA-engineered sEVs were fabricated by loading EA in sEVs. In vitro, EA-sEVs promoted the proliferation, migration, and transdifferentiation of HG-HDFs and the proliferation and migration of HG-HEKs. Mechanism analysis elucidated that epidermal growth factor receptor (EGFR) was the specific biological target of EA. EA interacting with EGFR was responsible for the functional improvement of HG-HDFs and HG-HEKs. In vivo, EA-sEVs encapsulated in GelMA promoted the healing of diabetic wounds by improving re-epithelialisation, collagen formation and the expression of EGFR. Gel-EA-sEVs promoted diabetic wound healing by improving biological functions of HDFs and HEKs. EGFR was first identified as the specific biological target of EA and was responsible for the functional improvement of HG-HDFs and HG-HEKs by Gel-EA-sEVs. Hence, Gel-EA-sEVs can serve as a new promising active dressing for diabetic wound treatment.
Collapse
Affiliation(s)
- Lige Tian
- College of Graduate, Tianjin Medical University, Tianjin, China
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
| | - Zihao Wang
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Shengqiu Chen
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- Innovation Research Center for Diabetic Foot, West China Hospital, Sichuan University, Chengdu, China
| | - Kailu Guo
- College of Graduate, Tianjin Medical University, Tianjin, China
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
| | - Yaying Hao
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Liqian Ma
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Kui Ma
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Junli Chen
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Xi Liu
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Linlin Li
- Beijing Key Laboratory of Micro-Nano Energy and Sensor, Center for High-Entropy Energy and Systems, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, China
| | - Xiaobing Fu
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| | - Cuiping Zhang
- Medical Innovation Research Department, PLA General Hospital, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine, Beijing, China
| |
Collapse
|
42
|
Zhang L, Zhan M, Sun H, Zou Y, Laurent R, Mignani S, Majoral JP, Cao X, Shen M, Shi X. Mesenchymal Stem-Cell-Derived Exosomes Loaded with Phosphorus Dendrimers and Quercetin Treat Parkinson's Disease by Modulating Inflammatory Immune Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40388599 DOI: 10.1021/acsami.5c05809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The intricate pathologic features of Parkinson's disease (PD) coupled with the obstacle posed by the blood-brain barrier (BBB) significantly limit the efficacy of most medications, leading to difficulties in PD treatments. Herein, we have developed a nanomedicine based on stem-cell-derived exosomes coloaded with hydroxyl-terminated phosphorus dendrimers (AK76) and quercetin (Que) for combined therapeutic intervention of PD. The engineered nanocomplexes (for short, QAE NPs) exhibit an optimal size of 269.7 nm, favorable drug release profile, and desired cytocompatibility, enabling penetration of the nasal mucosa to accumulate in the brain without BBB crossing. The developed QAE NPs can scavenge reactive oxygen species, promote M2 microglial polarization, attenuate inflammation, and protect neurons by inducing autophagy and restoring mitochondrial homeostasis through the integrated anti-inflammatory and antioxidant properties of exosomes, Que and AK76, collectively leading to improved motor functions, coordination, and alleviation of depression-like symptoms in PD mice. The formulated QAE NPs combined with several therapeutic components are able to simultaneously modulate both microglia and neurons, offering promising potential for the treatment of PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mengsi Zhan
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Huxiao Sun
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yu Zou
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Regis Laurent
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
- Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
| | - Serge Mignani
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
- Centre d'Etudes et de Recherche sur le Medicament de Normandie (CERMN), Université de Caen Normandie, Caen 14032, France
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
- Université Toulouse, 118 Route de Narbonne, CEDEX 4, 31077 Toulouse, France
| | - Xueyan Cao
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mingwu Shen
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory of Advanced Fiber Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
43
|
Ji R, Wang H, Zheng X, Shi D, Tian W, Gao P, Li Y, Wen Y, Wang J, Liu Z, Wong CCL, Chen Y. Tetraspanin 4 Mediates Cholesterol-Dependent Exosome Membrane Protection from Cryodamage. NANO LETTERS 2025. [PMID: 40387525 DOI: 10.1021/acs.nanolett.5c00572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Exosomes, nanosized extracellular vesicles carrying proteins, lipids, and nucleic acids, hold great potential in therapeutic applications. Cryopreservation, a widely used method for their preservation and transport, often causes irreversible damage. Understanding the molecular mechanisms underlying biomembrane resistance to cryodamage is crucial for advancing cryopreservation techniques. In this study, we find that tetraspanin 4 (TSPAN4) and other tetraspanin family proteins play an essential role in protecting exosomes from cryodamage, likely due to their role in cholesterol binding and membrane microdomain formation. Furthermore, we engineered TSPAN4-loaded exosomes, which demonstrated enhanced cryoprotection while maintaining a similar protein composition and uptake efficiency compared to wild-type exosomes. Our novel cryopreservation strategy, which does not rely on external agents, offers a promising approach for advancing the clinical translation of exosomes as therapeutic agents.
Collapse
Affiliation(s)
- Rui Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
- Peking University First Hospital, Beijing 100034, P. R. China
| | - Hongli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Xia Zheng
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Dongxue Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Wenmin Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Peizhen Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Yong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Yiling Wen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| | - Jianjun Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zhang Liu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, P. R. China
| | - Yang Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
- Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
44
|
Ghoshal B, Patkulkar PA, Bhatt P, Rana S, Sinharay S, Jhunjhunwala S. Neutrophil-Derived Extracellular Vesicles for Facile Delivery of Diagnostic Agents to Tumor Microenvironments. ACS APPLIED BIO MATERIALS 2025; 8:4132-4139. [PMID: 40276969 DOI: 10.1021/acsabm.5c00266] [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: 04/26/2025]
Abstract
Targeted delivery of diagnostic and therapeutic agents to tumor microenvironments using nanoparticles improves the efficacy of these agents and reduces their unwarranted side effects. Numerous synthetic nanoparticle systems have been designed for this very purpose, but few have translated clinically due to poor efficacy-to-cost and efficacy-to-toxicity ratios. Biological nanoparticles such as modified extracellular vesicles (EVs) that are likely to have lower toxicities have also been developed but face challenges in clinical translation as they have primarily been produced from cancerous cells/cell lines and have high batch-to-batch variability. To overcome these issues, herein, we demonstrate that EVs isolated from neutrophils may be loaded with a specific diagnostic agent (indocyanine green) in a facile manner within a few hours, that these agent-loaded EVs are retained in vivo in mouse tumors for longer with a ∼5-fold increase in retention when compared to free diagnostic agents and hence facilitate short-term longitudinal imaging of the tumor microenvironments. The use of an individual's own cell-derived EVs requiring minimal ex vivo manipulations increases the likelihood of translating such a system into the clinic.
Collapse
Affiliation(s)
- Bartika Ghoshal
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | | | - Preeti Bhatt
- Materials Research Centre, Indian Institute of Science, Bengaluru 560012, India
| | - Subinoy Rana
- Materials Research Centre, Indian Institute of Science, Bengaluru 560012, India
| | - Sanhita Sinharay
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | | |
Collapse
|
45
|
Islam MKB, Marcus RK. Effects of packing density and adsorption conditions on extracellular vesicle dynamic binding capacities for capillary-channeled polymer (C-CP) fiber columns. J Chromatogr A 2025; 1755:466068. [PMID: 40403650 DOI: 10.1016/j.chroma.2025.466068] [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: 03/12/2025] [Revised: 05/16/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
Extracellular vesicles (EVs) are membrane-bound nanoparticles (50-1000 nm) secreted by all cell types and play critical roles in various biological processes. Among these, exosomes, a smaller subset of EVs, have attracted considerable interest due to their potential applications in diagnostics and therapeutics. However, conventional EV isolation methods are often limited by inefficiencies in processing time, recovery, and scalability. Hydrophobic interaction chromatography utilizing capillary-channeled polymer (CCP) fiber stationary phases offers a promising alternative, enabling rapid (<15 min), cost-effective (∼$5 per column) EV isolation with high loading capacities (∼1010-10¹² particles) and minimal sample pre-processing. Despite these advantages, achieving high-throughput EV isolation for larger-scale applications using the CCP fiber platform is the present challenge. To this end, further optimization of stationary phase packing and adsorption conditions is necessary to maximize the available binding surface area in the current microbore column format. This study systematically investigates the influence of interstitial fraction (i.e. packing density) in polyester (PET) CCP fiber columns on the dynamic binding capacity (DBC) of EVs isolated from human urine using a high-performance liquid chromatography platform. Microbore columns (0.76 mm i.d. × 300 mm) packed with PET CCP fibers in both an eight-channel (PET-8) and a novel trilobal (PET-Y) configuration were evaluated using breakthrough curves and frontal analysis. The results reveal that lower packing densities correlate with higher mass- and surface area-based EV binding capacities, with a maximum DBCs of 2.86 × 10¹³ EVs g-1 fiber and 1.22 × 10¹⁴ EVs m⁻² fiber achieved in <2 min of sample loading. Under optimum conditions, surface utilization of >50 % is realized. These results establish a framework for optimizing CCP fiber-based platforms to enhance EV capture efficiency, facilitating the development of scalable EV isolation techniques for biomedical research and therapeutic applications.
Collapse
Affiliation(s)
- Md Khalid Bin Islam
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC 29634-0973, USA
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC 29634-0973, USA.
| |
Collapse
|
46
|
Frisk NLS, Jørgensen MM, Bæk R, Atic AI, Brodersen TR, Ostrowski SR, Larsen MH, Posselt D, Høgdall E, Høgdall C, Pedersen OBV, Dalgaard LT. Characterization of small extracellular vesicles from ovarian cancer patients and pre-diagnostic patient samples: Evidence from the Danish blood donor study. PLoS One 2025; 20:e0323529. [PMID: 40372993 PMCID: PMC12080785 DOI: 10.1371/journal.pone.0323529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/08/2025] [Indexed: 05/17/2025] Open
Abstract
AIM Ovarian cancer (OC) is the leading cause of gynecological cancer deaths. Current biomarkers of OC are not specific or sensitive enough. Extracellular vesicles (EVs), EV surface proteins and their cargo microRNA (miRNA) show potential as biomarkers. This study aimed to characterize the ability of EVs to identify early OC-biomarkers among blood donors six months before their diagnosis. METHODS Study groups of OC patients, benign tumor patients (B), healthy blood donors (Control), and blood donors with incident OC diagnosis within six months of the last blood draw (Pre-diagnostic; PD) were established. Small EVs were enriched from plasma using ultracentrifugation. EVs were characterized by Dynamic Light Scattering (DLS), EV Array, NanoFlow Cytometry, Nanoparticle Tracking Analysis, and Western blots. RNA from EVs was isolated. A discovery study was performed on OC and B patients using the TaqMan Array Human MicroRNA A card. A validation study of 9 specific miRNAs was performed using RT-qPCR. RESULTS With DLS, it was identified that the OC patients' EVs were more heterogeneous in size compared to the other groups. Western blot identified CD63 and TSG101 in the EV enrichments. EV Array assessed 22 known protein biomarkers. TaqMan MicroRNA Array cards indicated a differential miRNA abundance between OC and B; however, technical replication and validation could not validate this pattern. CONCLUSION This study has analyzed EVs in OC, B, Control, and PD women. More extensive investigations of EV CD9, CD151, and CD81 in conjunction with other risk factors and well-known biomarkers like CA125 or HE4 should be the main objectives of future research.
Collapse
Affiliation(s)
- Nanna Lond Skov Frisk
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Amila Iriskic Atic
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Novo Nordisk A/S, Måløv, Copenhagen, Denmark
| | | | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Dorthe Posselt
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Claus Høgdall
- Department of Gynaecology, Rigshospitalet, Copenhagen, Denmark
| | - Ole Birger Vesterager Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
47
|
Chen T, Chen D, Su W, Liang J, Liu X, Cai M. Extracellular vesicles as vital players in drug delivery: a focus on clinical disease treatment. Front Bioeng Biotechnol 2025; 13:1600227. [PMID: 40438295 PMCID: PMC12116468 DOI: 10.3389/fbioe.2025.1600227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
Extracellular vesicles (EVs), a diverse population of bilayer lipid-membrane vesicles secreted by cells, have emerged as ideal drug carriers due to their efficient cellular uptake and targeted delivery capabilities. Advancements in medical and bioengineering collaborations have enabled EVs to be engineered for specific marker expression or therapeutic cargo transport, positioning them as a promising modality for treating cancer, neurological disorders, cardiovascular diseases, and beyond. EV-based drug delivery strategies offer distinct advantages, including facilitation of intercellular communication and immune modulation, high biocompatibility and stability, the ability to traverse the blood-brain barrier, and potential synergistic interactions with encapsulated therapeutics to enhance efficacy. This review explores EV isolation and scalable production, emphasizing cost-effective and reproducible manufacturing strategies, cargo-loading methodologies, and therapeutic applications. Additionally, the current landscape of EV-based targeted drug delivery, clinical translation prospects, and prevailing challenges are examined to provide a comprehensive perspective on their potential in drug delivery systems.
Collapse
Affiliation(s)
| | | | | | | | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, hospital of Stomatology, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, China
| | - Mingxiang Cai
- The First Affiliated Hospital of Jinan University, hospital of Stomatology, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, China
| |
Collapse
|
48
|
Li L, Zheng Z, Lan W, Tang N, Zhang D, Ling J, Wu Y, Yang P, Fu L, Liu J, Zhang J, Yu P, Huang T. Role of Exosomes in Cardiovascular Disease: A Key Regulator of Intercellular Communication in Cardiomyocytes. ACS OMEGA 2025; 10:18145-18169. [PMID: 40385188 PMCID: PMC12079207 DOI: 10.1021/acsomega.4c11423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/27/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
In the cardiovascular system, different types of cardiovascular cells can secrete specific exosomes and participate in the maintenance of cardiovascular function and the occurrence and development of diseases. Exosomes carry biologically active substances such as proteins and nucleic acids from cells of origin and can be used as biomarkers for disease diagnosis and prognosis assessment. In addition, exosome-mediated intercellular communication plays a key role in the occurrence and development of cardiovascular diseases and has become a potential therapeutic target. This article emphasizes the importance of understanding the mechanism of exosomes in cardiovascular diseases and systematically details the current understanding of exosomes as regulators of intercellular communication in cardiomyocytes, providing a basis for future research and therapeutic intervention.
Collapse
Affiliation(s)
- Liuxin Li
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Zhidong Zheng
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Wenyu Lan
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Nan Tang
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Deju Zhang
- Food
and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 0000, Hong Kong
| | - Jitao Ling
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Yuting Wu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Pingping Yang
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Linhua Fu
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jianping Liu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jing Zhang
- Department
of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical
College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Peng Yu
- Department
of Metabolism and Endocrinology, The Second
Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Tieqiu Huang
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| |
Collapse
|
49
|
Wu Q, Liu S, Zhao M, Wang Y, Lv K, Zhu J, Liu J. Pyroptosis-preconditioned mesenchymal stromal cell-derived extracellular vesicles as advanced nanomedicines for treating inflammatory diseases. Biomater Sci 2025; 13:2690-2703. [PMID: 40165652 DOI: 10.1039/d4bm01505c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Uncontrolled inflammation is one of the major causes of various forms of tissue injury, and nanomedicines with immunoregulatory effects are needed. Mesenchymal stromal cell-derived extracellular vesicles (e.g., MSC-EVs) have been proposed as promising therapies, but the highly efficient generation of EVs with desirable properties is still a considerable challenge in this field. Here, we report that preconditioning MSCs with a critical immune process (pyroptosis) is a robust method for improving both the yield and anti-inflammatory potency of MSC-EVs. In brief, pyroptosis-preconditioned MSCs using a combined lipopolysaccharide (LPS) and adenosine triphosphate (ATP) stimulation showed elevated EV yields compared with those of MSCs cultured under normal conditions. Pyroptosis preconditioning upregulated multiple pathways (e.g., cell proliferation, DNA repair, and the immune response) in MSCs, leading to the enrichment of immunoregulatory cargos (e.g., PD-L2 and STC2) in MSC-EVs. In vitro, pyroptosis-preconditioned MSC-EVs (P-EVs) treatment has greater potential to suppress cytokine expression and cell death in pyroptotic macrophages than treatment with normal MSC-EVs (N-EVs). Compared with N-EV treatment, P-EV treatment showed superior potency in attenuating proinflammatory cell infiltration, cytokine/chemokine expression, resident tissue cell death, and the severity of pathological injury in different models of inflammatory diseases (acute lung or kidney injury), and these effects are likely the joint result of diverse functional cargos delivered by such EVs. This study highlights that pyroptosis preconditioning is a promising strategy for the highly efficient production of MSC-EVs with advanced therapeutic potential for treating diverse inflammatory diseases.
Collapse
Affiliation(s)
- Qianyi Wu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuyun Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Meng Zhao
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yizhuo Wang
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Ke Lv
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| | - Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jingping Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, China.
| |
Collapse
|
50
|
Phan N, Li Y, Yang M, Liu F. Tear fluid derived extracellular vesicles for new biomarker discovery. Ocul Surf 2025; 37:314-322. [PMID: 40368029 DOI: 10.1016/j.jtos.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
Various cell types release extracellular vesicles (EVs) containing proteins, DNA, and RNA essential for intercellular communication. The bioactive molecules from EVs can reflect disease status and monitor progression, while their communication abilities suggest therapeutic potential. We will review various EV isolation methods, EV-enriched fluids, and studies analyzing differential mi-RNA and protein levels extracted from EVs. Specifically, tear-derived EVs, which protect their molecular content and allow for real-time monitoring of ocular conditions such as Dry Eye Disease (DED), Sjögren's disease (SJD), Ocular graft-versus-host disease (oGVHD), and Diabetic Retinopathy (DR), which all currently remain undiagnosed in patients. EVs also provide potential as carriers for gene transfer, and mesenchymal stem cell (MSCs)-derived EVs are shown to be immunomodulatory, demonstrating promise for autoimmune ocular diseases. Through the multi-omic analysis of tear-fluid content, EVs are promising biomarkers and therapeutic agents in ocular diseases.
Collapse
Affiliation(s)
- Natalie Phan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, 94720, USA
| | - Yi Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|