1
|
Wang C, Rong X, Zhang F, Mu X, Jiang J. Cryo-trojan mesenchymal stem cells as non-living tumor-homing supercarriers for enhanced drug delivery and immune activation in prostate cancer. Mater Today Bio 2025; 32:101650. [PMID: 40151804 PMCID: PMC11937679 DOI: 10.1016/j.mtbio.2025.101650] [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: 01/25/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background Prostate cancer remains a leading cause of cancer-related mortality, with conventional therapies limited by systemic toxicity and poor tumor targeting. Developing innovative drug delivery systems that enhance therapeutic specificity while minimizing off-target effects is critical. Materials and methods We engineered cryo-trojan human umbilical cord mesenchymal stem cells (CT-MSCs) as non-living, tumor-homing carriers for mitoxantrone (MTX), termed CT-MTX. Cryo-treatment preserved structural integrity and chemokine receptors (CXCR4/CCR2) for tumor targeting while eliminating proliferative risks. Comprehensive evaluations included drug loading/release kinetics, in vitro tumor suppression, immunogenic cell death (ICD) induction, and in vivo efficacy/safety in prostate cancer models. Results CT-MTX demonstrated superior drug loading (116.38 μg/106 cells) and pH-sensitive release (74.10 % at pH 5.5), outperforming exosomes, liposomes, and living MSCs in stability and tumor-specific drug delivery. Compared to liposomes (low targeting) and nanomaterials (biocompatibility concerns), CT-MTX leveraged MSC-derived tropism without tumorigenic risks. In vitro, CT-MTX inhibited tumor proliferation (84.83 % MTX uptake), migration (4.42 % residual migration), and induced apoptosis (43.23 % late apoptosis). Mechanistically, CT-MTX triggered ICD via PAMPs release, activating CD8+ T cells and suppressing immunosuppressive Treg. In vivo, CT-MTX selectively accumulated in tumors, reducing growth by 87.88 % and extending survival (93.30 % vs. 66.70 % in controls) with negligible systemic toxicity. Proteomics revealed enriched immune pathways like NK cell cytotoxicity, validating its dual role in direct tumor killing and immune activation. Conclusion CT-MTX represents a novel, non-proliferative drug delivery platform that combines the tumor-homing capacity of MSCs with enhanced safety and controlled release, inducing ICDs for prostate cancer and other immunologically "cold" tumors to improve immune infiltration.
Collapse
Affiliation(s)
| | | | - Fuqiang Zhang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| |
Collapse
|
2
|
Dipalo LL, Mikkelsen JG, Gijsbers R, Carlon MS. Trojan Horse-Like Vehicles for CRISPR-Cas Delivery: Engineering Extracellular Vesicles and Virus-Like Particles for Precision Gene Editing in Cystic Fibrosis. Hum Gene Ther 2025. [PMID: 40295092 DOI: 10.1089/hum.2024.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
The advent of genome editing has kindled the hope to cure previously uncurable, life-threatening genetic diseases. However, whether this promise can be ultimately fulfilled depends on how efficiently gene editing agents can be delivered to therapeutically relevant cells. Over time, viruses have evolved into sophisticated, versatile, and biocompatible nanomachines that can be engineered to shuttle payloads to specific cell types. Despite the advances in safety and selectivity, the long-term expression of gene editing agents sustained by viral vectors remains a cause for concern. Cell-derived vesicles (CDVs) are gaining traction as elegant alternatives. CDVs encompass extracellular vesicles (EVs), a diverse set of intrinsically biocompatible and low-immunogenic membranous nanoparticles, and virus-like particles (VLPs), bioparticles with virus-like scaffold and envelope structures, but devoid of genetic material. Both EVs and VLPs can efficiently deliver ribonucleoprotein cargo to the target cell cytoplasm, ensuring that the editing machinery is only transiently active in the cell and thereby increasing its safety. In this review, we explore the natural diversity of CDVs and their potential as delivery vectors for the clustered regularly interspaced short palindromic repeats (CRISPR) machinery. We illustrate different strategies for the optimization of CDV cargo loading and retargeting, highlighting the versatility and tunability of these vehicles. Nonetheless, the lack of robust and standardized protocols for CDV production, purification, and quality assessment still hinders their widespread adoption to further CRISPR-based therapies as advanced "living drugs." We believe that a collective, multifaceted effort is urgently needed to address these critical issues and unlock the full potential of genome-editing technologies to yield safe, easy-to-manufacture, and pharmacologically well-defined therapies. Finally, we discuss the current clinical landscape of lung-directed gene therapies for cystic fibrosis and explore how CDVs could drive significant breakthroughs in in vivo gene editing for this disease.
Collapse
Affiliation(s)
- Laudonia Lidia Dipalo
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | | | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, Advanced Disease Modelling, Targeted Drug Discovery, and Gene Therapy (ADVANTAGE) labs, KU Leuven, Leuven, Belgium
- Leuven Viral Vector Core, group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Marianne S Carlon
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Zhou P, Chao Q, Li C, Wang N, Guo S, Wang P, Ge P, Li C, Guo R, Yang N, Lu Z, Tang Z, Zhang Q, Fu T, Xiao Q, Zhu H. Microglia-targeting nanosystems that cooperatively deliver Chinese herbal ingredients alleviate behavioral and cognitive deficits in Alzheimer's disease model mice. J Nanobiotechnology 2025; 23:313. [PMID: 40275381 PMCID: PMC12020378 DOI: 10.1186/s12951-025-03385-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The effective treatment of Alzheimer's disease (AD) is challenging because of its complex and controversial pathological mechanisms. Moreover, multiple barriers, such as the blood-brain barrier (BBB), reduce drug delivery efficiency. Microglia-related neuroinflammation has recently attracted increasing attention as a possible cause of AD and has become a novel therapeutic target. Therefore, overcoming the BBB and targeted delivery of anti-inflammatory agents to microglia seem to be effective practical strategies for treating AD. A large proportion of natural active extracts possess exceptional immunomodulating capabilities. In this study, the cooperative delivery of berberine (Ber) and palmatine (Pal) by transferrin-decorated extracellular vesicles (Tf-hEVs-Ber/Pal), which can cross the BBB and precisely target microglia, was performed. This nanosystem effectively cleared amyloid β-protein (Aβ) aggregates, significantly regulated the neuroinflammatory environment both in vitro and in vivo and markedly altered the behavior and improved the cognitive and learning abilities of AD model mice. The efficacy of a microglia-targeting combined therapeutic approach for AD was demonstrated, which broadens the potential application of Chinese herbal ingredients.
Collapse
Affiliation(s)
- Peng Zhou
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Quan Chao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ningjing Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Siqi Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingping Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingyuan Ge
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Caihong Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianyun Yang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhangdi Lu
- Polymer Science and Materials Chemistry, Exponent Ltd, Hong Kong, 999077, China
| | - Zhishu Tang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources, Shannxi University of Chinese Medicine, Xianyang, 712038, China
- China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qichun Zhang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingming Fu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingqing Xiao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Huaxu Zhu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
4
|
Jing B, Gao Y, Wang L, Guo F, Jiang D, Qin S, He M, Bai Y, An R, Xie M, Zhang L. Probiotic membrane vesicles ameliorate atherosclerotic plaques by promoting lipid efflux and polarization of foamy macrophages. J Nanobiotechnology 2025; 23:296. [PMID: 40241138 PMCID: PMC12004830 DOI: 10.1186/s12951-025-03360-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Foamy macrophages are pivotal contributors to the development and progression of atherosclerotic plaques, posing a substantial threat to human health. Presently, there is no pharmaceutical intervention available to effectively eliminate foamy macrophages. In this study, we demonstrate that probiotic membrane vesicles (MVs) can induce atherosclerotic plaque regression by modulating foamy macrophages. MVs isolated from Lactobacillus rhamnosus exhibited a specific uptake by foamy macrophages. Near-infrared fluorescence (NIRF) imaging, aortic oil red O staining, and hematoxylin and eosin staining showed reductions in the plaque area following MVs treatment. Mechanistically, bioinformatics analysis provided insights into how MVs exert their effects, revealing that they promote lipid efflux and macrophage polarization. Notably, MVs treatment upregulated NR1H3, which in turn increased ABCA1 expression, facilitating lipid efflux from foamy macrophages. Moreover, MVs shifted macrophage polarization from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype, highlighting their potential to create a more protective environment against plaque progression. This study is significant as it introduces MVs as a novel therapeutic platform for the targeted delivery of anti-inflammatory agents to atherosclerotic sites. By specifically modulating macrophage function, MVs hold considerable potential for the treatment of atherosclerosis and related cardiovascular diseases, addressing an unmet need in current therapeutic strategies.
Collapse
Affiliation(s)
- Boping Jing
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yu Gao
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Saimei Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengrong He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ying Bai
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rui An
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan, 430022, Hubei Province, China.
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
5
|
Kawiková I, Špička V, Lai JCK, Askenase PW, Wen L, Kejík Z, Jakubek M, Valeš K, Španiel F. Extracellular vesicles as precision therapeutics for psychiatric conditions: targeting interactions among neuronal, glial, and immune networks. Front Immunol 2025; 16:1454306. [PMID: 40264776 PMCID: PMC12011847 DOI: 10.3389/fimmu.2025.1454306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/14/2025] [Indexed: 04/24/2025] Open
Abstract
The critical role of the immune system in brain function and dysfunction is well recognized, yet development of immune therapies for psychiatric diseases has been slow due to concerns about iatrogenic immune deficiencies. These concerns are emphasized by the lack of objective diagnostic tools in psychiatry. A promise to resolve this conundrum lies in the exploitation of extracellular vesicles (EVs) that are physiologically produced or can be synthetized. EVs regulate recipient cell functions and offer potential for EVs-based therapies. Intranasal EVs administration enables the targeting of specific brain regions and functions, thereby facilitating the design of precise treatments for psychiatric diseases. The development of such therapies requires navigating four dynamically interacting networks: neuronal, glial, immune, and EVs. These networks are profoundly influenced by brain fluid distribution. They are crucial for homeostasis, cellular functions, and intercellular communication. Fluid abnormalities, like edema or altered cerebrospinal fluid (CSF) dynamics, disrupt these networks, thereby negatively impacting brain health. A deeper understanding of the above-mentioned four dynamically interacting networks is vital for creating diagnostic biomarker panels to identify distinct patient subsets with similar neuro-behavioral symptoms. Testing the functional pathways of these biomarkers could lead to new therapeutic tools. Regulatory approval will depend on robust preclinical data reflecting progress in these interdisciplinary areas, which could pave the way for the design of innovative and precise treatments. Highly collaborative interdisciplinary teams will be needed to achieve these ambitious goals.
Collapse
Affiliation(s)
- Ivana Kawiková
- National Institute of Mental Health, Klecany, Czechia
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
- Department of Biology, Hartford University, West Hartford, CT, United States
| | - Václav Špička
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - James C. K. Lai
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University College of Pharmacy, Pocatello, ID, United States
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Philip W. Askenase
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Li Wen
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Zdeněk Kejík
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Milan Jakubek
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Valeš
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| | - Filip Španiel
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| |
Collapse
|
6
|
Qin D, Huang P, Chen J, Wu C, Liang Y. The therapeutic potential of different mesenchymal stem cells and their derived exosomes in metabolic dysfunction-associated steatotic liver disease. Front Endocrinol (Lausanne) 2025; 16:1558194. [PMID: 40248144 PMCID: PMC12003127 DOI: 10.3389/fendo.2025.1558194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/19/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease is a metabolic disease with an increasing incidence. Its pathogenesis involves the interaction of multiple factors. There is currently no specific treatment, so early prevention and treatment are crucial. Mesenchymal stem cells are a type of cell with the ability to self-renew and differentiate in multiple directions. They have a wide range of sources, including umbilical cords, bone marrow, and fat, and have various biological functions such as anti-inflammation, immune regulation, anti-oxidation, and inhibition of fibrosis. They have shown significant potential in the treatment of non-alcoholic fatty liver disease. In recent years, mesenchymal stem cells derived exosomes have been shown to be rich in bioactive substances, and to be involved in intercellular communication, regulating metabolism, reducing inflammatory responses, improving lipid metabolism, inhibiting fibrosis, and other processes that contribute to the treatment of metabolic dysfunction-associated steatotic liver disease. Mesenchymal stem cells and mesenchymal stem cell-derived exosomes play an important role in the pathogenesis and treatment of metabolic dysfunction-associated steatotic liver disease and provide new potential and direction for the treatment of Metabolic dysfunction-associated steatotic liver disease. This article reviews the role and effects of mesenchymal stem cells and mesenchymal stem cell-derived exosomes from different sources in Metabolic dysfunction-associated steatotic liver disease and discusses their prospects as potential therapeutic strategies.
Collapse
Affiliation(s)
- Dan Qin
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Pingping Huang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jialing Chen
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Changjun Wu
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuzhen Liang
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
7
|
Li W, Zhang H, Chen L, Huang C, Jiang Z, Zhou H, Zhu X, Liu X, Zheng Z, Yu Q, He Y, Gao Y, Ma J, Yang L. Cell membrane-derived nanovesicles as extracellular vesicle-mimetics in wound healing. Mater Today Bio 2025; 31:101595. [PMID: 40104636 PMCID: PMC11914519 DOI: 10.1016/j.mtbio.2025.101595] [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: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Cell membrane-derived nanovesicles (NVs) have emerged as promising alternatives to extracellular vesicles (EVs) for wound healing applications, addressing the limitations of traditional EVs, which include insufficient targeting capability, low production yield, and limited drug-loading capacity. Through mechanical cell extrusion methods, NVs exhibit superior characteristics, demonstrating enhanced yield, stability, and purity compared to natural EVs. These NVs can be derived from various membrane sources, including single cell types (stem cells, blood cells, immune cells, and bacterial membranes), hybrid cell membranes and cell membranes mixed with liposomes, with each offering unique therapeutic properties. The integration of genetic engineering and surface modifications has further enhanced NV functionality, enabling precise targeting and improved drug delivery capabilities. Recent advances in NV-based therapies have demonstrated their potential across multiple biomedical applications. Although challenges persist in terms of standardization, storage stability, and clinical translation, the combination of natural cell-derived functions with artificial modification potential positions NVs as a promising platform for next-generation therapeutic delivery systems, thereby offering new possibilities in wound healing applications. Finally, we explore the challenges and future prospects of translating NV-based therapeutics into clinical practice, providing insights into the future development of this innovative approach in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxi Zhu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zesen Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
8
|
Tiwari P, Shukla RP, Yadav K, Sharma M, Bakshi AK, Panwar D, Singh N, Agarwal N, Mugale MN, Mishra PR. YIGSR Functionalized Hybrid Exosomes Spatially Target Dasatinib to Laminin Receptors for Precision Therapy in Breast Cancer. Adv Healthc Mater 2025; 14:e2402673. [PMID: 39962816 DOI: 10.1002/adhm.202402673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/17/2024] [Indexed: 04/08/2025]
Abstract
In this study, YIGSR-functionalized exosomes (Exo) are engineered and hybridized with lipid polymeric nanoparticles (LPNPs) followed by loading of chemotherapy Dasatinib (DST) to spatially target laminin receptors on tumors. Exo derived from differentiated macrophages are engineered with YIGSR targeting peptides.These YIGSR-Exo are subsequently fused with LPNPs membranes using the freeze-thaw method, resulting in fused hybrid YIGSR-Exo, which are then loaded with DST, creating DST-FuNP@YIGSR-Exo and targeted breast cancer (BC), leading to enhanced mitochondrial membrane potential (54.50 ±5.0%), increased reactive oxygen species (59.50 ± 6.0%), and apoptosis (63 ± 6.5%), ultimately inducing cell death. Further, cellular uptake and receptor blocking studies confirm the binding affinity and interaction of DST-FuNP@YIGSR-Exo with laminin receptors, Intravenous pharmacokinetic analysis of DST-FuNP@YIGSR-Exo reveals a significant improvement in AUC0-∞, with a 20.84-fold increase compared to free DST and a 1.61-fold enhancement over DST-FuNP@Exo. This is further supported by in vivo imaging and demonstrated improved tumor localization. A tumor regression study shows a 6.8-fold reduction in tumors. Tumor tissue-specific IHC for the Ki67 proliferative marker is significantly reduced in the targeted formulation. The potential of DST-FuNP@YIGSR-Exo as an effective carrier for delivering chemotherapeutic drugs, paving the path for the advancement of biologically obtained nanocarriers for targeted breast cancer.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
9
|
Vahab SA, V VK, Kumar VS. Exosome-based drug delivery systems for enhanced neurological therapeutics. Drug Deliv Transl Res 2025; 15:1121-1138. [PMID: 39325272 DOI: 10.1007/s13346-024-01710-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are small extracellular vesicles naturally secreted by cells into body fluids, enriched with bioactive molecules such as RNAs, proteins, and lipids. These nanosized vesicles play a crucial role in physiological and pathological processes by facilitating intercellular communication and modulating cellular responses, particularly within the central nervous system (CNS). Their ability to cross the blood-brain barrier and reflect the characteristics of their parent cells makes exosomal cargo a promising candidate for biomarkers in the early diagnosis and clinical assessment of neurological conditions. This review offers a comprehensive overview of current knowledge on the characterization of mammalian-derived exosomes, their application as drug delivery systems for neurological disorders, and ongoing clinical trials involving exosome-loaded cargo. Despite their promising attributes, a significant challenge remains the lack of standardized isolation methods, as current techniques are often complex, costly, and require sophisticated equipment, affecting the scalability and affordability of exosome-based therapies. The review highlights the engineering potential of exosomes, emphasizing their ability to be customized for targeted therapeutic delivery through surface modification or conjugation. Future advancements in addressing these challenges and leveraging the unique properties of exosomes could lead to innovative and effective therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Safa A Vahab
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India
| | - Vyshma K V
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India
| | - Vrinda S Kumar
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India.
| |
Collapse
|
10
|
Tran V, Nguyen N, Renkes S, Nguyen KT, Nguyen T, Alexandrakis G. Current and Near-Future Technologies to Quantify Nanoparticle Therapeutic Loading Efficiency and Surface Coating Efficiency with Targeted Moieties. Bioengineering (Basel) 2025; 12:362. [PMID: 40281721 PMCID: PMC12025210 DOI: 10.3390/bioengineering12040362] [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: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Active targeting nanoparticles are a new generation of drug and gene delivery systems with the potential for greatly improved therapeutics delivery compared to conventional nanomedicine approaches. Despite their potential, the translation of active targeting nanoparticles faces challenges in production scale-up and batch consistency. Accurate quality control methods for nanoparticle therapeutic payload and coating characterization are critical for attaining the desired levels of batch repeatability, drug/gene loading efficiency, targeting molecule coating effectiveness, and safety profiles. Current limitations in nanoparticle characterization technologies, such as relying on ensemble-average analysis, pose challenges in assessing the drug/gene content and surface modification heterogeneity, which can greatly affect therapeutic outcomes. Single-molecule analysis technologies have emerged as a promising alternative, offering rich information on heterogeneity and stochastic variations between nanoparticle batches. This review first evaluates and identifies the challenges of traditional nanoparticle characterization tools that rely on indirect, bulk solution quantification methods. Subsequently, newly emerging characterization technologies are introduced for the quantification of therapeutic loading and targeted moiety coating efficiencies with single-nanoparticle resolution, to help guide researchers towards advancing the translation of active targeting nanoparticles into the clinical setting.
Collapse
Affiliation(s)
| | | | | | | | - Tam Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA; (V.T.); (N.N.); (S.R.); (K.T.N.)
| | - George Alexandrakis
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA; (V.T.); (N.N.); (S.R.); (K.T.N.)
| |
Collapse
|
11
|
Wu P, Wang Z, Sun Y, Cheng Z, Wang M, Wang B. Extracellular vesicles: a new frontier in diagnosing and treating graft-versus-host disease after allogeneic hematopoietic cell transplantation. J Nanobiotechnology 2025; 23:251. [PMID: 40133949 PMCID: PMC11938667 DOI: 10.1186/s12951-025-03297-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Graft-versus-host disease (GvHD) is a prevalent complication following allogeneic hematopoietic stem cell transplantation (HSCT) and is characterized by relatively high morbidity and mortality rates. GvHD can result in extensive systemic damage in patients following allogeneic HSCT (allo-HSCT), with the skin, gastrointestinal tract, and liver frequently being the primary target organs affected. The severe manifestations of acute intestinal GvHD often indicate a poor prognosis for patients after allo-HSCT. Endoscopy and histopathological evaluation remain employed to diagnose GvHD, and auxiliary examinations exclude differential diagnoses. Currently, reliable serum biomarkers for the diagnosis and differential diagnosis of GvHD are scarce. As an essential part of standard transplant protocols, early application of immunosuppressive drugs effectively prevents GvHD. Among them, steroids represent first-line therapeutic agents, and the JAK2 inhibitor ruxolitinib represents the second-line therapeutic agent. Currently, no efficacious treatment modality exists for steroid-resistant aGvHD. Therefore, the diagnosis and treatment of GvHD still face significant medical demands. Extracellular vesicles (EVs) are nanometer to micrometer-scale biomembrane vesicles containing various bioactive components, such as proteins, nucleotides, and metabolites. Distinctive changes in serum-derived EV components occur in patients after allo-HSCT; Hence, EVs are expected to be potential biomarkers for diagnosing and treating GvHD. Furthermore, cell-free therapeutics characterized by EVs derived from mesenchymal stem cells (MSCs) have manifested remarkable therapeutic efficacy in preclinical models and preclinical trials of GvHD. Customized engineered EVs with fewer toxic and side effects for the combined treatment of GvHD hold broad prospects for clinical translation. This review article examines the potential value of translating EVs into clinical applications for the diagnosis and treatment of GvHD. It summarizes the latest advancements and prospects of engineered EVs applying GvHD.
Collapse
Affiliation(s)
- Peipei Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Zhangfei Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yongping Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhixiang Cheng
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Min Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China.
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
12
|
Ghufran SM, Brown ML, Beierle EA. Role of exosomes in diagnosis, prognostication, and treatment of pediatric solid tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200930. [PMID: 39895692 PMCID: PMC11783428 DOI: 10.1016/j.omton.2024.200930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cancer is the second leading cause of death in children, and solid tumors make up 30% of childhood cancers. Molecular profiling of pediatric solid tumors allows a personalized approach to therapy, but this approach mostly relies on surgical biopsy, which is invasive and carries the risk of complications. Liquid biopsy serves as a reliable alternative and a minimally invasive tool for diagnosing, prognosticating, and residual disease monitoring in childhood cancers. This review outlines the potential of exosomes as informative liquid biopsies in pediatric solid tumors. Studies highlighting the potential applications and clinical utility of exosomes and their molecular constituents as prognosticators and therapies in common childhood solid tumors, including neuroblastoma, medulloblastoma, sarcoma, and hepatoblastoma, have been overviewed. We also discuss the limitations and technical challenges of utilizing exosomes for pediatric solid tumors.
Collapse
Affiliation(s)
- Shaikh M. Ghufran
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Morgan L. Brown
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Elizabeth A. Beierle
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| |
Collapse
|
13
|
López RR, Ben El Khyat CZ, Chen Y, Tsering T, Dickinson K, Bustamante P, Erzingatzian A, Bartolomucci A, Ferrier ST, Douanne N, Mounier C, Stiharu I, Nerguizian V, Burnier JV. A synthetic model of bioinspired liposomes to study cancer-cell derived extracellular vesicles and their uptake by recipient cells. Sci Rep 2025; 15:8430. [PMID: 40069225 PMCID: PMC11897354 DOI: 10.1038/s41598-025-91873-5] [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: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Extracellular vesicles (EVs) are secreted by most cell types and play a central role in cell-cell communication. These naturally occurring nanoparticles have been particularly implicated in cancer, but EV heterogeneity and lengthy isolation methods with low yield make them difficult to study. To circumvent the challenges in EV research, we aimed to develop a unique synthetic model by engineering bioinspired liposomes to study EV properties and their impact on cellular uptake. We produced EV-like liposomes mimicking the physicochemical properties as cancer EVs. First, using a panel of cancer and non-cancer cell lines, small EVs were isolated by ultracentrifugation and characterized by dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). Cancer EVs ranged in mean size from 107.9 to 161 nm by NTA, hydrodynamic diameter from 152 to 355 nm by DLS, with a zeta potential ranging from - 25 to -6 mV. EV markers TSG101 and CD81 were positive on all EVs. Using a microfluidics bottom-up approach, liposomes were produced using the nanoprecipitation method adapted to micromixers developed by our group. A library of liposome formulations was created that mimicked the ranges of size (90-222 nm) and zeta potential (anionic [-47 mV] to neutral [-1 mV]) at a production throughput of up to 41 mL/h and yielding a concentration of 1 × 1012 particles per mL. EV size and zeta potential were reproduced by controlling the flow conditions and lipid composition set by a statistical model based on the response surface methodology. The model was fairly accurate with an R-squared > 70% for both parameters between the targeted EV and the obtained liposomes. Finally, the internalization of fluorescently labeled EV-like liposomes was assessed by confocal microscopy and flow cytometry, and correlated with decreasing liposome size and less negative zeta potential, providing insights into the effects of key EV physicochemical properties. Our data demonstrated that liposomes can be used as a powerful synthetic model of EVs. By mimicking cancer cell-derived EV properties, the effects on cellular internalization can be assessed individually and in combination. Taken together, we present a novel system that can accelerate research on the effects of EVs in cancer models.
Collapse
Grants
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 190179 Canadian Institutes for Health Research
- 190179 Canadian Institutes for Health Research
- 177808 National Sciences and Engineering Research Council of Canada (NSERC)
- NFRFE-2019-01587 Government of Canada's New Frontiers in Research Fund (NFRF)
- Government of Canada’s New Frontiers in Research Fund (NFRF)
Collapse
Affiliation(s)
- Rubén R López
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Chaymaa Zouggari Ben El Khyat
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Prisca Bustamante
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Sarah Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Catherine Mounier
- Department of biological sciences, Université du Québec à Montréal, 141 avenue du président Kennedy, Montreal, QC, H2X 1Y4, Canada
- Department of Mechanical, Industrial and Aerospace Engineering, Concordia University, 1455 de Maisonneuve Blvd. West, Montreal, QC, H3G 1M8, Canada
| | - Ion Stiharu
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montreal, QC, H4A 3T2, Canada
| | - Vahé Nerguizian
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada.
- Department of Pathology, McGill University, Quebec, Canada.
| |
Collapse
|
14
|
Bang S, Park B, Park JC, Jin H, Shim JS, Koo J, Lee KH, Shim MK, Kim H. Exosome-Inspired Lipid Nanoparticles for Enhanced Tissue Penetration. ACS NANO 2025; 19:8882-8894. [PMID: 40017353 DOI: 10.1021/acsnano.4c16629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules with varying pore sizes, posing a challenge for the effective penetration of lipid nanoparticles. In contrast, cell-derived lipid nanoparticles, such as exosomes, have demonstrated the ability to travel to distant organs, indicating their capacity to penetrate the ECM. Here, we designed exosome-like vesicles (ELVs) inspired by exosomes' distinct transport phenomena. Specifically, we integrated three exosomal components (anionic lipid, cholesterol, and aquaporin-1) associated with transport into our ELVs to mimic the superior diffusion behavior of exosomes over synthetic lipid nanoparticles. Surprisingly, both bulk- and single-particle-diffusion studies revealed a more than 33 times increase in the effective diffusion coefficient within model ECM compared to conventional lipid nanoparticles. Furthermore, ELVs show an 80% increase in the effective diffusion coefficient within biological tissues. The excellent transport behavior of ELVs was further validated in vivo, where intratumoral injection showcased their superior transport. These findings provide insights into lipid nanoparticle design for improved tissue penetration.
Collapse
Affiliation(s)
- Seunghwan Bang
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Byeongmin Park
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jae Chul Park
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Harin Jin
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Ji Sung Shim
- Department of Urology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jahyun Koo
- School of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Kwan Hyi Lee
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hojun Kim
- Division of Bio-Medical Science & Technology, KIST school, University of Science and Technology, Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
15
|
Li L, Wang F, Zhu D, Hu S, Cheng K, Li Z. Engineering exosomes and exosome-like nanovesicles for improving tissue targeting and retention. FUNDAMENTAL RESEARCH 2025; 5:851-867. [PMID: 40242543 PMCID: PMC11997600 DOI: 10.1016/j.fmre.2024.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/18/2025] Open
Abstract
Exosomes are natural nano-size particles secreted by human cells, containing numerous bioactive cargos. Serving as crucial mediators of intercellular communication, exosomes are involved in many physiological and pathological processes, such as inflammation, tissue injury, cardiovascular diseases, tumorigenesis and tumor development. Exosomes have exhibited promising results in the diagnosis and treatment of cancer, cardiovascular diseases and others. They are a rapidly growing class of drug delivery vehicles with many advantages over conventional synthetic carriers. Exosomes used in therapeutic applications encounter several challenges, such as the lack of tissue targeting capabilities and short residence time. In this review, we discuss recent advances in exosome engineering to improve tissue targeting and describe the current types of engineered exosome-like nanovesicles, and summarize their preclinical applications in the treatment of diseases. Further, we also highlight the latest engineering strategies developed to extend exosomes retention time in vivo and exosome-like nanovesicles.
Collapse
Affiliation(s)
- Lanya Li
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Southern Medical University, Guangdong 510515, China
| | - Fei Wang
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Southern Medical University, Guangdong 510515, China
| | - Dashuai Zhu
- Department of Biomedical Engineering, Columbia University, New York 10032, USA
| | - Shiqi Hu
- Department of Biomedical Engineering, Columbia University, New York 10032, USA
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York 10032, USA
| | - Zhenhua Li
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Southern Medical University, Guangdong 510515, China
| |
Collapse
|
16
|
Pozy E, Brown AC. Transfusion biomaterials for hemostasis. J Thromb Haemost 2025; 23:790-803. [PMID: 39675569 DOI: 10.1016/j.jtha.2024.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
Bleeding is a leading cause of trauma deaths and surgical complications. Excessive bleeding has traditionally been treated with the transfusion of donated blood. However, the complicated logistics of sourcing and storing donated blood increases the cost and reduces the accessibility of treatment, particularly as rates of blood donation decline. Advances in biomaterials for targeted drug delivery have presented the opportunity for alternative synthetic injectable hemostats. Among these leading technologies are lipid and polymeric particles and polymer platforms that bind to ligands present at wound sites and amplify hemostatic pathways. As leading hemostatic biomaterials advance toward clinical application, we review current preclinical research models and findings as well as future research directions for next-generation biomaterial injectable hemostatic technologies.
Collapse
Affiliation(s)
- Ethan Pozy
- Joint Department of Biomedical Engineering of University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering of University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA.
| |
Collapse
|
17
|
Chen Y, Douanne N, Wu T, Kaur I, Tsering T, Erzingatzian A, Nadeau A, Juncker D, Nerguizian V, Burnier JV. Leveraging nature's nanocarriers: Translating insights from extracellular vesicles to biomimetic synthetic vesicles for biomedical applications. SCIENCE ADVANCES 2025; 11:eads5249. [PMID: 40009680 PMCID: PMC11864201 DOI: 10.1126/sciadv.ads5249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Naturally occurring extracellular vesicles (EVs) and synthetic nanoparticles like liposomes have revolutionized precision diagnostics and medicine. EVs excel in biocompatibility and cell targeting, while liposomes offer enhanced drug loading capacity and scalability. The clinical translation of EVs is hindered by challenges including low yield and heterogeneity, whereas liposomes face rapid immune clearance and limited targeting efficiency. To bridge these gaps, biomimetic synthetic vesicles (SVs) have emerged as innovative platforms, combining the advantageous properties of EVs and liposomes. This review emphasizes critical aspects of EV biology, such as mechanisms of EV-cell interaction and source-dependent functionalities in targeting, immune modulation, and tissue regeneration, informing biomimetic SV engineering. We reviewed a broad array of biomimetic SVs, with a focus on lipid bilayered vesicles functionalized with proteins. These include cell-derived nanovesicles, protein-functionalized liposomes, and hybrid vesicles. By addressing current challenges and highlighting opportunities, this review aims to advance biomimetic SVs for transformative biomedical applications.
Collapse
Affiliation(s)
- Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Tad Wu
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Ishman Kaur
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- École de technologie supérieure ÉTS, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - David Juncker
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | | | - Julia V. Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Quan J, Liu Q, Li P, Yang Z, Zhang Y, Zhao F, Zhu G. Mesenchymal stem cell exosome therapy: current research status in the treatment of neurodegenerative diseases and the possibility of reversing normal brain aging. Stem Cell Res Ther 2025; 16:76. [PMID: 39985030 PMCID: PMC11846194 DOI: 10.1186/s13287-025-04160-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 02/23/2025] Open
Abstract
With the exacerbation of the aging population trend, a series of neurodegenerative diseases caused by brain aging have become increasingly common, significantly impacting the daily lives of the elderly and imposing heavier burdens on nations and societies. Brain aging is a complex process involving multiple mechanisms, including oxidative stress, apoptosis of damaged neuronal cells, chronic inflammation, and mitochondrial dysfunction, and research into new therapeutic strategies to delay brain aging has gradually become a research focus in recent years. Mesenchymal stem cells (MSCs) have been widely used in cell therapy due to their functions such as antioxidative stress, anti-inflammation, and tissue regeneration. However, accompanying safety issues such as immune rejection, tumor development, and pulmonary embolism cannot be avoided. Studies have shown that using exosome derived from mesenchymal stem cells (MSC-Exo) for the treatment of neurodegenerative diseases is a safe and effective method. It not only has the therapeutic effects of stem cells but also avoids the risks associated with cell therapy. Therefore, exploring new therapeutic strategies to delay normal brain aging from the mechanism of MSC-Exo in the treatment of neurodegenerative diseases is feasible. This review summarizes the characteristics of MSC-Exo and their clinical progress in the treatment of neurodegenerative diseases, aiming to explore the possibility and potential mechanisms of MSC-Exo in reversing brain aging.
Collapse
Affiliation(s)
- Jinglan Quan
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Qing Liu
- Department of Library, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Pinghui Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Zhiyu Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Yaohui Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Fuxing Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Gaohong Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China.
| |
Collapse
|
19
|
Wang Q, Sun J, Jiang H, Yu M. Emerging roles of extracellular vesicles in oral and maxillofacial areas. Int J Oral Sci 2025; 17:11. [PMID: 39900916 PMCID: PMC11791077 DOI: 10.1038/s41368-024-00341-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 02/05/2025] Open
Abstract
The oral and maxillofacial region is a highly complex area composed of multiple tissue types and bears various critical functions of the human body. Diseases in this region pose significant diagnostic and management challenges; therefore, exploring new strategies for early diagnosis, targeted treatment, and tissue reconstruction is key to improving patient prognosis and quality of life. Extracellular vesicles are a group of heterogeneous lipid-bilayer membrane structures secreted by most cell types, including exosomes, microvesicles, and apoptotic bodies. Present in various body fluids and tissues, they act as messengers via the transfer of nucleic acids, proteins, and metabolites to recipient cells. To date, studies have revealed the different roles of extracellular vesicles in physiological or pathological processes, as well as applications in disease diagnosis, prognosis, and treatment. The importance and tissue specificity of the dental and maxillofacial tissues indicate that extracellular vesicles derived from this region are promising for further research. This paper reviews the published data on extracellular vesicles derived from cells, body fluids, and tissues in oral and maxillofacial regions, summarizes the latest advances in extracellular vesicles from extensive sources, and concludes with a focus on the current research progress and application prospects of engineered exosomes in oral science.
Collapse
Affiliation(s)
- Qianting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Jiayu Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Haci Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
20
|
Li J, Wang J, Chen Z. Emerging role of exosomes in cancer therapy: progress and challenges. Mol Cancer 2025; 24:13. [PMID: 39806451 PMCID: PMC11727182 DOI: 10.1186/s12943-024-02215-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
This review highlights recent progress in exosome-based drug delivery for cancer therapy, covering exosome biogenesis, cargo selection mechanisms, and their application across multiple cancer types. As small extracellular vesicles, exosomes exhibit high biocompatibility and low immunogenicity, making them ideal drug delivery vehicles capable of efficiently targeting cancer cells, minimizing off-target damage and side effects. This review aims to explore the potential of exosomes in cancer therapy, with a focus on applications in chemotherapy, gene therapy, and immunomodulation. Additionally, challenges related to exosome production and standardization are analyzed, highlighting the importance of addressing these issues for their clinical application. In conclusion, exosome-based drug delivery systems offer promising potential for future cancer therapies. Further research should aim to enhance production efficiency and facilitate clinical translation, paving the way for innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Jiachong Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| | - Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| |
Collapse
|
21
|
Lou S, Hu W, Wei P, He D, Fu P, Ding K, Chen Z, Dong Z, Zheng J, Wang K. Artificial Nanovesicles Derived from Cells: A Promising Alternative to Extracellular Vesicles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22-41. [PMID: 39692623 DOI: 10.1021/acsami.4c12567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
As naturally secreted vesicles by cells, extracellular vesicles (EVs) play essential roles in modulating cell-cell communication and have significant potential in tissue regeneration, immune regulation, and drug delivery. However, the low yield and uncontrollable heterogeneity of EVs have been obstacles to their widespread translation into clinical practice. Recently, it has been discovered that artificial nanovesicles (NVs) produced by cell processing can inherit the components and functions of the parent cells and possess similar structures and functions to EVs, with significantly higher yields and more flexible functionalization, making them a powerful complement to natural EVs. This review focuses on recent advances in the research of artificial NVs as replacements for natural EVs. We provide an overview comparing natural EVs and artificial NVs and summarize the top-down preparation strategies of NVs. The applications of NVs prepared from stem cells, differentiated cells, and engineered cells are presented, as well as the latest advances in NV engineering. Finally, the main challenges of artificial NVs are discussed.
Collapse
Affiliation(s)
- Saiyun Lou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Wei Hu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Pengyao Wei
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Dongdong He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kejian Ding
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhenyi Chen
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo,Zhejiang 315211, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Jianping Zheng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo 315300, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Carnicer‐Lombarte A, Malliaras GG, Barone DG. The Future of Biohybrid Regenerative Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2408308. [PMID: 39564751 PMCID: PMC11756040 DOI: 10.1002/adma.202408308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/13/2024] [Indexed: 11/21/2024]
Abstract
Biohybrid regenerative bioelectronics are an emerging technology combining implantable devices with cell transplantation. Once implanted, biohybrid regenerative devices integrate with host tissue. The combination of transplant and device provides an avenue to both replace damaged or dysfunctional tissue, and monitor or control its function with high precision. While early challenges in the fusion of the biological and technological components limited development of biohybrid regenerative technologies, progress in the field has resulted in a rapidly increasing number of applications. In this perspective the great potential of this emerging technology for the delivery of therapy is discussed, including both recent research progress and potential new directions. Then the technology barriers are discussed that will need to be addressed to unlock the full potential of biohybrid regenerative devices.
Collapse
Affiliation(s)
| | - George G. Malliaras
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
| | - Damiano G. Barone
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
- Department of Neurosurgery, Houston MethodistHouston77030USA
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| |
Collapse
|
23
|
Guo F, Wu Y, Wang G, Liu J. Role of PCBP2 in regulating nanovesicles loaded with curcumin to mitigate neuroferroptosis in neural damage caused by heat stroke. J Nanobiotechnology 2024; 22:800. [PMID: 39731111 DOI: 10.1186/s12951-024-02889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/01/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the mechanisms by which nanovesicles (NVs) transport curcumin(CUR) across the blood-brain barrier to treat hypothalamic neural damage induced by heat stroke by regulating the expression of poly(c)-binding protein 2 (PCBP2). METHODS Initially, NVs were prepared from macrophages using a continuous extrusion method. Subsequently, CUR was loaded into NVs using sonication, yielding engineered cell membrane Nanovesicles loaded with curcumin (NVs-CUR), which were characterized and subjected to in vitro and in vivo tracking analysis. Evaluations included assessing the toxicity of NVs-CUR using the MTT assay, evaluating neuroprotection of NVs-CUR against H2O2-induced oxidative stress damage in PC12 cells, examining effects on cell morphology and quantity, and detecting ferroptosis-related markers through Western blot and transmission electron microscopy (TEM). Proteomic analysis was conducted on PC12 cells treated with NVs (n = 3) and NVs-CUR (n = 3) to identify downstream key factors. Subsequently, the expression of key factors was modulated, and rescue experiments were performed to validate the impact of NVs-CUR through the regulation of key factor expression. Furthermore, a mouse model of hypothalamic neural damage induced by heat stroke was established, where CUR, NVs-CUR, and ferroptosis inducer Erastin were administered to observe mouse survival rates, conduct nerve function deficit scoring, perform histological staining, and measure levels of inflammation and oxidative stress factors in hypothalamic tissue. RESULTS NVs-CUR was successfully prepared with excellent stability, serving as an advantageous drug delivery system that effectively targets brain injury sites or neurons both in vitro and in vivo. Subsequent in vitro cell experiments demonstrated the biocompatibility of NVs-CUR, showing superior protective effects against H2O2-induced PC12 cell damage and reduced ferroptosis compared to CUR. Moreover, in the mouse model of hypothalamic neural damage induced by heat stroke, NVs-CUR exhibited enhanced therapeutic effects. Proteomic analysis revealed that NVs-CUR exerted its effects through the regulation of key protein PCBP2; silencing PCBP2 reversed the protective effect of NVs-CUR on neural damage and its inhibition of ferroptosis. Additionally, NVs-CUR regulated solute carrier family 7 member 11 (SLC7A11) expression by PCBP2; overexpression of SLC7A11 reversed the promotion of neural damage and ferroptosis by silencing PCBP2. Animal experiments indicated that ferroptosis inducers reversed the improved survival and nerve function observed with NVs-CUR, silencing PCBP2 reversed the ameliorative effects of NVs-CUR on hypothalamic neural injury induced by heat stroke, and overexpression of SLC7A11 further reversed the adverse effects of silencing PCBP2 on hypothalamic neural injury induced by heat stroke. This suggests that NVs-CUR alleviates hypothalamic neural damage induced by heat stroke by targeting the PCBP2/SLC7A11 axis to reduce neuronal ferroptosis. CONCLUSION This study successfully developed engineered cell membrane NVs-CUR with neuron-targeting properties. NVs-CUR increased the expression of PCBP2, maintained the stability of SLC7A11 mRNA, reduced ferroptosis, and ultimately alleviated hypothalamic neuroinflammation induced by heatstroke.
Collapse
Affiliation(s)
- Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yizhan Wu
- Graduate School of Xinjiang Medical University, Urumqi, China
| | - Guangjun Wang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, No. 359, Youhao North Road, Urumqi, Xinjiang, China.
| |
Collapse
|
24
|
Tang B, Huang R, Ma W. Advances in nanotechnology-based approaches for the treatment of head and neck squamous cell carcinoma. RSC Adv 2024; 14:38668-38688. [PMID: 39654926 PMCID: PMC11626385 DOI: 10.1039/d4ra07193j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), one of the most common types of cancers occurring in the head and neck region, is often associated with high mortality rates due to its invasiveness and morbidity. The mainstream treatment methods in clinical settings, including surgery, chemotherapy, and radiotherapy, may cause poor overall survival rate and prognosis, with issues such as drug resistance, damage to adjacent healthy tissues, and potential recurrences. Other treatment approaches such as immunotherapy, photodynamic therapy (PDT), and photothermal therapy (PPT) also suffer from inefficient tumor targeting and suboptimal therapeutic outcomes. Early detection is vital for HNSCC patients, but it is always limited by insensitivity and confusing clinical manifestations. Hence, it is highly desirable to develop optimized therapeutic and diagnostic strategies. With the boom in nanomaterials, nanotechnology-conducted HNSCC therapy has attracted widespread attention. Nanoparticles (NPs) are distinguished by their unique morphology and superior physicochemical property, and some can exhibit direct antitumor activity, while others serve as promising candidates for drug delivery. In addition, NPs offer the potential for structural modification for drug delivery and tumor targeting, enabling specific delivery to tumor cells through conjugation with biomarker ligands and improving cargo biocompatibility. This work reviews current therapies and diagnosis methods for HNSCC, highlights the characteristics of the major NPs, surveys their uses and advantages in the treatment of HNSCC, and discusses the obstacles and prospects in clinical applications, aiming to enlighten future research directions for nanotechnology-based therapy for HNSCC.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Rui Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
25
|
Ma YN, Hu X, Karako K, Song P, Tang W, Xia Y. Exploring the multiple therapeutic mechanisms and challenges of mesenchymal stem cell-derived exosomes in Alzheimer's disease. Biosci Trends 2024; 18:413-430. [PMID: 39401895 DOI: 10.5582/bst.2024.01306] [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: 11/19/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder, and the current treatment options are limited. Mesenchymal stem cell-derived exosomes (MSC-Exos) have garnered significant attention due to their unique biological properties, showcasing tremendous potential as an acellular alternative therapy for AD. MSC-Exos exhibit excellent biocompatibility and low immunogenicity, enabling them to effectively cross the blood-brain barrier (BBB) and deliver therapeutic molecules directly to target cells. They are highly efficacious in delivering nucleic acid-based drugs. Moreover, the production process of MSC-Exos benefits from a high proliferation capacity and multilineage differentiation potential, allowing for production while maintaining a stable composition. Despite the significant theoretical advantages of MSC-Exos, their clinical use still faces multiple challenges, including cross-contamination during isolation and purification processes, the complexity of their components, and the presence of potential adverse paracrine factors. Future research needs to focus on optimizing separation and purification techniques, enhancing delivery methods to improve therapeutic efficacy, and performing detailed analyses of the components of MSC-Exos. In summary, MSC-Exos hold promise as an effective option for the treatment of AD and other neurodegenerative diseases, driving their clinical research and use in related fields.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Kenji Karako
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Peipei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
26
|
Zhang SH, Peng LL, Chen YF, Xu Y, Moradi V. Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy. Inflamm Regen 2024; 44:45. [PMID: 39490997 PMCID: PMC11533312 DOI: 10.1186/s41232-024-00358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.
Collapse
Affiliation(s)
- Si-Heng Zhang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China
| | - Ling-Long Peng
- Wuhu Hospital, East China Normal University (The Second People's Hospital of Wuhu), Wuhu, 241000, China
| | - Yi-Fei Chen
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yan Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China.
| | - Vahid Moradi
- Hematology and Bood Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Hu Z, Wang W, Lin Y, Guo H, Chen Y, Wang J, Yu F, Rao L, Fan Z. Extracellular Vesicle-Inspired Therapeutic Strategies for the COVID-19. Adv Healthc Mater 2024; 13:e2402103. [PMID: 38923772 DOI: 10.1002/adhm.202402103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Emerging infectious diseases like coronavirus pneumonia (COVID-19) present significant challenges to global health, extensively affecting both human society and the economy. Extracellular vesicles (EVs) have demonstrated remarkable potential as crucial biomedical tools for COVID-19 diagnosis and treatment. However, due to limitations in the performance and titer of natural vesicles, their clinical use remains limited. Nonetheless, EV-inspired strategies are gaining increasing attention. Notably, biomimetic vesicles, inspired by EVs, possess specific receptors that can act as "Trojan horses," preventing the virus from infecting host cells. Genetic engineering can enhance these vesicles by enabling them to carry more receptors, significantly increasing their specificity for absorbing the novel coronavirus. Additionally, biomimetic vesicles inherit numerous cytokine receptors from parent cells, allowing them to effectively mitigate the "cytokine storm" by adsorbing pro-inflammatory cytokines. Overall, this EV-inspired strategy offers new avenues for the treatment of emerging infectious diseases. Herein, this review systematically summarizes the current applications of EV-inspired strategies in the diagnosis and treatment of COVID-19. The current status and challenges associated with the clinical implementation of EV-inspired strategies are also discussed. The goal of this review is to provide new insights into the design of EV-inspired strategies and expand their application in combating emerging infectious diseases.
Collapse
Affiliation(s)
- Ziwei Hu
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Wei Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Ying Lin
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Hui Guo
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P. R. China
| | - Yiwen Chen
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Junjie Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Feng Yu
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, P. R. China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, Kunming, 650500, P. R. China
| |
Collapse
|
28
|
Chen H, Sun H, Yang Y, Wang P, Chen X, Yin J, Li A, Zhang L, Cai J, Huang J, Zhang S, Zhang Z, Feng X, Yin J, Wang Y, Xiong W, Wan B. Engineered melatonin-pretreated plasma exosomes repair traumatic spinal cord injury by regulating miR-138-5p/SOX4 axis mediated microglia polarization. J Orthop Translat 2024; 49:230-245. [PMID: 39512441 PMCID: PMC11541837 DOI: 10.1016/j.jot.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/08/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Background Neuroinflammation plays a crucial role in the repair of spinal cord injury (SCI), with microglia, pivotal in neuroinflammation, driving either degeneration or recovery in this pathological process. Recently, plasma-derived exosomes (denoted Exos) have presented a high capacity for promoting functional recovery of SCI through the anti-inflammatory effects, and pretreated exosomes are associated with better outcomes. Thus, we aimed to explore whether melatonin-pretreated plasma-derived exosomes (denoted MExo) could exert superior effects on SCI, and attempted to elucidate the potential mechanisms. Methods Electron microscopy, nanoparticle tracking analysis, and western blot were applied to delineate the distinctions between Exos and MExos. To assess their therapeutic potentials, we established a contusion SCI rat model, complemented by a battery of in vitro experiments comparing both groups. Subsequently, a miRNA microarray analysis was conducted, followed by a series of rescue experiments to elucidate the specific role of miRNAs in MExos. To further delve into the molecular mechanisms involved, we employed western blot analysis and the luciferase reporter gene assay. Results Melatonin promoted the release of exosome from plasma, concurrently amplifying their anti-inflammatory properties. Furthermore, it was discerned that MExos facilitated a transition in microglia polarization from M1 to M2 phenotype, a phenomenon more pronounced than that observed with Exos. In an endeavor to elucidate this variance, we scrutinized miRNAs exhibiting elevated expression levels in MExos, pinpointing miR-138-5p as a pivotal element in this dynamic. Following this, an in-depth investigation into the role of miR-138-5p was undertaken, which uncovered its efficacy in driving phenotypic alterations within microglia. The analysis of downstream genes targeted by miR-138-5p revealed that it exerted a negative regulatory influence on SOX4, which was found to obstruct the generation of M2-type microglia and the secretion of anti-inflammatory cytokines, thereby partially elucidating the mechanism behind miR-138-5p's regulation of microglia polarization. Conclusions We innovatively observed that melatonin enhanced the anti-inflammatory function of Exos, which further decreased the expression of SOX4 by delivering miR-138-5p. This inhibition promoted the conversion of M1 microglia to M2 microglia, thus offering a viable option for the treatment of SCI. The translational potential of this article This study highlights that melatonin enhances the anti-inflammatory function of Exos through delivery of miR-138-5p. Activation of miR-138-5p/SOX4 axis by engineered melatonin-pretreated plasma exosomes may be a potential target for SCI treatment.
Collapse
Affiliation(s)
- Hao Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Huihui Sun
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yaqing Yang
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Pingchuan Wang
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Xizhao Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Junxiang Yin
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Aoying Li
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Liang Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jun Cai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jijun Huang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Shengfei Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Zhiqiang Zhang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xinmin Feng
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Orthopedics, the Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of Orthopedics, Northern Jiangsu People's Hospital, Affiliated Hospital of Nanjing University Medical School, Yangzhou, China
| | - Wu Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bowen Wan
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
29
|
Wang P, Chen W, li B, Yang S, Li W, Zhao S, Ning J, Zhou X, Cheng F. Exosomes on the development and progression of renal fibrosis. Cell Prolif 2024; 57:e13677. [PMID: 38898750 PMCID: PMC11533081 DOI: 10.1111/cpr.13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/09/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Renal fibrosis is a prevalent pathological alteration that occurs throughout the progression of primary and secondary renal disorders towards end-stage renal disease. As a complex and irreversible pathophysiological phenomenon, it includes a sequence of intricate regulatory processes at the molecular and cellular levels. Exosomes are a distinct category of extracellular vesicles that play a crucial role in facilitating intercellular communication. Multiple pathways are regulated by exosomes produced by various cell types, including tubular epithelial cells and mesenchymal stem cells, in the context of renal fibrosis. Furthermore, research has shown that exosomes present in bodily fluids, including urine and blood, may be indicators of renal fibrosis. However, the regulatory mechanism of exosomes in renal fibrosis has not been fully elucidated. This article reviewed and analysed the various mechanisms by which exosomes regulate renal fibrosis, which may provide new ideas for further study of the pathophysiological process of renal fibrosis and targeted treatment of renal fibrosis with exosomes.
Collapse
Affiliation(s)
- Peihan Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Wu Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Bojun li
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Songyuan Yang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Wei Li
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Sheng Zhao
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jinzhuo Ning
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Xiangjun Zhou
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
30
|
Liao J, Gu Q, Liu Z, Wang H, Yang X, Yan R, Zhang X, Song S, Wen L, Wang Y. Edge advances in nanodrug therapies for osteoarthritis treatment. Front Pharmacol 2024; 15:1402825. [PMID: 39539625 PMCID: PMC11559267 DOI: 10.3389/fphar.2024.1402825] [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: 03/18/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
As global population and lifestyles change, osteoarthritis (OA) is becoming a major healthcare challenge world. OA, a chronic condition characterized by inflammatory and degeneration, often present with joint pain and can lead to irreversible disability. While there is currently no cure for OA, it is commonly managed using nonsteroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, and glucosamine. Although these treatments can alleviate symptoms, it is difficult to effectively deliver and sustain therapeutic agents within joints. The emergence of nanotechnology, particularly in form of smart nanomedicine, has introduced innovative therapeutic approaches for OA treatment. Nanotherapeutic strategies offer promising advantages, including more precise targeting of affected areas, prolonged therapeutic effects, enhanced bioavailability, and reduced systemic toxicity compared to traditional treatments. While nanoparticles show potential as a viable delivery system for OA therapies based on encouraging lab-based and clinical trials results, there remails a considerable gap between current research and clinical application. This review highlights recent advances in nanotherapy for OA and explore future pathways to refine and optimize OA treatments strategies.
Collapse
Affiliation(s)
- Jinfeng Liao
- Department of Dermatology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Qingjia Gu
- Department of ENT, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Zheng Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Xian Yang
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongkai Yan
- Department of Radiology, Ohio state university, Columbus, OH, United States
| | - Xiaofeng Zhang
- Greenwich Hospital, Yale New Haven Health, Greenwich, CT, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lebin Wen
- Department of Thyroid, Sichuan Second Hospital of TCM, Chengdu, China
| | - Yi Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
31
|
Yang Z, Yang M, Rui S, Hao W, Wu X, Guo L, Armstrong DG, Yang C, Deng W. Exosome-based cell therapy for diabetic foot ulcers: Present and prospect. Heliyon 2024; 10:e39251. [PMID: 39498056 PMCID: PMC11532254 DOI: 10.1016/j.heliyon.2024.e39251] [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: 04/25/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Diabetic foot ulcers (DFUs) represent a serious complication of diabetes with high incidence, requiring intensive treatment, prolonged hospitalization, and high costs. It poses a severe threat to the patient's life, resulting in substantial burdens on patient and healthcare system. However, the therapy of DFUs remains challenging. Therefore, exploring cell-free therapies for DFUs is both critical and urgent. Exosomes, as crucial mediators of intercellular communication, have been demonstrated potentially effective in anti-inflammation, angiogenesis, cell proliferation and migration, and collagen deposition. These functions have been proven beneficial in all stages of diabetic wound healing. This review aims to summarize the role and mechanisms of exosomes from diverse cellular sources in diabetic wound healing research. In addition, we elaborate on the challenges for clinical application, discuss the advantages of membrane vesicles as exosome mimics in wound healing, and present the therapeutic potential of exosomes and their mimetic vesicles for future clinical applications.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Mengling Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Shunli Rui
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wei Hao
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Xiaohua Wu
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Lian Guo
- Department of Endocrinology, School of Medicine, Chongqing University Three Gorges Central Hospital, Chongqing, 404000, China
| | - David G. Armstrong
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Cheng Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| |
Collapse
|
32
|
Song Y, Yin C, Kong N. Stem Cell-Derived Exosomes: Natural Intercellular Messengers with Versatile Mechanisms for the Treatment of Diabetic Retinopathy. Int J Nanomedicine 2024; 19:10767-10784. [PMID: 39469447 PMCID: PMC11514697 DOI: 10.2147/ijn.s475234] [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: 05/26/2024] [Accepted: 10/05/2024] [Indexed: 10/30/2024] Open
Abstract
Diabetic retinopathy is one of the complications of diabetes mellitus that occurs in the early stages. It is a disease that has a serious impact, and may lead to blindness when the disease progresses to advanced stages. Currently, treatments for diabetic retinopathy are mainly limited to its advanced stages of the disease, being restricted to a single therapeutic mechanism. Stem cells hold the promise of regenerative therapy and have the potential to comprehensively improve diabetic retinopathy. However, direct stem cell therapy carries some risk of carcinogenesis. Exosomes secreted by stem cells have shown a similar overall improvement in disease as stem cells. Exosomes can carry a number of biologically active materials from donor cells to recipient cells or distant organs, regulating intercellular signaling. Exosomes have shown remarkable efficacy in alleviating oxidative stress, inhibiting inflammatory responses, suppressing angiogenesis, reducing apoptosis and protecting neural tissues. Currently, the experimental literature using stem cell exosomes in the treatment of diabetic retinopathy tends to converge on the above experimental results. With this in mind, we have chosen to explore exosomes in depth from a subtle molecular perspective. We will elaborate on this perspective in this paper and propose to advocate exosome therapy as one promising approach for the treatment of diabetic retinopathy to ameliorate the lesions through multiple mechanisms.
Collapse
Affiliation(s)
- Yameng Song
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, People’s Republic of China
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People’s Republic of China
| | - Caiyun Yin
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People’s Republic of China
| | - Ning Kong
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
33
|
Tikhonov A, Kachanov A, Yudaeva A, Danilik O, Ponomareva N, Karandashov I, Kostyusheva A, Zamyatnin AA, Parodi A, Chulanov V, Brezgin S, Kostyushev D. Biomimetic Nanoparticles for Basic Drug Delivery. Pharmaceutics 2024; 16:1306. [PMID: 39458635 PMCID: PMC11510494 DOI: 10.3390/pharmaceutics16101306] [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: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Biomimetic nanoparticles (BMNPs) are innovative nanovehicles that replicate the properties of naturally occurring extracellular vesicles, facilitating highly efficient drug delivery across biological barriers to target organs and tissues while ensuring maximal biocompatibility and minimal-to-no toxicity. BMNPs can be utilized for the delivery of therapeutic payloads and for imparting novel properties to other nanotechnologies based on organic and inorganic materials. The application of specifically modified biological membranes for coating organic and inorganic nanoparticles has the potential to enhance their therapeutic efficacy and biocompatibility, presenting a promising pathway for the advancement of drug delivery technologies. This manuscript is grounded in the fundamentals of biomimetic technologies, offering a comprehensive overview and analytical perspective on the preparation and functionalization of BMNPs, which include cell membrane-coated nanoparticles (CMCNPs), artificial cell-derived vesicles (ACDVs), and fully synthetic vesicles (fSVs). This review examines both "top-down" and "bottom-up" approaches for nanoparticle preparation, with a particular focus on techniques such as cell membrane coating, cargo loading, and microfluidic fabrication. Additionally, it addresses the technological challenges and potential solutions associated with the large-scale production and clinical application of BMNPs and related technologies.
Collapse
Affiliation(s)
- Andrey Tikhonov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Oleg Danilik
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Ivan Karandashov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
34
|
Qiao L, Du X, Wang H, Wang Z, Gao S, Zhao CQ. Research Progress on the Strategies for Crossing the Blood-Brain Barrier. Mol Pharm 2024; 21:4786-4803. [PMID: 39231367 DOI: 10.1021/acs.molpharmaceut.4c00447] [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: 09/06/2024]
Abstract
Recently, the incidence of brain diseases, such as central nervous system degenerative diseases, brain tumors, and cerebrovascular diseases, has increased. However, the blood-brain barrier (BBB) limits the effective delivery of drugs to brain disease areas. Therefore, the mainstream direction of new drug development for these diseases is to engineer drugs that can better cross the BBB to exert their effects in the brain. This paper reviews the research progress and application of the main trans-BBB drug delivery strategies (receptor/transporter-mediated BBB crossing, focused ultrasound to open the BBB, adenosine agonist reversible opening of the BBB, aromatic resuscitation, transnasal administration, cell-mediated trans-BBB crossing, and viral vector system-mediated brain drug delivery). Meanwhile, the potential applications, advantages, and disadvantages of these strategies for crossing the BBB are analyzed. Finally, the future development prospects of strategies for crossing the BBB are also discussed. These strategies have potential value for treating brain diseases.
Collapse
Affiliation(s)
- Li Qiao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Xiuwei Du
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Hua Wang
- College of Intelligence and Information Engineering, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Zhiyi Wang
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Shijie Gao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Chun-Qin Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| |
Collapse
|
35
|
Zhen X, Li Y, Yuan W, Zhang T, Li M, Huang J, Kong N, Xie X, Wang S, Tao W. Biointerface-Engineered Hybrid Nanovesicles for Targeted Reprogramming of Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401495. [PMID: 38851884 DOI: 10.1002/adma.202401495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/21/2024] [Indexed: 06/10/2024]
Abstract
The tumor microenvironment (TME) of typical tumor types such as triple-negative breast cancer is featured by hypoxia and immunosuppression with abundant tumor-associated macrophages (TAMs), which also emerge as potential therapeutic targets for antitumor therapy. M1-like macrophage-derived exosomes (M1-Exos) have emerged as a promising tumor therapeutic candidate for their tumor-targeting and macrophage-polarization capabilities. However, the limited drug-loading efficiency and stability of M1-Exos have hindered their effectiveness in antitumor applications. Here, a hybrid nanovesicle is developed by integrating M1-Exos with AS1411 aptamer-conjugated liposomes (AApt-Lips), termed M1E/AALs. The obtained M1E/AALs are loaded with perfluorotributylamine (PFTBA) and IR780, as P-I, to construct P-I@M1E/AALs for reprogramming TME by alleviating tumor hypoxia and engineering TAMs. P-I@M1E/AAL-mediated tumor therapy enhances the in situ generation of reactive oxygen species, repolarizes TAMs toward an antitumor phenotype, and promotes the infiltration of T lymphocytes. The synergistic antitumor therapy based on P-I@M1E/AALs significantly suppresses tumor growth and prolongs the survival of 4T1-tumor-bearing mice. By integrating multiple treatment modalities, P-I@M1E/AAL nanoplatform demonstrates a promising therapeutic approach for overcoming hypoxic and immunosuppressive TME by targeted TAM reprogramming and enhanced tumor photodynamic immunotherapy. This study highlights an innovative TAM-engineering hybrid nanovesicle platform for the treatment of tumors characterized by hypoxic and immunosuppressive TME.
Collapse
Affiliation(s)
- Xueyan Zhen
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wanqing Yuan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Tingting Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Min Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Jinhai Huang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University; NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China; Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200030, China
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaoyu Xie
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
| | - Sicen Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an, 710061, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
36
|
Wang A, Walden M, Ettlinger R, Kiessling F, Gassensmith JJ, Lammers T, Wuttke S, Peña Q. Biomedical Metal-Organic Framework Materials: Perspectives and Challenges. ADVANCED FUNCTIONAL MATERIALS 2024; 34:adfm.202308589. [PMID: 39726715 PMCID: PMC7617264 DOI: 10.1002/adfm.202308589] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Indexed: 12/28/2024]
Abstract
Metal-organic framework (MOF) materials are gaining significant interest in biomedical research, owing to their high porosity, crystallinity, and structural and compositional diversity. Their versatile hybrid organic/inorganic chemistry endows MOFs with the capacity to retain organic (drug) molecules, metals, and gases, to effectively channel electrons and photons, to survive harsh physiological conditions such as low pH, and even to protect sensitive biomolecules. Extensive preclinical research has been carried out with MOFs to treat several pathologies and, recently, their integration with other biomedical materials such as stents and implants has demonstrated promising performance in regenerative medicine. However, there remains a significant gap between MOF preclinical research and translation into clinically and societally relevant medicinal products. Here, we outline the intrinsic features of MOFs and discuss how these are suited to specific biomedical applications like detoxification, drug and gas delivery, or as (combination) therapy platforms. We furthermore describe relevant examples of how MOFs have been engineered and evaluated in different medical indications, including cancer, microbial, and inflammatory diseases. Finally, we critically examine the challenges facing their translation into the clinic, with the goal of establishing promising research directions and more realistic approaches that can bridge the translational gap of MOFs and MOF-containing (nano)materials.
Collapse
Affiliation(s)
- Alec Wang
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Forckenbeckstrasse 55, 52074Aachen, Germany
| | - Madeline Walden
- BCMaterials (Basque Centre for Materials, Applications & Nanostructures), Bld. Martina Casiano, 3rd. Floor UPV/EHU Science Park Barrio Sarriena s/n, 48940Leioa, Spain
| | - Romy Ettlinger
- EastChem School of Chemistry, University of St Andrews, North Haugh, St AndrewsKY16 9ST, UK
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Forckenbeckstrasse 55, 52074Aachen, Germany
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry & Biomedical Engineering, University of Texas at Dallas, Richardson, TX75080-3021
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Forckenbeckstrasse 55, 52074Aachen, Germany
| | - Stefan Wuttke
- BCMaterials (Basque Centre for Materials, Applications & Nanostructures), Bld. Martina Casiano, 3rd. Floor UPV/EHU Science Park Barrio Sarriena s/n, 48940Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013Bilbao, Spain
| | - Quim Peña
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Forckenbeckstrasse 55, 52074Aachen, Germany
| |
Collapse
|
37
|
Mirgh D, Sonar S, Ghosh S, Adhikari MD, Subramaniyan V, Gorai S, Anand K. Landscape of exosomes to modified exosomes: a state of the art in cancer therapy. RSC Adv 2024; 14:30807-30829. [PMID: 39328877 PMCID: PMC11426072 DOI: 10.1039/d4ra04512b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Exosomes are a subpopulation of extracellular vesicles (EVs) that naturally originate from endosomes. They play a significant role in cellular communication. Tumor-secreted exosomes play a crucial role in cancer development and significantly contribute to tumorigenesis, angiogenesis, and metastasis by intracellular communication. Tumor-derived exosomes (TEXs) are a promising biomarker source of cancer detection in the early stages. On the other hand, they offer revolutionary cutting-edge approaches to cancer therapeutics. Exosomes offer a cell-free approach to cancer therapeutics, which overcomes immune cell and stem cell therapeutics-based limitations (complication, toxicity, and cost of treatment). There are multiple sources of therapeutic exosomes present (stem cells, immune cells, plant cells, and synthetic and modified exosomes). This article explores the dynamic source of exosomes (plants, mesenchymal stem cells, and immune cells) and their modification (chimeric, hybrid exosomes, exosome-based CRISPR, and drug delivery) based on cancer therapeutic development. This review also highlights exosomes based clinical trials and the challenges and future orientation of exosome research. We hope that this article will inspire researchers to further explore exosome-based cancer therapeutic platforms for precision oncology.
Collapse
Affiliation(s)
- Divya Mirgh
- Vaccine and Immunotherapy Centre, Massachusetts General Hospital Boston USA
| | - Swarup Sonar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu 602105 India
| | - Srestha Ghosh
- Department of Microbiology, Lady Brabourne College Kolkata West Bengal 700017 India
| | - Manab Deb Adhikari
- Department of Biotechnology, University of North Bengal Darjeeling West Bengal India
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University Bandar Sunway Subang Jaya Selangor 47500 Malaysia
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center Chicago IL USA
| | - Krishnan Anand
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Faculty of Health Sciences, University of the Free State Bloemfontein 9300 South Africa
| |
Collapse
|
38
|
Neupane KR, Aryal SP, Harvey BT, Ramon GS, Chun B, McCorkle JR, Kolesar JM, Kekenes-Huskey PM, Richards CI. Organelle Specific Macrophage Engineered Vesicles Differentially Reprogram Macrophage Polarization. Adv Healthc Mater 2024:e2401906. [PMID: 39240019 DOI: 10.1002/adhm.202401906] [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: 05/22/2024] [Revised: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Tumor-associated macrophages (TAMs) represent the majority of the immune cells present in the tumor microenvironment. These macrophages exhibit an anti-inflammatory (M2)-like physiological state and execute immune-suppressive and tumor-supporting properties. With TAMs being plastic, there is a growing interest in reprogramming them toward a pro-inflammatory (M1)-like phenotype that exhibits anti-tumoral properties. Recent studies have demonstrated that both engineered vesicles derived from macrophages and endogenous extracellular vesicles produced by macrophages can be programmed to alter macrophage phenotype. Here it is demonstrated that pro-inflammatory macrophage-engineered subcellular vesicles (MEVs) have differential properties based on their organelle of origin. Endoplasmic reticulum specific MEVs (erMEVs) treated M2 macrophages exhibit enhanced pro-inflammatory cytokine production compared to plasma membrane specific MEVs (pmMEVs) treated M2 macrophages. In addition, under in vitro co-culture conditions, erMEVs elicit superior efficacy in suppressing the viability of cancer cells compared to the same concentration of pmMEVs. Furthermore, erMEVs and pmMEVs maintain differences in their membrane proteins, that regulate the repolarization efficacy of M2 macrophages toward an M1-like phenotype. In addition, The M2 to M1 repolarizing efficacy of MEVs can be altered by changing the activity of the membrane proteins present on erMEVs or pmMEVs.
Collapse
Affiliation(s)
- Khaga R Neupane
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Surya P Aryal
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Brock T Harvey
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Geraldine San Ramon
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, IL, 60153, USA
| | - Byeong Chun
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, IL, 60153, USA
| | - J Robert McCorkle
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, 40508, USA
| | - Jill M Kolesar
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, 40508, USA
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, IL, 60153, USA
| | | |
Collapse
|
39
|
González Á, López-Borrego S, Sandúa A, Vales-Gomez M, Alegre E. Extracellular vesicles in cancer: challenges and opportunities for clinical laboratories. Crit Rev Clin Lab Sci 2024; 61:435-457. [PMID: 38361287 DOI: 10.1080/10408363.2024.2309935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/03/2024] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
Extracellular vesicles (EVs) are nano-sized particles secreted by most cells. They transport different types of biomolecules (nucleic acids, proteins, and lipids) characteristic of their tissue or cellular origin that can mediate long-distance intercellular communication. In the case of cancer, EVs participate in tumor progression by modifying the tumor microenvironment, favoring immune tolerance and metastasis development. Consequently, EVs have great potential in liquid biopsy for cancer diagnosis, prognosis and follow-up. In addition, EVs could have a role in cancer treatment as a targeted drug delivery system. The intense research in the EV field has resulted in hundreds of patents and the creation of biomedical companies. However, methodological issues and heterogeneity in EV composition have hampered the advancement of EV validation trials and the development of EV-based diagnostic and therapeutic products. Consequently, only a few EV biomarkers have moved from research to clinical laboratories, such as the ExoDx Prostate IntelliScore (EPI) test, a CLIA/FDA-approved EV prostate cancer diagnostic test. In addition, the number of large-scale multicenter studies that would clearly define biomarker performance is limited. In this review, we will critically describe the different types of EVs, the methods for their enrichment and characterization, and their biological role in cancer. Then, we will specially focus on the parameters to be considered for the translation of EV biology to the clinic laboratory, the advances already made in the field of EVs related to cancer diagnosis and treatment, and the issues still pending to be solved before EVs could be used as a routine tool in oncology.
Collapse
Affiliation(s)
- Álvaro González
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Silvia López-Borrego
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council, Madrid, Spain
| | - Amaia Sandúa
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mar Vales-Gomez
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council, Madrid, Spain
| | - Estibaliz Alegre
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
40
|
Chen C, Chang ZH, Yao B, Liu XY, Zhang XW, Liang J, Wang JJ, Bao SQ, Chen MM, Zhu P, Li XH. 3D printing of interferon γ-preconditioned NSC-derived exosomes/collagen/chitosan biological scaffolds for neurological recovery after TBI. Bioact Mater 2024; 39:375-391. [PMID: 38846528 PMCID: PMC11153920 DOI: 10.1016/j.bioactmat.2024.05.026] [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: 03/14/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
The reconstruction of neural function and recovery of chronic damage following traumatic brain injury (TBI) remain significant clinical challenges. Exosomes derived from neural stem cells (NSCs) offer various benefits in TBI treatment. Numerous studies confirmed that appropriate preconditioning methods enhanced the targeted efficacy of exosome therapy. Interferon-gamma (IFN-γ) possesses immunomodulatory capabilities and is widely involved in neurological disorders. In this study, IFN-γ was employed for preconditioning NSCs to enhance the efficacy of exosome (IFN-Exo, IE) for TBI. miRNA sequencing revealed the potential of IFN-Exo in promoting neural differentiation and modulating inflammatory responses. Through low-temperature 3D printing, IFN-Exo was combined with collagen/chitosan (3D-CC-IE) to preserve the biological activity of the exosome. The delivery of exosomes via biomaterial scaffolds benefited the retention and therapeutic potential of exosomes, ensuring that they could exert long-term effects at the injury site. The 3D-CC-IE scaffold exhibited excellent biocompatibility and mechanical properties. Subsequently, 3D-CC-IE scaffold significantly improved impaired motor and cognitive functions after TBI in rat. Histological results showed that 3D-CC-IE scaffold markedly facilitated the reconstruction of damaged neural tissue and promoted endogenous neurogenesis. Further mechanistic validation suggested that IFN-Exo alleviated neuroinflammation by modulating the MAPK/mTOR signaling pathway. In summary, the results of this study indicated that 3D-CC-IE scaffold engaged in long-term pathophysiological processes, fostering neural function recovery after TBI, offering a promising regenerative therapy avenue.
Collapse
Affiliation(s)
- Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Bin Yao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Xiao-Yin Liu
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
41
|
Zhao J, Ma Y, Zheng X, Sun Z, Lin H, Du C, Cao J. Bladder cancer: non-coding RNAs and exosomal non-coding RNAs. Funct Integr Genomics 2024; 24:147. [PMID: 39217254 DOI: 10.1007/s10142-024-01433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Bladder cancer (BCa) is a highly prevalent type of cancer worldwide, and it is responsible for numerous deaths and cases of disease. Due to the diverse nature of this disease, it is necessary to conduct significant research that delves deeper into the molecular aspects, to potentially discover novel diagnostic and therapeutic approaches. Lately, there has been a significant increase in the focus on non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), due to their growing recognition for their involvement in the progression and manifestation of BCa. The interest in exosomes has greatly grown due to their potential for transporting a diverse array of active substances, including proteins, nucleic acids, carbohydrates, and lipids. The combination of these components differs based on the specific cell and its condition. Research indicates that using exosomes could have considerable advantages in identifying and forecasting BCa, offering a less invasive alternative. The distinctive arrangement of the lipid bilayer membrane found in exosomes is what makes them particularly effective for administering treatments aimed at managing cancer. In this review, we have tried to summarize different ncRNAs that are involved in BCa pathogenesis. Moreover, we highlighted the role of exosomal ncRNAs in BCa.
Collapse
Affiliation(s)
- Jingang Zhao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Yangyang Ma
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Xiaodong Zheng
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Zhen Sun
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Hongxiang Lin
- Department of Urology, Ganzhou Donghe Hospital, Ganzhou, 341000, Jiang'xi, China
| | - Chuanjun Du
- Department of Urology, Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310009, Zhe'jiang, China
| | - Jing Cao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China.
| |
Collapse
|
42
|
Zhang Z, Fu Y, Ju X, Zhang F, Zhang P, He M. Advances in Engineering Circular RNA Vaccines. Pathogens 2024; 13:692. [PMID: 39204292 PMCID: PMC11356823 DOI: 10.3390/pathogens13080692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Engineered circular RNAs (circRNAs) are a class of single-stranded RNAs with head-to-tail covalently linked structures that integrate open reading frames (ORFs) and internal ribosome entry sites (IRESs) with the function of coding and expressing proteins. Compared to mRNA vaccines, circRNA vaccines offer a more improved method that is safe, stable, and simple to manufacture. With the rapid revelation of the biological functions of circRNA and the success of Severe Acute Respiratory Coronavirus Type II (SARS-CoV-2) mRNA vaccines, biopharmaceutical companies and researchers around the globe are attempting to develop more stable circRNA vaccines for illness prevention and treatment. Nevertheless, research on circRNA vaccines is still in its infancy, and more work and assessment are needed for their synthesis, delivery, and use. In this review, based on the current understanding of the molecular biological properties and immunotherapeutic mechanisms of circRNA, we summarize the current preparation methods of circRNA vaccines, including design, synthesis, purification, and identification. We discuss their delivery strategies and summarize the challenges facing the clinical application of circRNAs to provide references for circRNA vaccine-related research.
Collapse
Affiliation(s)
- Zhongyan Zhang
- School of Pharmacy, Yantai University, Yantai 264005, China;
| | - Yuanlei Fu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264005, China; (Y.F.); (X.J.); (F.Z.)
| | - Xiaoli Ju
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264005, China; (Y.F.); (X.J.); (F.Z.)
| | - Furong Zhang
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264005, China; (Y.F.); (X.J.); (F.Z.)
| | - Peng Zhang
- School of Pharmacy, Yantai University, Yantai 264005, China;
| | - Meilin He
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264005, China; (Y.F.); (X.J.); (F.Z.)
| |
Collapse
|
43
|
Song X, Song Y, Zhang J, Hu Y, Zhang L, Huang Z, Abbas Raza SH, Jiang C, Ma Y, Ma Y, Wu H, Wei D. Regulatory role of exosome-derived miRNAs and other contents in adipogenesis. Exp Cell Res 2024; 441:114168. [PMID: 39004201 DOI: 10.1016/j.yexcr.2024.114168] [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: 05/18/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Intramuscular fat (IMF) content significantly impacts meat quality. influenced by complex interactions between skeletal muscle cells and adipocytes. Adipogenesis plays a pivotal role in IMF formation. Exosomes, extracellular membranous nanovesicles, facilitate intercellular communication by transporting proteins, nucleic acids (DNA and RNA), and other biomolecules into target cells, thereby modulating cellular behaviors. Recent studies have linked exosome-derived microRNAs (miRNAs) and other cargo to adipogenic processes. Various cell types, including skeletal muscle cells, interact with adipocytes via exosome secretion and uptake. Exosomes entering adipocytes regulate adipogenesis by modulating key signaling pathways, thereby influencing the extent and distribution of IMF deposition. This review comprehensively explores the origin, formation, and mechanisms of exosome action, along with current research and their applications in adipogenesis. Emphasis is placed on exosome-mediated regulation of miRNAs, non-coding RNAs (ncRNAs), proteins, lipids, and other biomolecules during adipogenesis. Leveraging exosomal contents for genetic breeding and treating obesity-related disorders is discussed. Insights gathered contribute to advancing understanding and potential therapeutic applications of exosome-regulated adipogenesis mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Song
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yaping Song
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Jiupan Zhang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, 750021, China
| | - Yamei Hu
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Lingkai Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | | | - Sayed Haidar Abbas Raza
- Xichang University, Xichang, 615000, China; Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
| | - Chao Jiang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yanfen Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Yun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Hao Wu
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Dawei Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China; Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, 750021, China.
| |
Collapse
|
44
|
Tamimi A, Javid M, Sedighi-Pirsaraei N, Mirdamadi A. Exosome prospects in the diagnosis and treatment of non-alcoholic fatty liver disease. Front Med (Lausanne) 2024; 11:1420281. [PMID: 39144666 PMCID: PMC11322140 DOI: 10.3389/fmed.2024.1420281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
The growing prevalence of NAFLD and its global health burden have provoked considerable research on possible diagnostic and therapeutic options for NAFLD. Although various pathophysiological mechanisms and genetic factors have been identified to be associated with NAFLD, its treatment remains challenging. In recent years, exosomes have attracted widespread attention for their role in metabolic dysfunctions and their efficacy as pathological biomarkers. Exosomes have also shown tremendous potential in treating a variety of disorders. With increasing evidence supporting the significant role of exosomes in NAFLD pathogenesis, their theragnostic potential has become a point of interest in NAFLD. Expectedly, exosome-based treatment strategies have shown promise in the prevention and amelioration of NAFLD in preclinical studies. However, there are still serious challenges in preparing, standardizing, and applying exosome-based therapies as a routine clinical option that should be overcome. Due to the great potential of this novel theragnostic agent in NAFLD, further investigations on their safety, clinical efficacy, and application standardization are highly recommended.
Collapse
|
45
|
Zhou X, Liu J, Wu F, Mao J, Wang Y, Zhu J, Hong K, Xie H, Li B, Qiu X, Xiao X, Wen C. The application potential of iMSCs and iMSC-EVs in diseases. Front Bioeng Biotechnol 2024; 12:1434465. [PMID: 39135947 PMCID: PMC11317264 DOI: 10.3389/fbioe.2024.1434465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
The immune system, functioning as the body's "defense army", plays a role in surveillance, defense. Any disruptions in immune system can lead to the development of immune-related diseases. Extensive researches have demonstrated the crucial immunoregulatory role of mesenchymal stem cells (MSCs) in these diseases. Of particular interest is the ability to induce somatic cells under specific conditions, generating a new cell type with stem cell characteristics known as induced pluripotent stem cell (iPSC). The differentiation of iPSCs into MSCs, specifically induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs), hold promise as a potential solution to the challenges of MSCs, potentially serving as an alternative to traditional drug therapies. Moreover, the products of iMSCs, termed induced pluripotent stem cell-derived mesenchymal stem cell-derived extracellular vesicles (iMSC-EVs), may exhibit functions similar to iMSCs. With the biological advantages of EVs, they have become the focus of "cell-free therapy". Here, we provided a comprehensive summary of the biological impact of iMSCs on immune cells, explored the applications of iMSCs and iMSC-EVs in diseases, and briefly discussed the fundamental characteristics of EVs. Finally, we overviewed the current advantages and challenges associated with iMSCs and iMSC-EVs. It is our hope that this review related to iMSCs and iMSC-EVs will contribute to the development of new approaches for the treatment of diseases.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinyu Liu
- Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiangbin Xiao
- Department of Cardiovascular, People’s Hospital of Jianyang, Jianyang, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
46
|
Bahadorani M, Nasiri M, Dellinger K, Aravamudhan S, Zadegan R. Engineering Exosomes for Therapeutic Applications: Decoding Biogenesis, Content Modification, and Cargo Loading Strategies. Int J Nanomedicine 2024; 19:7137-7164. [PMID: 39050874 PMCID: PMC11268655 DOI: 10.2147/ijn.s464249] [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: 03/04/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Exosomes emerge from endosomal invagination and range in size from 30 to 200 nm. Exosomes contain diverse proteins, lipids, and nucleic acids, which can indicate the state of various physiological and pathological processes. Studies have revealed the remarkable clinical potential of exosomes in diagnosing and prognosing multiple diseases, including cancer, cardiovascular disorders, and neurodegenerative conditions. Exosomes also have the potential to be engineered and deliver their cargo to a specific target. However, further advancements are imperative to optimize exosomes' diagnostic and therapeutic capabilities for practical implementation in clinical settings. This review highlights exosomes' diagnostic and therapeutic applications, emphasizing their engineering through simple incubation, biological, and click chemistry techniques. Additionally, the loading of therapeutic agents onto exosomes, utilizing passive and active strategies, and exploring hybrid and artificial exosomes are discussed.
Collapse
Affiliation(s)
- Mehrnoosh Bahadorani
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Mahboobeh Nasiri
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Shyam Aravamudhan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| | - Reza Zadegan
- Department of Nanoengineering, Joint School of Nanoscience & Nanoengineering, North Carolina Agriculture and Technical State University, Greensboro, NC, USA
| |
Collapse
|
47
|
Xu H, Ma H, Zha L, Li Q, Pan H, Zhang L. Engineered exosomes transporting the lncRNA, SVIL-AS1, inhibit the progression of lung cancer via targeting miR-21-5p. Am J Cancer Res 2024; 14:3335-3347. [PMID: 39113865 PMCID: PMC11301303 DOI: 10.62347/yrjk5888] [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: 04/18/2024] [Accepted: 06/20/2024] [Indexed: 08/10/2024] Open
Abstract
In this study, we constructed engineered exosomes carrying the long non-coding RNA (lncRNA) SVIL-AS1 (SVIL-AS1 Exos), and explored its role and mechanism in lung cancer. After the construction of SVIL-AS1 Exos, their physicochemical characteristics were identified. Then, their function and effect in three different cell lines, A549, HeLa, and HepG2, were detected using western blot, the quantitative reverse transcriptase polymerase chain reaction, flow cytometry, 5-ethynyl-2'-deoxyuridine, and Cell Counting Kit-8 experiments. Finally, a mouse xenograft model was constructed to analyze tumor growth and explore the in vivo utility of SVIL-AS1 Exos using hematoxylin and eosin staining, immunohistochemistry, and the TdT-mediated dUTP nick end labeling assay. The results demonstrated that SVIL-AS1 Exos preferentially targeted A549 lung cancer cells over HeLa and HepG2 cells. SVIL-AS1 Exos promoted apoptosis and inhibited A549 cell proliferation by elevating expression of the lncRNA, SVIL-AS1. In vivo, SVIL-AS1 Exos effectively inhibited the growth of lung cancer A549 cells. Furthermore, SVIL-AS1 Exos suppressed the expression of miR-21-5p and upregulated the expression of caspase-9, indicating that SVIL-AS1 may regulate the development of lung cancer through the miR-21-5p/caspase-9 pathway. In conclusion, the engineered SVIL-AS1 Exos targeted lung cancer cells to inhibit the expression of miR-21-5p, upregulate the expression of caspase-9, and inhibit the development of lung cancer.
Collapse
Affiliation(s)
- Hao Xu
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Hongda Ma
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Lifen Zha
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Qian Li
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Huiming Pan
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| | - Ladi Zhang
- Department of Respiratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University Danyang 212300, Jiangsu, China
| |
Collapse
|
48
|
Yang Y, Chen H, Li Y, Liang J, Huang F, Wang L, Miao H, Nanda HS, Wu J, Peng X, Zhou Y. Hydrogel Loaded with Extracellular Vesicles: An Emerging Strategy for Wound Healing. Pharmaceuticals (Basel) 2024; 17:923. [PMID: 39065772 PMCID: PMC11280375 DOI: 10.3390/ph17070923] [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: 05/30/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
An increasing number of novel biomaterials have been applied in wound healing therapy. Creating beneficial environments and containing various bioactive molecules, hydrogel- and extracellular vesicle (EV)-based therapies have respectively emerged as effective approaches for wound healing. Moreover, the synergistic combination of these two components demonstrates more favorable outcomes in both chronic and acute wound healing. This review provides a comprehensive discussion and summary of the combined application of EVs and hydrogels to address the intricate scenario of wounds. The wound healing process and related biological mechanisms are outlined in the first section. Subsequently, the utilization of EV-loaded hydrogels during the wound healing process is evaluated and discussed. The moist environment created by hydrogels is conducive to wound tissue regeneration. Additionally, the continuous and controlled release of EVs from various origins could be achieved by hydrogel encapsulation. Finally, recent in vitro and in vivo studies reported on hydrogel dressings loaded with EVs are summarized and challenges and opportunities for the future clinical application of this therapeutic approach are outlined.
Collapse
Affiliation(s)
- Yucan Yang
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Huizhi Chen
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Yunjie Li
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Junting Liang
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Feng Huang
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Liyan Wang
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Huilai Miao
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
| | - Himansu Sekhar Nanda
- Biomaterials and Biomanufacturing Laboratory, Discipline of Mechanical Engineering, PDPM Indian Institute of Information Technology Design and Manufacturing, Jabalpur 482005, Madhya Pradesh, India;
| | - Jin Wu
- State Key Laboratory of Optoelectronic Materials and Technologies and the Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510275, China;
| | - Xinsheng Peng
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| | - Yubin Zhou
- Key Laboratory of Liver Injury Diagnosis and Repair, and Department of Hepatobiliary Surgery, The 2nd Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Y.); (H.C.); (Y.L.); (J.L.); (F.H.); (L.W.); (H.M.)
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Dongguan Key Laboratory of Advanced Drug Delivery and Biosensing Research and Development, School of Pharmacy, and Dongguan Innovation Institute, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
49
|
Sun DS, Chang HH. Extracellular vesicles: Function, resilience, biomarker, bioengineering, and clinical implications. Tzu Chi Med J 2024; 36:251-259. [PMID: 38993825 PMCID: PMC11236075 DOI: 10.4103/tcmj.tcmj_28_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as key players in intercellular communication, disease pathology, and therapeutic innovation. Initially overlooked as cellular debris, EVs are now recognized as vital mediators of cell-to-cell communication, ferrying a cargo of proteins, nucleic acids, and lipids, providing cellular resilience in response to stresses. This review provides a comprehensive overview of EVs, focusing on their role as biomarkers in disease diagnosis, their functional significance in physiological and pathological processes, and the potential of bioengineering for therapeutic applications. EVs offer a promising avenue for noninvasive disease diagnosis and monitoring, reflecting the physiological state of originating cells. Their diagnostic potential spans a spectrum of diseases, including cancer, cardiovascular disorders, neurodegenerative diseases, and infectious diseases. Moreover, their presence in bodily fluids such as blood, urine, and cerebrospinal fluid enhances their diagnostic utility, presenting advantages over traditional methods. Beyond diagnostics, EVs mediate crucial roles in intercellular communication, facilitating the transfer of bioactive molecules between cells. This communication modulates various physiological processes such as tissue regeneration, immune modulation, and neuronal communication. Dysregulation of EV-mediated communication is implicated in diseases such as cancer, immune disorders, and neurodegenerative diseases, highlighting their therapeutic potential. Bioengineering techniques offer avenues for manipulating EVs for therapeutic applications, from isolation and purification to engineering cargo and targeted delivery systems. These approaches hold promise for developing novel therapeutics tailored to specific diseases, revolutionizing personalized medicine. However, challenges such as standardization, scalability, and regulatory approval need addressing for successful clinical translation. Overall, EVs represent a dynamic frontier in biomedical research with vast potential for diagnostics, therapeutics, and personalized medicine.
Collapse
Affiliation(s)
- Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
50
|
Li W, Wang C, Zhang Y, Lu Y. Lipid Nanocarrier-Based mRNA Therapy: Challenges and Promise for Clinical Transformation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310531. [PMID: 38287729 DOI: 10.1002/smll.202310531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Indexed: 01/31/2024]
Abstract
Due to the outbreak of novel coronavirus pneumonia, messenger RNA (mRNA) technology has attracted heated attention. A specific, safe, and efficient mRNA delivery system is needed. Lipid nanocarriers have become attractive carriers for mRNA delivery due to their high delivery efficiency, few side effects, and easy modification to change their structures and functions. To achieve the desired biological effect, lipid nanocarriers must reach the designated location for effective drug delivery. Therefore, the effects of the composition of lipid nanocarriers on their key properties are briefly reviewed. In addition, the progress of smart drug delivery by changing the composition of lipid nanocarriers is summarized, and the importance of component design and structure is emphasized. Subsequently, this review summarizes the latest progress in lipid nanocarrier-based mRNA technology and provides corresponding strategies for its current challenges, putting forward valuable information for the future design of lipid nanocarriers and mRNA.
Collapse
Affiliation(s)
- Wenchao Li
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Yifei Zhang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|